1
|
Endo Y, Aoki T, Jafari D, Rolston DM, Hagiwara J, Ito-Hagiwara K, Nakamura E, Kuschner CE, Becker LB, Hayashida K. Acute lung injury and post-cardiac arrest syndrome: a narrative review. J Intensive Care 2024; 12:32. [PMID: 39227997 PMCID: PMC11370287 DOI: 10.1186/s40560-024-00745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Post-cardiac arrest syndrome (PCAS) presents a multifaceted challenge in clinical practice, characterized by severe neurological injury and high mortality rates despite advancements in management strategies. One of the important critical aspects of PCAS is post-arrest lung injury (PALI), which significantly contributes to poor outcomes. PALI arises from a complex interplay of pathophysiological mechanisms, including trauma from chest compressions, pulmonary ischemia-reperfusion (IR) injury, aspiration, and systemic inflammation. Despite its clinical significance, the pathophysiology of PALI remains incompletely understood, necessitating further investigation to optimize therapeutic approaches. METHODS This review comprehensively examines the existing literature to elucidate the epidemiology, pathophysiology, and therapeutic strategies for PALI. A comprehensive literature search was conducted to identify preclinical and clinical studies investigating PALI. Data from these studies were synthesized to provide a comprehensive overview of PALI and its management. RESULTS Epidemiological studies have highlighted the substantial prevalence of PALI in post-cardiac arrest patients, with up to 50% of survivors experiencing acute lung injury. Diagnostic imaging modalities, including chest X-rays, computed tomography, and lung ultrasound, play a crucial role in identifying PALI and assessing its severity. Pathophysiologically, PALI encompasses a spectrum of factors, including chest compression-related trauma, pulmonary IR injury, aspiration, and systemic inflammation, which collectively contribute to lung dysfunction and poor outcomes. Therapeutically, lung-protective ventilation strategies, such as low tidal volume ventilation and optimization of positive end-expiratory pressure, have emerged as cornerstone approaches in the management of PALI. Additionally, therapeutic hypothermia and emerging therapies targeting mitochondrial dysfunction hold promise in mitigating PALI-related morbidity and mortality. CONCLUSION PALI represents a significant clinical challenge in post-cardiac arrest care, necessitating prompt diagnosis and targeted interventions to improve outcomes. Mitochondrial-related therapies are among the novel therapeutic strategies for PALI. Further clinical research is warranted to optimize PALI management and enhance post-cardiac arrest care paradigms.
Collapse
Affiliation(s)
- Yusuke Endo
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Tomoaki Aoki
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Daniel Jafari
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Daniel M Rolston
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Jun Hagiwara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Kanako Ito-Hagiwara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Eriko Nakamura
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Cyrus E Kuschner
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Lance B Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Kei Hayashida
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA.
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
2
|
Leroy V, Manual Kollareth DJ, Tu Z, Valisno JAC, Woolet-Stockton M, Saha B, Emtiazjoo AM, Rackauskas M, Moldawer LL, Efron PA, Cai G, Atkinson C, Upchurch GR, Sharma AK. MerTK-dependent efferocytosis by monocytic-MDSCs mediates resolution of ischemia/reperfusion injury after lung transplant. JCI Insight 2024; 9:e179876. [PMID: 39172530 PMCID: PMC11466183 DOI: 10.1172/jci.insight.179876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024] Open
Abstract
Lung transplantation (LTx) outcomes are impeded by ischemia/reperfusion injury (IRI) and subsequent chronic lung allograft dysfunction (CLAD). We examined the undefined role of receptor Mer tyrosine kinase (MerTK) on monocytic myeloid-derived suppressor cells (M-MDSCs) in efferocytosis to facilitate resolution of lung IRI. Single-cell RNA sequencing of lung tissue and bronchoalveolar lavage (BAL) from patients after LTx were analyzed. Murine lung hilar ligation and allogeneic orthotopic LTx models of IRI were used with BALB/c (WT), Cebpb-/- (MDSC-deficient), Mertk-/-, or MerTK-cleavage-resistant mice. A significant downregulation in MerTK-related efferocytosis genes in M-MDSC populations of patients with CLAD was observed compared with healthy individuals. In the murine IRI model, a significant increase in M-MDSCs, MerTK expression, and efferocytosis and attenuation of lung dysfunction was observed in WT mice during injury resolution that was absent in Cebpb-/- and Mertk-/- mice. Adoptive transfer of M-MDSCs in Cebpb-/- mice significantly attenuated lung dysfunction and inflammation. Additionally, in a murine orthotopic LTx model, increases in M-MDSCs were associated with resolution of lung IRI in the transplant recipients. In vitro studies demonstrated the ability of M-MDSCs to efferocytose apoptotic neutrophils in a MerTK-dependent manner. Our results suggest that MerTK-dependent efferocytosis by M-MDSCs can substantially contribute to the resolution of post-LTx IRI.
Collapse
Affiliation(s)
- Victoria Leroy
- Department of Surgery
- Department of Pharmacology and Therapeutics
| | | | - Zhenxiao Tu
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | | | - Biplab Saha
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Amir M. Emtiazjoo
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | | | | | - Guoshuai Cai
- Department of Surgery
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Carl Atkinson
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | - Ashish K. Sharma
- Department of Surgery
- Department of Pharmacology and Therapeutics
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
3
|
Kollareth DJM, Leroy V, Tu Z, Woolet-Stockton MJ, Kamat M, Garrett TJ, Atkinson C, Cai G, Upchurch GR, Sharma AK. Lipoxin A 4 /FPR2 signaling mitigates ferroptosis of alveolar epithelial cells via NRF2-dependent pathway during lung ischemia-reperfusion injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590127. [PMID: 38712069 PMCID: PMC11071405 DOI: 10.1101/2024.04.22.590127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
BACKGROUND Post-lung transplantation (LTx) injury can involve sterile inflammation due to ischemia-reperfusion injury (IRI). We investigated the cell-specific role of ferroptosis (excessive iron-mediated cell death) in mediating lung IRI and determined if specialized pro-resolving mediators such as Lipoxin A4 (LxA 4 ) can protect against ferroptosis in lung IRI. METHODS Single-cell RNA sequencing of lung tissue from post-LTx patients was analyzed. Lung IRI was evaluated in C57BL/6 (WT), formyl peptide receptor 2 knockout ( Fpr2 -/- ) and nuclear factor erythroid 2-related factor 2 knockout ( Nrf2 -/- ) mice using a hilar-ligation model with or without LxA 4 administration. Furthermore, the protective efficacy of LxA 4 was evaluated employing a murine orthotopic LTx model and in vitro studies using alveolar type II epithelial (ATII) cells. RESULTS Differential expression of ferroptosis-related genes was observed in post-LTx patient samples compared to healthy controls. A significant increase in the levels of oxidized lipids and reduction in the levels of intact lipids were observed in mice subjected to IRI compared to shams. Furthermore, pharmacological inhibition of ferroptosis with liproxstatin-1 mitigated lung IRI and lung dysfunction. Importantly, LxA 4 treatment attenuated pulmonary dysfunction, ferroptosis and inflammation in WT mice subjected to lung IRI, but not in Fpr2 -/- or Nrf2 -/- mice, after IRI. In the murine LTx model, LxA 4 treatment increased PaO 2 levels and attenuated lung IRI. Mechanistically, LxA 4 -mediated protection involves increase in NRF2 activation and glutathione concentration as well as decrease in MDA levels in ATII cells. CONCLUSIONS LxA 4 /FPR2 signaling on ATII cells mitigates ferroptosis via NRF2 activation and protects against lung IRI.
Collapse
|
4
|
Li Q, Nie H. Advances in lung ischemia/reperfusion injury: unraveling the role of innate immunity. Inflamm Res 2024; 73:393-405. [PMID: 38265687 DOI: 10.1007/s00011-023-01844-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/03/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Lung ischemia/reperfusion injury (LIRI) is a common occurrence in clinical practice and represents a significant complication following pulmonary transplantation and various diseases. At the core of pulmonary ischemia/reperfusion injury lies sterile inflammation, where the innate immune response plays a pivotal role. This review aims to investigate recent advancements in comprehending the role of innate immunity in LIRI. METHODS A computer-based online search was performed using the PubMed database and Web of Science database for published articles concerning lung ischemia/reperfusion injury, cell death, damage-associated molecular pattern molecules (DAMPs), innate immune cells, innate immunity, inflammation. RESULTS During the process of lung ischemia/reperfusion, cellular injury even death can occur. When cells are injured or undergo cell death, endogenous ligands known as DAMPs are released. These molecules can be recognized and bound by pattern recognition receptors (PRRs), leading to the recruitment and activation of innate immune cells. Subsequently, a cascade of inflammatory responses is triggered, ultimately exacerbating pulmonary injury. These steps are complex and interrelated rather than being in a linear relationship. In recent years, significant progress has been made in understanding the immunological mechanisms of LIRI, involving novel types of cell death, the ability of receptors other than PRRs to recognize DAMPs, and a more detailed mechanism of action of innate immune cells in ischemia/reperfusion injury (IRI), laying the groundwork for the development of novel diagnostic and therapeutic approaches. CONCLUSIONS Various immune components of the innate immune system play critical roles in lung injury after ischemia/reperfusion. Preventing cell death and the release of DAMPs, interrupting DAMPs receptor interactions, disrupting intracellular inflammatory signaling pathways, and minimizing immune cell recruitment are essential for lung protection in LIRI.
Collapse
Affiliation(s)
- Qingqing Li
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Hanxiang Nie
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China.
| |
Collapse
|
5
|
Leroy V, Manual Kollareth DJ, Tu Z, Valisno JAC, Woolet-Stockton M, Saha B, Emtiazjoo AM, Rackauskas M, Moldawer LL, Efron PA, Cai G, Atkinson C, Upchurch GR, Sharma AK. MerTK-dependent efferocytosis by monocytic-MDSCs mediates resolution of post-lung transplant injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576261. [PMID: 38328174 PMCID: PMC10849528 DOI: 10.1101/2024.01.18.576261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Rationale Patients with end stage lung diseases require lung transplantation (LTx) that can be impeded by ischemia-reperfusion injury (IRI) leading to subsequent chronic lung allograft dysfunction (CLAD) and inadequate outcomes. Objectives We examined the undefined role of MerTK (receptor Mer tyrosine kinase) on monocytic myeloid-derived suppressor cells (M-MDSCs) in efferocytosis (phagocytosis of apoptotic cells) to facilitate resolution of lung IRI. Methods Single-cell RNA sequencing of lung tissue and BAL from post-LTx patients was analyzed. Murine lung hilar ligation and allogeneic orthotopic LTx models of IRI were used with Balb/c (WT), cebpb -/- (MDSC-deficient), Mertk -/- or MerTK-CR (cleavage resistant) mice. Lung function, IRI (inflammatory cytokine and myeloperoxidase expression, immunohistology for neutrophil infiltration), and flow cytometry of lung tissue for efferocytosis of apoptotic neutrophils were assessed in mice. Measurements and Main Results A significant downregulation in MerTK-related efferocytosis genes in M-MDSC populations of CLAD patients compared to healthy subjects was observed. In the murine IRI model, significant increase in M-MDSCs, MerTK expression and efferocytosis was observed in WT mice during resolution phase that was absent in cebpb -/- Land Mertk -/- mice. Adoptive transfer of M-MDSCs in cebpb -/- mice significantly attenuated lung dysfunction, and inflammation leading to resolution of IRI. Additionally, in a preclinical murine orthotopic LTx model, increases in M-MDSCs were associated with resolution of lung IRI in the transplant recipients. In vitro studies demonstrated the ability of M-MDSCs to efferocytose apoptotic neutrophils in a MerTK-dependent manner. Conclusions Our results suggest that MerTK-dependent efferocytosis by M-MDSCs can significantly contribute to the resolution of post-LTx IRI.
Collapse
|
6
|
He X, Li Z, Ye M, Zhao C, Wu S, Qin Y, Guo Y, Zhang L, Lin F. Near-infrared laser-irradiated upconversion nanoparticles with dexamethasone precise released for alleviating lung ischemia-reperfusion injury. Front Bioeng Biotechnol 2023; 11:1176369. [PMID: 37214302 PMCID: PMC10196198 DOI: 10.3389/fbioe.2023.1176369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction: Dexamethasone (DEX), as an important enduring-effect glucocorticoid (GC), holds great promise in the field of lung ischemia-reperfusion injury (LIRI) comprehensive therapy owing to its immunomodulatory properties, such as inducing apoptosis and cell cycle distribution. However, its potent anti-inflammatory application is still restricted because of multiple internal physiologic barriers. Methods: Herein, we developed upconversion nanoparticles (UCNPs) coated with photosensitizer/capping agent/fluorescent probe-modified mesoporous silica (UCNPs@mSiO2[DEX]-Py/β-CD/FITC, USDPFs) for precise DEX release synergistic LIRI comprehensive therapy. The UCNPs were designed by covering an inert YOF:Yb shell on the YOF:Yb, Tm core to achieve high-intensity blue and red upconversion emission upon Near-Infrared (NIR) laser irradiation. Results: Under suitable compatibility conditions, the molecular structure of photosensitizer can be damaged along with capping agent shedding, which endowed USDPFs with an outstanding capability to carry out DEX release controlling and fluorescent indicator targeting. Furthermore, the hybrid encapsulating of DEX significantly increased utilization of nano-drugs, improving the water solubility and bioavailability, which was conducive to developing the anti-inflammatory performance of USDPFs in the complex clinical environment. Discussion: The response-controlled release of DEX in the intrapulmonary microenvironment can reduce normal cell damage, which can effectively avoid the side effects of nano-drugs in anti-inflammatory application. Meanwhile, the multi-wavelength of UCNPs endowed nano-drugs with the fluorescence emission imaging capacity in an intrapulmonary microenvironment, providing precise guidance for LIRI.
Collapse
Affiliation(s)
- Xiaojing He
- Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Zhining Li
- Guangxi Medical University Cancer Hospital, Nanning, China
| | - Mengling Ye
- Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Chen Zhao
- Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Siyi Wu
- Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Yi Qin
- Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Youyuan Guo
- Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Lu Zhang
- Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| | - Fei Lin
- Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Anesthesiology, Nanning, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Nanning, China
- Guangxi Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Nanning, China
| |
Collapse
|
7
|
Johns CE, Gattu M, Camilli S, Desaraju A, Kolliputi N, Galam L. The Cd/Zn Axis: Emerging Concepts in Cellular Fate and Cytotoxicity. Biomolecules 2023; 13:biom13020316. [PMID: 36830685 PMCID: PMC9953193 DOI: 10.3390/biom13020316] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
Cadmium (Cd) is a toxic and carcinogenic substance that is present in the natural environment. The underlying biomolecular mechanisms of Cd toxicity are not completely understood, and it continues to be a significant research target due to its impact on public health. The primary routes of exposure are through ingestion of contaminated food and water and inhalation. Cd's long biological half-life of 10-30 years allows it to accumulate in the body, leading to organ dysfunction notably in the kidney, liver, bone, and lungs. Cd has similar biochemical characteristics to Zinc (Zn). It shares the import transporters, ZIP8 and ZIP14, to enter the cells. This competitive behavior can be observed in multiple instances throughout the progression of Cd toxicity. Future studies on the biochemical interactions of Cd and Zn will elucidate the potential protective effects of Zn supplementation in reducing the effects of Cd toxicity. In addition, research can be focused on discovering key proteins and effective pathways for Cd elimination that confer fewer adverse effects than current antioxidant therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Lakshmi Galam
- Correspondence: ; Tel.: +1-813-974-5419; Fax: +1-813-974-8575
| |
Collapse
|
8
|
Calabrese F, Pezzuto F, Fortarezza F, Lunardi F, Faccioli E, Lorenzoni G, Boscolo A, Sella N, Gregori D, Schiavon M, Navalesi P, Dell’Amore A, Rea F. Evaluation of Tissue Ischemia/Reperfusion Injury in Lung Recipients Supported by Intraoperative Extracorporeal Membrane Oxygenation: A Single-Center Pilot Study. Cells 2022; 11:cells11223681. [PMID: 36429108 PMCID: PMC9688824 DOI: 10.3390/cells11223681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Intraoperative veno-arterial (VA) extracorporeal membrane oxygenation (ECMO) as intraoperative hemodynamic support during lung transplantation is becoming a standard practice due to promising clinical results. Nevertheless, studies on tissue/molecular pathways investigating ischemia/reperfusion injury are still lacking. Patients receiving a bilateral lung transplantation between January 2012 and December 2018 at the University Hospital of Padova were included in this retrospective single-center observational study. The present study aimed to investigate ischemia/reperfusion injury in 51 tissue specimens obtained from 13 recipients supported by intraoperative VA-ECMO and 38 who were not. Several tissue analyses, including apoptosis evaluation and inducible nitric oxide synthase expression, were performed on the biopsies at the time of transplantation. Lung samples from the ECMO group (both pre- and post-reperfusion) were comparable, or for some parameters better, than samples from the non-ECMO group. Leukocyte margination was significantly lower in the ECMO group than in the non-ECMO group. Primary graft dysfunction, mainly at 24 and 48 h, was correlated with the tissue injury score of the post-reperfusion biopsy. The interquartile ranges for all morphological parameters showed high grade variability between pre- and post-reperfusion in the non-ECMO group. These preliminary data support the use of intraoperative ECMO based on lower lung tissue ischemia/reperfusion injury. Larger case series are mandatory to confirm our findings.
Collapse
Affiliation(s)
- Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35128 Padova, Italy
- Correspondence: ; Tel.: +39-0498272268
| | - Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35128 Padova, Italy
| | - Francesco Fortarezza
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35128 Padova, Italy
| | - Francesca Lunardi
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35128 Padova, Italy
| | - Eleonora Faccioli
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35128 Padova, Italy
| | - Giulia Lorenzoni
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35128 Padova, Italy
| | - Annalisa Boscolo
- Institute of Anesthesia and Intensive Care, Padova University Hospital, 35128 Padova, Italy
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Nicolò Sella
- Institute of Anesthesia and Intensive Care, Padova University Hospital, 35128 Padova, Italy
| | - Dario Gregori
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35128 Padova, Italy
| | - Marco Schiavon
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35128 Padova, Italy
| | - Paolo Navalesi
- Institute of Anesthesia and Intensive Care, Padova University Hospital, 35128 Padova, Italy
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Andrea Dell’Amore
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35128 Padova, Italy
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, 35128 Padova, Italy
| |
Collapse
|
9
|
Acetate, a gut bacterial product, ameliorates ischemia-reperfusion induced acute lung injury in rats. Int Immunopharmacol 2022; 111:109136. [PMID: 35964409 DOI: 10.1016/j.intimp.2022.109136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/27/2022] [Accepted: 08/05/2022] [Indexed: 11/21/2022]
Abstract
Recent data suggest that short-chain fatty acids (SCFAs), the major fermentation product from gut microbial degradation of dietary fiber, have protective effects against renal ischemia-reperfusion (IR) injury, colitis, and allergic asthma. However, the effect of SCFAs on acute lung injury (ALI) caused by IR is still unclear. In this study, we examine whether SCFAs have protective effects against IR-induced ALI and explore possible protective mechanisms. IR-induced ALI was established by 40 min ischemia followed by 60 min reperfusion in isolated perfused rat lungs. Rats were randomly assigned to one of six groups: control, control + acetate (400 mg/kg), IR, and IR + acetate at one of three dosages (100, 200, 400 mg/kg). Bronchoalveolar lavage fluids (BALF) and lung tissues were obtained and analyzed at the end of the experiment. In vitro, mouse lung epithelial cells (MLE-12) subjected to hypoxia-reoxygenation (HR) were pretreated with acetate (25 mmol/L) and GPR41 or GPR43 siRNA. Acetate decreased lung weight gain, lung weight/body weight ratios, wet/dry weight ratios, pulmonary artery pressure, and protein concentration of the BALF in a dose-dependent manner for IR-induced ALI. Acetate also significantly inhibited the production of TNF-α, IL-6 and CINC-1 in the BALF. Moreover, acetate treatment restored suppressed IκB-α levels and reduced nuclear NF-κB p65 levels in lung tissues. In addition, acetate mitigated IR-induced apoptosis and tight junction disruption in injured lung tissue. In vitro analyses showed that acetate attenuated NF-κB activation and KC/CXCL-1 levels in MLE-12 cells exposed to HR. The protective effects of acetate in vitro were significantly abrogated by GPR41 or GPR43 siRNA. Acetate ameliorates IR-induced acute lung inflammation and its protective mechanism appears to be via the GPR41/43 signaling pathway. Based on our findings, acetate may provide a novel adjuvant therapeutic approach for IR-induced lung injury.
Collapse
|
10
|
Zhou P, Song NC, Zheng ZK, Li YQ, Li JS. MMP2 and MMP9 contribute to lung ischemia-reperfusion injury via promoting pyroptosis in mice. BMC Pulm Med 2022; 22:230. [PMID: 35705936 PMCID: PMC9202153 DOI: 10.1186/s12890-022-02018-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 06/01/2022] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Lung ischemia-reperfusion injury (LIRI) is a cause of poor prognosis in several lung diseases and after lung transplantation. In LIRI, matrix metalloproteinases and pyroptosis indicators change in parallel, both of them involvement of inflammatory modulation, but it is unclear whether they are related to each other. METHODS We analyzed the matrix metalloproteinases (MMPs) changes from RNA sequencing (RNA-Seq) data of human transplantation and rat ischemia-reperfusion lung tissues in the Group on Earth Observations (GEO) database. Then established the mouse LIRI model to validate the changes. Further, the severity of lung injury was measured after intervening the matrix metalloproteinases changes with their selective inhibitor during Lung ischemia-reperfusion. Meanwhile, lung, pyroptosis was assessed by assaying the activity of Caspase-1 and interleukin 1β (IL-1β) before and after intervening the matrix metalloproteinases changes. RESULTS The RNA-Seq data revealed that matrix metallopeptidase 2 (MMP2), matrix metallopeptidase 9 (MMP9) mRNA expression was elevated both in human lung transplantation and rat lung ischemia-reperfusion tissues, consistent with the change in our mouse model. At the same time, the activity of Caspase-1 and IL-1β were increased after LIRI. While, the lung injury was attenuated for the use of MMP2 and MMP9 selective inhibitor SB-3CT. Likewise, lung pyroptosis alleviated when treatment the mice with SB-3CT in LIRI. CONCLUSION We conclude that MMP2 and MMP9 are involved in the process of LIRI, the mechanism of which is related to the promotion of lung pyroptosis.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nai-Cheng Song
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Kun Zheng
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yi-Qing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jin-Song Li
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
11
|
Shi ZA, Li TT, Kang DL, Su H, Tu FP. Fingolimod attenuates renal ischemia/reperfusion-induced acute lung injury by inhibiting inflammation and apoptosis and modulating S1P metabolism. J Int Med Res 2021; 49:3000605211032806. [PMID: 34340580 PMCID: PMC8358582 DOI: 10.1177/03000605211032806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective This study examined whether the immunomodulator fingolimod (FTY720) could alleviate renal ischemia/reperfusion (I/R)-induced lung injury and explored the potential mechanisms. Methods Renal I/R was established in a rat model, and FTY720 (0.5, 1, or 2 mg/kg) was injected intraperitoneally after 15 minutes of ischemia. Pro-inflammatory cytokine levels, oxidative stress, apoptosis, and the mRNA expression of the sphingosine-1-phosphate (S1P)-related signaling pathway genes sphingosine kinase-1 (SphK1) and sphingosine kinase-2 were analyzed in lung tissue. Results Increased pro-inflammatory cytokine levels; decreased total superoxide dismutase, catalase, and glutathione peroxidase levels; increased apoptosis; and increased S1P lyase and SphK1 expression were observed following renal I/R. FTY720 reversed renal I/R-induced changes and effectively attenuated lung injury. Conclusion FTY720 protected against acute lung injury in rats subjected to renal I/R by decreasing pulmonary inflammation and apoptosis, increasing oxidative stress, and modulating S1P metabolism.
Collapse
Affiliation(s)
- Zu-An Shi
- Department of Anesthesiology, Nanchong Central Hospital, the Second Clinical College of North Sichuan Medical College, Nanchong, P.R. China
| | - Ting-Ting Li
- Department of Pharmacy, the Second Affiliated Hospital of North Sichuan Medical College, Nanchong, P.R. China
| | - Dao-Ling Kang
- Department of Anesthesiology, 117913Affiliated Hospital of North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong, P.R. China
| | - Hang Su
- Department of Anesthesiology, 117913Affiliated Hospital of North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong, P.R. China
| | - Fa-Ping Tu
- Department of Anesthesiology, 117913Affiliated Hospital of North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong, P.R. China
| |
Collapse
|
12
|
Tang SE, Liao WI, Pao HP, Hsu CW, Wu SY, Huang KL, Chu SJ. Poloxamer 188 Attenuates Ischemia-Reperfusion-Induced Lung Injury by Maintaining Cell Membrane Integrity and Inhibiting Multiple Signaling Pathways. Front Pharmacol 2021; 12:650573. [PMID: 34335242 PMCID: PMC8319770 DOI: 10.3389/fphar.2021.650573] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 07/05/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Poloxamer 188 (P188) possesses anti-inflammatory properties and can help to maintain plasma membrane function. P188 has been reported to exert beneficial effects in the treatment of various disorders. However, the effects of P188 in ischemia/reperfusion (IR)-induced acute lung injury have not been examined. Methods: We investigated the ability of P188 to attenuate IR-induced acute lung injury in rats and hypoxia/reoxygenation (HR) injury in murine epithelial cells. Isolated perfused rat lungs were exposed to 40 min ischemia followed by 60 min reperfusion to induce IR injury. Results: IR led to lung edema, increased pulmonary arterial pressure, promoted lung tissue inflammation and oxidative stress, and upregulated the levels of TNF-α, IL-6 and CINC-1, and increased Lactic dehydrogenase (LDH) activity in bronchoalveolar lavage fluid. IR also downregulated the levels of inhibitor of κB (IκB-α), upregulated nuclear factor (NF)-κB (NF-κB), and promoted apoptosis in lung tissues. P188 significantly suppressed all these effects. In vitro, P188 also exerted a similar effect in murine lung epithelial cells exposed to HR. Furthermore, P188 reduced the number of propidium iodide-positive cells, maintained cell membrane integrity, and enhanced cell membrane repair following HR. Conclusion: We conclude that P188 protects against lung IR injury by suppressing multiple signaling pathways and maintaining cell membrane integrity.
Collapse
Affiliation(s)
- Shih-En Tang
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan.,Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chin-Wang Hsu
- Department of Emergency and Critical Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan.,Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shi-Jye Chu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
13
|
Lung Transplantation, Pulmonary Endothelial Inflammation, and Ex-Situ Lung Perfusion: A Review. Cells 2021; 10:cells10061417. [PMID: 34200413 PMCID: PMC8229792 DOI: 10.3390/cells10061417] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/31/2022] Open
Abstract
Lung transplantation (LTx) is the gold standard treatment for end-stage lung disease; however, waitlist mortality remains high due to a shortage of suitable donor lungs. Organ quality can be compromised by lung ischemic reperfusion injury (LIRI). LIRI causes pulmonary endothelial inflammation and may lead to primary graft dysfunction (PGD). PGD is a significant cause of morbidity and mortality post-LTx. Research into preservation strategies that decrease the risk of LIRI and PGD is needed, and ex-situ lung perfusion (ESLP) is the foremost technological advancement in this field. This review addresses three major topics in the field of LTx: first, we review the clinical manifestation of LIRI post-LTx; second, we discuss the pathophysiology of LIRI that leads to pulmonary endothelial inflammation and PGD; and third, we present the role of ESLP as a therapeutic vehicle to mitigate this physiologic insult, increase the rates of donor organ utilization, and improve patient outcomes.
Collapse
|
14
|
Shen Y, Lu H, Song G. MiR-221-3p and miR-92a-3p enhances smoking-induced inflammation in COPD. J Clin Lab Anal 2021; 35:e23857. [PMID: 34097306 PMCID: PMC8274981 DOI: 10.1002/jcla.23857] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/29/2021] [Accepted: 04/10/2021] [Indexed: 12/23/2022] Open
Abstract
Background Smoking is likely to facilitate airway inflammation and finally contributes to chronic obstructive pulmonary disease (COPD). This investigation was intended to elucidate miRNAs that were involved in smoking‐induced COPD. Methods Altogether 155 COPD patients and 77 healthy volunteers were recruited, and their serum levels of miR‐221‐3p and miR‐92a‐3p were determined. Besides, human bronchial epithelial cells (16HBECs) were purchased, and they were treated by varying concentrations of cigarette smoke extract (CSE). The 16HBECs were, additionally, transfected by miR‐221‐3p mimic, miR‐92a‐3p mimic, miR‐221‐3p inhibitor or miR‐92a‐3p inhibitor, and cytokines released by them, including TNF‐α, IL‐8, IL‐1β, and TGF‐β1, were monitored using enzyme linked immunosorbent assay (ELISA) kits. Results Chronic obstructive pulmonary disease patients possessed higher serum levels of miR‐221‐3p and miR‐92a‐3p than healthy volunteers (p < 0.05), and both miR‐221‐3p and miR‐92a‐3p were effective biomarkers in diagnosing stable COPD from acute exacerbation COPD. Moreover, viability of 16HBECs was undermined by CSE treatment (p < 0.05), and exposure to CSE facilitated 16HBECs’ release of TNF‐α, IL‐8, IL‐1β, and TGF‐β1 (p < 0.05). Furthermore, miR‐221‐3p/miR‐92a‐3p expression in 16HBECs was significantly suppressed after transfection of miR‐221‐3p/miR‐92a‐3p inhibitor (p < 0.05), which abated CSE‐triggered increase in cytokine production and decline in viability of 16HBECs (p < 0.05). Conclusion MiR‐221‐3p and miR‐92a‐3p were involved in CSE‐induced hyperinflammation of COPD, suggesting that they were favorable alternatives in diagnosing COPD patients with smoking history.
Collapse
Affiliation(s)
- Yahui Shen
- Department of Respiratory and Critical Care Medicine, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, China
| | - Huiyu Lu
- Department of Respiratory and Critical Care Medicine, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, China
| | - Guixian Song
- Department of Cardiology, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, China
| |
Collapse
|
15
|
Pan J, Chen S, Guo W, Cao S, Shi X, Zhang J, Zhang H, Zhang S. Alpinetin protects against hepatic ischemia/reperfusion injury in mice by inhibiting the NF-κB/MAPK signaling pathways. Int Immunopharmacol 2021; 95:107527. [PMID: 33743314 DOI: 10.1016/j.intimp.2021.107527] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/05/2021] [Accepted: 02/21/2021] [Indexed: 12/22/2022]
Abstract
Liver damage induced by ischemia/reperfusion (I/R) remains a primary issue in liver transplantation and resection. Alpinetin, a novel plant flavonoid derived from Alpinia katsumadai Hayata, is widely used to treat various inflammatory diseases. However, the effects of alpinetin on hepatic I/R injury remain unclear. The present study investigated the protective effects of alpinetin pretreatment on hepatic I/R injury in mice. C57BL/6 mice were subjected to 1 h of partial hepatic ischemia followed by 6 h of reperfusion. Alpinetin (50 mg/kg) was given by intraperitoneal injection 1 h before liver ischemia. The blood and liver tissues were collected to assess biochemical indicators, hepatocyte damage, and levels of proteins related to signaling pathways. Furthermore, a hepatocytes hypoxia/reoxygenation (H/R) model was established for in vitro experiments. In vivo, we observed that alpinetin significantly attenuated the increases in alanine aminotransferase, aspartate transaminase, proinflammatory cytokines, hepatocyte damage, and apoptosis caused by hepatic I/R. Moreover, the hepatic I/R-induced nuclear factor kappa-B (NF-κB)/mitogen-activated protein kinase (MAPK) pathways were suppressed by alpinetin. In vitro, we also observed that alpinetin inhibited the inflammatory response, apoptosis, and activation of the NF-κB/MAPK pathways in hepatocytes after H/R treatment. Our data indicate that alpinetin ameliorated the inflammatory response and apoptosis induced by hepatic I/R injury in mice. The protective effects of alpinetin on hepatic I/R injury may be due to its ability to inhibit the NF-κB/MAPK signaling pathways. These results suggest that alpinetin is a promising potential therapeutic reagent for hepatic I/R injury.
Collapse
Affiliation(s)
- Jie Pan
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, China; ZhengZhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, China
| | - Sanyang Chen
- Department of Emergency Surgery, the First Affiliated Hospital of Zhengzhou University, China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, China; ZhengZhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, China
| | - Shengli Cao
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, China; ZhengZhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, China
| | - Xiaoyi Shi
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, China; ZhengZhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, China
| | - Jiakai Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, China; ZhengZhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, China
| | - Huapeng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, China; ZhengZhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, China; ZhengZhou Key Laboratory of Hepatobiliary & Pancreatic Diseases and Organ Transplantation, China.
| |
Collapse
|
16
|
Bag-1L Protects against Cell Apoptosis in an In Vitro Model of Lung Ischemia-Reperfusion Injury through the C-Terminal "Bag" Domain. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8822807. [PMID: 34056003 PMCID: PMC8123090 DOI: 10.1155/2021/8822807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/13/2021] [Accepted: 02/08/2021] [Indexed: 11/17/2022]
Abstract
Bcl-2-associated athanogene 1 (Bag-1) is a multifunctional and antiapoptotic protein that binds to the antiapoptosis regulator Bcl-2 and promotes cell survival. To investigate the protective function of Bag-1, we examined the effects of Bag-1L, one isoform of Bag-1, in an in vitro cell culture model of lung ischemia-reperfusion injury (LIRI) generated by treatment of A549 cells with hypoxia/reoxygenation. Overexpression of full-length Bag-1L increased the viability of A549 cells and reduced cell apoptosis in response to 6 h of hypoxia/reoxygenation treatment. Furthermore, Bag-1L overexpression enhanced the heat shock protein 70 (HSP70) and Bcl-2 protein levels, increased the phosphorylation of AKT, decreased Bax and cleaved caspase-3 levels, and was able to overcome cell cycle arrest. These effects were not observed in A549 cells overexpressing a truncated form of Bag-1L lacking the "Bag domain," denoted Bag-1L△C. The "Bag domain" is the C-terminal 47 amino acids. Taken together, the results of this study suggest that Bag-1L overexpression can protect against oxidative stress and apoptosis in an in vitro LIRI model, with a dependence on the Bag domain.
Collapse
|
17
|
Liao WI, Wu SY, Tsai SH, Pao HP, Huang KL, Chu SJ. 2-Methoxyestradiol Protects Against Lung Ischemia/Reperfusion Injury by Upregulating Annexin A1 Protein Expression. Front Immunol 2021; 12:596376. [PMID: 33796096 PMCID: PMC8007881 DOI: 10.3389/fimmu.2021.596376] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/22/2021] [Indexed: 11/13/2022] Open
Abstract
Background: 2-Methoxyestradiol (2ME), a natural 17-β estradiol metabolite, is a potent anti-inflammatory agent, but its effect on ischemia/reperfusion (IR)-induced acute lung inflammation remains unknown. Annexin A1 (AnxA1), a glucocorticoid-regulated protein, is effective at inhibiting neutrophil transendothelial migration by binding the formyl peptide receptors (FPRs). We aimed to investigate whether 2ME upregulates the expression of AnxA1 and protects against IR-induced lung damage. Methods: IR-mediated acute lung inflammation was induced by ischemia for 40 min followed by reperfusion for 60 min in an isolated, perfused rat lung model. The rat lungs were randomly treated with vehicle or 2ME, and the functional relevance of AnxA1 was determined using an anti-AnxA1 antibody or BOC2 (a pan-receptor antagonist of the FPR). In vitro, human primary alveolar epithelial cells (HPAECs) and rat neutrophils were pretreated with 2ME and an AnxA1 siRNA or anti-AnxA1 antibody and subjected to hypoxia-reoxygenation (HR). Results: 2ME significantly decreased all lung edema parameters, neutrophil infiltration, oxidative stress, proinflammatory cytokine production, lung cell apoptosis, tight junction protein disruption, and lung tissue injury in the IR-induced acute lung inflammation model. 2ME also increased the expression of the AnxA1 mRNA and protein and suppressed the activation of nuclear factor-κB (NF-κB). In vitro, 2ME attenuated HR-triggered NF-κB activation and interleukin-8 production in HPAECs, decreased transendothelial migration, tumor necrosis factor-α production, and increased apoptosis in neutrophils exposed to HR. These protective effects of 2ME were significantly abrogated by BOC2, the anti-AnxA1 antibody, or AnxA1 siRNA. Conclusions: 2ME ameliorates IR-induced acute lung inflammation by increasing AnxA1 expression. Based on these results, 2ME may be a promising agent for attenuating IR-induced lung injury.
Collapse
Affiliation(s)
- Wen-I Liao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hung Tsai
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shi-Jye Chu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
18
|
Chen SY, Zhang HP, Li J, Shi JH, Tang HW, Zhang Y, Zhang JK, Wen PH, Wang ZH, Shi XY, He YT, Hu BW, Yang H, Guo WZ, Zhang SJ. Tripartite Motif-Containing 27 Attenuates Liver Ischemia/Reperfusion Injury by Suppressing Transforming Growth Factor β-Activated Kinase 1 (TAK1) by TAK1 Binding Protein 2/3 Degradation. Hepatology 2021; 73:738-758. [PMID: 32343849 PMCID: PMC7898667 DOI: 10.1002/hep.31295] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/31/2020] [Accepted: 04/07/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Hepatic ischemia-reperfusion (I/R) injury, which mainly involves inflammatory responses and apoptosis, is a common cause of organ dysfunction in liver transplantation (LT). As a critical mediator of inflammation and apoptosis in various cell types, the role of tripartite motif-containing (TRIM) 27 in hepatic I/R injury remains worthy of study. APPROACH AND RESULTS This study systemically evaluated the putative role of TRIM27/transforming growth factor β-activated kinase 1 (TAK1)/JNK (c-Jun N-terminal kinase)/p38 signaling in hepatic I/R injury. TRIM27 expression was significantly down-regulated in liver tissue from LT patients, mice subjected to hepatic I/R surgery, and hepatocytes challenged by hypoxia/reoxygenation (H/R) treatment. Subsequently, using global Trim27 knockout mice (Trim27-KO mice) and hepatocyte-specific Trim27 transgenic mice (Trim27-HTG mice), TRIM27 functions to ameliorate liver damage, reduce the inflammatory response, and prevent cell apoptosis. In parallel in vitro studies, activating TRIM27 also prevented H/R-induced hepatocyte inflammation and apoptosis. Mechanistically, TRIM27 constitutively interacted with the critical components, TAK1 and TAK1 binding protein 2/3 (TAB2/3), and promoted the degradation of TAB2/3, leading to inactivation of TAK1 and the subsequent suppression of downstream JNK/p38 signaling. CONCLUSIONS TRIM27 is a key regulator of hepatic I/R injury by mediating the degradation of TAB2/3 and suppression of downstream TAK1-JNK/p38 signaling. TRIM27 may be a promising approach to protect the liver against I/R-mediated hepatocellular damage in transplant recipients.
Collapse
Affiliation(s)
- San-Yang Chen
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Hua-Peng Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Jie Li
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Ji-Hua Shi
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Hong-Wei Tang
- Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Yi Zhang
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jia-Kai Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Pei-Hao Wen
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Zhi-Hui Wang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Xiao-Yi Shi
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Yu-Ting He
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Bo-Wen Hu
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Han Yang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Wen-Zhi Guo
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| | - Shui-Jun Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina.,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouChina.,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouChina
| |
Collapse
|
19
|
Ischemia-reperfusion Injury in the Transplanted Lung: A Literature Review. Transplant Direct 2021; 7:e652. [PMID: 33437867 PMCID: PMC7793349 DOI: 10.1097/txd.0000000000001104] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 02/07/2023] Open
Abstract
Lung ischemia-reperfusion injury (LIRI) and primary graft dysfunction are leading causes of morbidity and mortality among lung transplant recipients. Although extensive research endeavors have been undertaken, few preventative and therapeutic treatments have emerged for clinical use. Novel strategies are still needed to improve outcomes after lung transplantation. In this review, we discuss the underlying mechanisms of transplanted LIRI, potential modifiable targets, current practices, and areas of ongoing investigation to reduce LIRI and primary graft dysfunction in lung transplant recipients.
Collapse
|
20
|
Oshima Y, Otsuki A, Endo R, Nakasone M, Harada T, Takahashi S, Inagaki Y. The Effects of Volatile Anesthetics on Lung Ischemia-Reperfusion Injury: Basic to Clinical Studies. J Surg Res 2020; 260:325-344. [PMID: 33373852 DOI: 10.1016/j.jss.2020.11.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/01/2020] [Indexed: 02/08/2023]
Abstract
Case reports from as early as the 1970s have shown that intravenous injection of even a small dose of volatile anesthetics result in fatal lung injury. Direct contact between volatile anesthetics and pulmonary vasculature triggers chemical damage in the vessel walls. A wide variety of factors are involved in lung ischemia-reperfusion injury (LIRI), such as pulmonary endothelial cells, alveolar epithelial cells, alveolar macrophages, neutrophils, mast cells, platelets, proinflammatory cytokines, and surfactant. With a constellation of factors involved, the assessment of the protective effect of volatile anesthetics in LIRI is difficult. Multiple animal studies have reported that with regards to LIRI, sevoflurane demonstrates an anti-inflammatory effect in immunocompetent cells and an anti-apoptotic effect on lung tissue. Scattered studies have dismissed a protective effect of desflurane against LIRI. While a single-center randomized controlled trial (RCT) found that volatile anesthetics including desflurane demonstrated a lung-protective effect in thoracic surgery, a multicenter RCT did not demonstrate a lung-protective effect of desflurane. LIRI is common in lung transplantation. One study, although limited due to its small sample size, found that the use of volatile anesthetics in organ procurement surgery involving "death by neurologic criteria" donors did not improve lung graft survival. Future studies on the protective effect of volatile anesthetics against LIRI must examine not only the mechanism of the protective effect but also differences in the effects of different types of volatile anesthetics, their optimal dosage, and the appropriateness of their use in the event of marked alveolar capillary barrier damage.
Collapse
Affiliation(s)
- Yoshiaki Oshima
- Department of Anesthesiology, Yonago Medical Center, Yonago, Tottori, Japan.
| | - Akihiro Otsuki
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Ryo Endo
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Masato Nakasone
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Tomomi Harada
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Shunsaku Takahashi
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Yoshimi Inagaki
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| |
Collapse
|
21
|
Calabrese F, Schiavon M, Perissinotto E, Lunardi F, Marulli G, Di Gregorio G, Pezzuto F, Edith Vuljan S, Forin E, Wiegmann B, Jonigk D, Warnecke G, Rea F. Organ Care System Lung resulted in lower apoptosis and iNOS expression in donor lungs. Am J Transplant 2020; 20:3639-3648. [PMID: 32652873 DOI: 10.1111/ajt.16187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/24/2020] [Accepted: 06/28/2020] [Indexed: 01/25/2023]
Abstract
Ischemia-reperfusion (IR) injury after lung transplantation is still today an important complication in up to 25% of patients. The Organ Care System (OCS) Lung, an advanced normothermic ex vivo lung perfusion system, was found to be effective in reducing primary graft dysfunction compared to standard organ care (SOC) but studies on tissue/molecular pathways that could explain these more effective clinical results are lacking. This observational longitudinal study aimed to investigate IR injury in 68 tissue specimens collected before and after reperfusion from 17 OCS and 17 SOC preserved donor lungs. Several tissue analyses including apoptosis evaluation and inducible nitric oxide synthase (iNOS) expression (by immunohistochemistry and real-time reverse transcriptase-polymerase chain reaction) were performed. Lower iNOS expression and apoptotic index were distinctive of OCS preserved tissues at pre- and post-reperfusion times, independently from potential confounding factors. Moreover, OCS recipients had lower acute cellular rejection at the first 6-month follow-up. In conclusion, IR injury, in terms of apoptosis and iNOS expression, was less frequent in OCS- than in SOC-preserved lungs, which could eventually explain a better clinical outcome. Further studies are needed to validate our data and determine the role of iNOS expression as a predictive biomarker of the complex IR injury mechanism.
Collapse
Affiliation(s)
- Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Marco Schiavon
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Egle Perissinotto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Francesca Lunardi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Giuseppe Marulli
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | | | - Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Stefania Edith Vuljan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Edoardo Forin
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Bettina Wiegmann
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Center of Lung Research, Hannover, Germany
| | - Danny Jonigk
- German Center of Lung Research, Hannover, Germany.,Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Gregor Warnecke
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Center of Lung Research, Hannover, Germany
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
22
|
Leligdowicz A, Ross JT, Nesseler N, Matthay MA. The endogenous capacity to produce proinflammatory mediators by the ex vivo human perfused lung. Intensive Care Med Exp 2020; 8:56. [PMID: 32955627 PMCID: PMC7505905 DOI: 10.1186/s40635-020-00343-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/04/2020] [Indexed: 12/19/2022] Open
Abstract
Background The ex vivo human perfused lung model has enabled optimizing donor lungs for transplantation and delineating mechanisms of lung injury. Perfusate and airspace biomarkers are a proxy of the lung response to experimental conditions. However, there is a lack of studies evaluating biomarker kinetics during perfusion and after exposure to stimuli. In this study, we analyzed the ex vivo-perfused lung response to three key perturbations: exposure to the perfusion circuit, exogenous fresh whole blood, and bacteria. Results Ninety-nine lungs rejected for transplantation underwent ex vivo perfusion. One hour after reaching experimental conditions, fresh whole blood was added to the perfusate (n = 55). Two hours after reaching target temperature, Streptococcus pneumoniae was added to the perfusate (n = 42) or to the airspaces (n = 17). Perfusate and airspace samples were collected at baseline (once lungs were equilibrated for 1 h, but before blood or bacteria were added) and 4 h later. Interleukin (IL)-6, IL-8, angiopoietin (Ang)-2, and soluble tumor necrosis factor receptor (sTNFR)-1 were quantified. Baseline perfusate and airspace biomarker levels varied significantly, and this was not related to pre-procurement PaO2:FiO2 ratio, cold ischemia time, and baseline alveolar fluid clearance (AFC). After 4 h of ex vivo perfusion, the lung demonstrated a sustained production of proinflammatory mediators. The change in biomarker levels was not influenced by baseline donor lung characteristics (cold ischemia time, baseline AFC) nor was it associated with measures of experimental epithelial (final AFC) or endothelial (percent weight gain) injury. In the presence of exogenous blood, the rise in biomarkers was attenuated. Lungs exposed to intravenous (IV) bacteria relative to control lungs demonstrated a significantly higher rise in perfusate IL-6. Conclusions The ex vivo-perfused lung has a marked endogenous capacity to produce inflammatory mediators over the course of short-term perfusion that is not significantly influenced by donor lung characteristics or the presence of exogenous blood, and only minimally affected by the introduction of systemic bacteremia. The lack of association between biomarker change and donor lung cold ischemia time, final alveolar fluid clearance, and experimental percent weight gain suggests that the maintained ability of the human lung to produce biomarkers is not merely a marker of lung epithelial or endothelial injury, but may support the function of the lung as an immune cell reservoir.
Collapse
Affiliation(s)
- Aleksandra Leligdowicz
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA. .,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - James T Ross
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Nicolas Nesseler
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA.,Department of Anesthesia and Critical Care, Pontchaillou, University Hospital of Rennes, Rennes, France.,Univ Rennes, CHU de Rennes, Inra, Inserm, Institut NUMECAN-UMR_A 1341, UMR_S 1241, 35000, Rennes, France.,Univ Rennes, CHU Rennes, Inserm, CIC 1414 (Centre d'Investigation Clinique de Rennes), 35000, Rennes, France
| | - Michael A Matthay
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA.,Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA, USA.,Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
23
|
Kong Q, Yuan M, Ming T, Fang Q, Wu X, Song X. Expression and regulation of tumor necrosis factor-α-induced protein-8-like 2 is associated with acute lung injury induced by myocardial ischemia reperfusion in diabetic rats. Microvasc Res 2020; 130:104009. [PMID: 32333940 DOI: 10.1016/j.mvr.2020.104009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/17/2020] [Accepted: 04/17/2020] [Indexed: 10/24/2022]
Abstract
AIMS The purpose of the present study was to investigate the possible role of TIPE2 on acute lung injury (ALI) induced by myocardial ischemia/reperfusion (MIR) in diabetic rats. METHODS Sprague-Dawley (SD) rats were randomly separated into four groups: control+sham (C + sham); control+MIR (C + MIR); diabetes+sham (D + sham); diabetes+MIR (D + MIR). Diabetes was induced using streptozotocin. Eight weeks after diabetes induction, MIR was conducted. At 2 h after MIR, myocardial injury indices were assessed; arterial blood, bronchoalveolar lavage fluid (BALF) and lung tissues were collected for corresponding detection. RESULTS Rats subjected to MIR showed serious ALI (estimated via pathological changes, lung injury score and Wet/Dry weight ratio), lung inflammation and pulmonary cell apoptosis compared with sham groups, especially in D + MIR group. Evaluation of protein expression in lung tissues showed that p-JNK and nuclear NF-κB p65 protein levels were higher in D + MIR group as compared with C + MIR group. Besides, either hyperglycemia or MIR can significantly upregulate TIPE2 protein levels. CONCLUSIONS In conclusion, diabetic lungs are more susceptible to MIR. TIPE2 may involve in this pathological process, possibly through regulation of inflammation, oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Qian Kong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Min Yuan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Tingqian Ming
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Qing Fang
- Department of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China.
| | - Xuemin Song
- Department of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China.
| |
Collapse
|
24
|
Qiao X, Yang Y, Huang R, Shi X, Chen H, Wang J, Chen Y, Tan Y, Tan Z. E-Jet 3D-Printed Scaffolds as Sustained Multi-Drug Delivery Vehicles in Breast Cancer Therapy. Pharm Res 2019; 36:182. [PMID: 31741089 DOI: 10.1007/s11095-019-2687-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/15/2019] [Indexed: 01/09/2023]
Abstract
PURPOSE Combination chemotherapy is gradually receiving more attention because of its potential synergistic effect and reduced drug doses in clinical application. However, how to precisely control drug release dose and time using vehicles remains a challenge. This work developed an efficient drug delivery system to combat breast cancer, which can enhance drug effects despite reducing its concentration. METHODS Controlled-release poly-lactic-co-glycolic acid (PLGA) scaffolds were fabricated by E-jet 3D printing to deliver doxorubicin (DOX) and cisplatin (CDDP) simultaneously. RESULTS This drug delivery system allowed the use of a reduced drug dosage resulting in a better effect on the human breast cancer cell apoptosis and inhibiting tumor growth, compared with the effect of each drug and the two drugs administrated without PLGA scaffolds. Our study suggested that DOX-CDDP-PLGA scaffolds could efficiently destroy MDA-MB-231 cells and restrain tumor growth. CONCLUSIONS The 3D printed PLGA scaffolds with their time-programmed drug release might be useful as a new multi-drug delivery vehicle in cancer therapy, which has a potential advantage in a long term tumor cure and prevention of tumor recurrence.
Collapse
Affiliation(s)
- Xiaoyin Qiao
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Yikun Yang
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Ruiying Huang
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Xuelei Shi
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Haoxiang Chen
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Jian Wang
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Yanxiang Chen
- Department of Obstetrics and Gynecology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China.
| | - Yongjun Tan
- College of Biology, Hunan University, Changsha, 410082, Hunan, China
| | - Zhikai Tan
- College of Biology, Hunan University, Changsha, 410082, Hunan, China. .,Shenzhen Institute, Hunan University, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
25
|
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: 0.6] [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
|
26
|
Yeşil H, Tuğlu I. The relation of oxidative stress and apoptosis to histopathologic alterations in the lungs as a result of global cerebral ischemia. Biotech Histochem 2019; 94:555-568. [PMID: 31373845 DOI: 10.1080/10520295.2019.1601768] [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: 10/26/2022] Open
Abstract
Heart attack and oxygen deficiency may cause necrosis in the brain and other tissues. We investigated the histopathological effects of nitric oxide (NO) on ischemia/reperfusion in lung and hippocampus using a rat brain bilateral occlusion ischemia model. Male rats were assigned to sham (SH), ischemic preconditioning (PC), global ischemia (GI) and ischemic reperfusion (IR) groups. Before ischemia was induced, blood was drawn to induce hypovolemic hypotension and for blood gas testing. After sacrifice, samples of hippocampus were harvested. Sections were examined using hematoxylin and eosin (H & E) staining and immunostaining using primary antibodies for GFAP, S100β, iNOS, eNOS and the TUNEL method. Following ischemia, we found evidence of gliosis induced oxidative stress and apoptosis in the hippocampus. No significant differences were detected between the SH and PC groups. In the GI and IR groups, apoptosis and necrosis were observed in the hippocampus. Lung sections were stained with H & E and Masson's trichrome (MT) and immunostained for iNOS and eNOS. The TUNEL method was used to detect apoptosis. Interstitial edema, vascular congestion, intra-alveolar hemorrhage, perivascular edema, neutrophil infiltration and disruption of alveoli were observed after global ischemia and ischemic reperfusion. Inflammatory cells were detected in the connective tissue. The IR and GI groups exhibited significantly more apoptotic cells than the SH or PC groups. Free radicals, such as nitric oxide (NO), that appear following ischemia and reperfusion in the brain may also injure the lungs. Increased NO in both lung and brain tissue suggests that apoptosis in these organs can be induced by reactive nitrogen species.
Collapse
Affiliation(s)
- H Yeşil
- Departments of Midwifery, Celal Bayar University Manisa, Manisa, Turkey
| | - I Tuğlu
- Histology and Embryology, Faculty of Medicine, Celal Bayar University Manisa, Manisa, Turkey
| |
Collapse
|
27
|
Zhai C, Lv J, Wang K, Li Q, Qu Y. HSP70 silencing aggravates apoptosis induced by hypoxia/reoxygenation in vitro. Exp Ther Med 2019; 18:1013-1020. [PMID: 31363363 PMCID: PMC6614734 DOI: 10.3892/etm.2019.7697] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 02/28/2019] [Indexed: 01/09/2023] Open
Abstract
Lung ischemia-reperfusion can cause acute lung injury, which is closely associated with apoptosis. Heat shock protein 70 (HSP70) is an anti-apoptotic protein that promotes cell survival under a variety of different stress conditions. However, the role and mechanism of HSP70 in lung ischemia-reperfusion injury is yet to be fully elucidated. In the present study, an in vitro hypoxia/reoxygenation model of A549 cells was established to simulate lung ischemia-reperfusion and HSP70 was silenced by transfecting A549 cells with an shRNA sequence targeting HSP70. Western blotting, reverse transcription-quantitative polymerase chain reaction, Cell Counting kit-8 and flow cytometry were used to detect protein levels, RNA expression, cell activity and apoptosis. The results revealed that silencing HSP70 reduced cell viability, aggravated apoptosis, increased lactate dehydrogenase levels and induced a G2/M blockade in a hypoxia-reoxygenation A549 cell model. Furthermore, silencing HSP70 decreased the phosphorylation levels of protein kinase B (AKT) and extracellular signal-regulated kinase (ERK); however, the total AKT and ERK levels did not change significantly. Pretreating A549 cells with the AKT pathway inhibitor, LY294002 and the ERK pathway inhibitor, U0216 led to a decrease in HSP70 expression. These results indicate that silencing HSP70 may aggravate apoptosis in hypoxia-reoxygenation cell models, potentially via the mitogen-activated protein kinase/ERK and phosphoinositide 3-kinase/AKT signaling pathways.
Collapse
Affiliation(s)
- Congying Zhai
- Intensive Care Unit, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong 266071, P.R. China.,Department of Respiratory Medicine, Zibo First Hospital, Zibo, Shandong 255200, P.R. China
| | - Jiling Lv
- Department of Respiratory Medicine, Zibo First Hospital, Zibo, Shandong 255200, P.R. China
| | - Keke Wang
- Department of Emergency, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Qingshu Li
- Intensive Care Unit, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Yan Qu
- Intensive Care Unit, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
28
|
Li Q, Cui S, Jing G, Ding H, Xia Z, He X. The role of PI3K/Akt signal pathway in the protective effects of propofol on intestinal and lung injury induced by intestinal ischemia/reperfusion1. Acta Cir Bras 2019; 34:e20190010000005. [PMID: 30785506 PMCID: PMC6585923 DOI: 10.1590/s0102-865020190010000005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/15/2019] [Indexed: 12/15/2022] Open
Abstract
Purpose To investigate the role of PI3k/Akt signal pathway in the protective effects
of propofol on intestinal and lung injury induced by intestinal
ischemia/reperfusion(I/R). Methods Male Sprague-Dawley rats were subjected to 45 min of ischemia by occluding
the superior mesenteric artery and to 2h of reperfusion to establish the
model of I/R. Twenty four rats were randomly divided into four groups: Sham,
intestinal I/R (II/R), propofol (P), wortmannin (W). In groups P, W,
propofol was injected intravenously and continuously at the onset of
reperfusion via infusion pump. PI3K inhibitor (wortmannin) was administered
intravenously in group W 25 min before ischemia. Intestinal tissues and lung
tissues were obtained for determination of histologic injury, wet/dry weight
ratio, malondialdehyde (MDA) levels, superoxide dismutase (SOD) and
myeloperoxidase (MPO) activities. Meanwhile, the expressions of caspase-3
and phosphorylated Akt (p-Akt) in intestines and lungs were detected by
western blot. Results Propofol treatment alleviated intestinal and lung morphological changes which
were observed in II/R group , Moreover, wet/dry weight ratio, the MDA level,
MPO activity and expression of caspase-3 were significantly decreased
whereas the SOD activity and p-Akt expression were significantly increased.
Notably, the protections were significantly reversed by pretreatment of
wortmannin. Conclusion: PI3K/Akt pathway activation play a critical role in the protective effects of
propofol on intestinal and lung injury induced by ischemia/reperfusion.
Collapse
Affiliation(s)
- Qingwen Li
- MD, Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei, China. Manuscript preparation and writng
| | - Shanshan Cui
- MD, Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei, China. Conception and design of the study, acquisiton of data
| | - Guoqing Jing
- MD, Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei, China. Analysis and interpretation of data, technical procedures
| | - Huang Ding
- MD, Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei, China. Histopathological examinations, statistics analysis
| | - Zhongyuan Xia
- MD, Department of Anesthesiology, Renmin Hospital of Wuhan University, Hubei, China. Final approval
| | - Xianghu He
- MD, Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Hubei, China. Critical revision
| |
Collapse
|
29
|
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: 133] [Impact Index Per Article: 26.6] [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
|
30
|
Garutti I, Gonzalez-Moraga F, Sanchez-Pedrosa G, Casanova J, Martin-Piñeiro B, Rancan L, Simón C, Vara E. The effect of anesthetic preconditioning with sevoflurane on intracellular signal-transduction pathways and apoptosis, in a lung autotransplant experimental model. BRAZILIAN JOURNAL OF ANESTHESIOLOGY (ENGLISH EDITION) 2019. [PMID: 30459087 PMCID: PMC9391783 DOI: 10.1016/j.bjane.2018.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background Anesthetic pre-conditioning attenuates inflammatory response during ischemia-reperfusion lung injury. The molecular mechanisms to explain it are not fully understood. The aim of our investigation was to analyze the molecular mechanism that explain the anti-inflammatory effects of anesthetic pre-conditioning with sevoflurane focusing on its effects on MAPKs (mitogen-activated protein kinases), NF-κB (nuclear factor kappa beta) pathways, and apoptosis in an experimental lung autotransplant model. Methods Twenty large white pigs undergoing pneumonectomy plus lung autotransplant were divided into two 10-member groups on the basis of the anesthetic received (propofol or sevoflurane). Anesthetic pre-conditioning group received sevoflurane 3% after anesthesia induction and it stopped when one-lung ventilation get started. Control group did not receive sevoflurane in any moment during the whole study period. Intracellular signal-transduction pathways (MAPK family), transcription factor (NF-κB), and apoptosis (caspases 3 and 9) were analyzed during experiment. Results Pigs that received anesthetic pre-conditioning with sevoflurane have shown significant lower values of MAPK-p38, MAPK-P-p38, JNK (c-Jun N-terminal kinases), NF-κB p50 intranuclear, and caspases (p < 0.05) than pigs anesthetized with intravenous propofol. Conclusions Lung protection of anesthetic pre-conditioning with sevoflurane during experimental lung autotransplant is, at least, partially associated with MAPKs and NF κB pathways attenuation, and antiapoptotic effects.
Collapse
Affiliation(s)
- Ignacio Garutti
- Hospital General Universitario Gregorio Marañon, Departamento de Anestesiologia, Madri, Espanha; Universidad Complutense de Madrid, Departamento de Farmacologia, Madri, Espanha.
| | | | | | - Javier Casanova
- Hospital General Universitario Gregorio Marañon, Departamento de Anestesiologia, Madri, Espanha
| | | | - Lisa Rancan
- Universidad Complutense de Madrid, Facultad de Medicina, Departamento de Bioquímica, Madri, Espanha
| | - Carlos Simón
- Hospital General Universitario Gregorio Marañon, Departamento de Cirugía Torácica, Madri, Espanha; Universidad Complutense de Madrid, Departamento de Cirugía, Madri, Espanha
| | - Elena Vara
- Universidad Complutense de Madrid, Facultad de Medicina, Departamento de Bioquímica, Madri, Espanha
| |
Collapse
|
31
|
Pao HP, Liao WI, Wu SY, Hung KY, Huang KL, Chu SJ. PG490-88, a derivative of triptolide, suppresses ischemia/reperfusion-induced lung damage by maintaining tight junction barriers and targeting multiple signaling pathways. Int Immunopharmacol 2018; 68:17-29. [PMID: 30599444 DOI: 10.1016/j.intimp.2018.12.058] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/08/2018] [Accepted: 12/25/2018] [Indexed: 12/27/2022]
Abstract
Previous studies demonstrated that triptolide (PG490) has many anti-inflammatory and immunosuppressive effects. However, little is known about the effect of PG490-88 (a water-soluble derivative of triptolide) on ischemia/reperfusion (I/R)-induced acute lung injury. We assessed the effects of PG490-88 on I/R-induced acute lung injury in rats and on hypoxia/reoxygenation (H/R) in a line of murine epithelial cells. Isolated perfused rat lungs were subjected to 40 min of ischemia, followed by 60 min of reperfusion to induce I/R injury. Induction of I/R led to lung edema, elevated pulmonary arterial pressure, histological evidence of lung inflammation, oxidative stress, and increased levels of TNF-α and CINC-1 in bronchoalveolar lavage fluid. PG490-88 significantly suppressed all of these responses. Additionally, induction of I/R reduced the expression of claudin-4, occludin, and ZO-1, and increased apoptosis in lung tissue. PG490-88 also significantly suppressed these effects. I/R reduced the levels of IκB-α and MKP-1, and increased the levels of nuclear NF-κB and mitogen-activated protein kinase in lung tissue, and PG490-88 suppressed these effects. In vitro studies using mouse lung alveolar epithelial cells indicated that H/R increased the levels of phosphorylated p65 and MIP-2, but decreased the level of IκB-α. PG490-88 also suppressed these effects. In I/R damaged lungs, PG490-88 suppresses the inflammatory response, disruption of tight junction structure, and apoptosis. PG490-88 has the potential as a prophylactic agent to prevent I/R-induced lung injury.
Collapse
Affiliation(s)
- Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan; Department of Emergency Medicine, Tri-Service General Hospital, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kuei-Yi Hung
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan.
| | - Shi-Jye Chu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
32
|
Garutti I, Gonzalez-Moraga F, Sanchez-Pedrosa G, Casanova J, Martin-Piñeiro B, Rancan L, Simón C, Vara E. [The effect of anesthetic preconditioning with sevoflurane on intracellular signal-transduction pathways and apoptosis, in a lung autotransplant experimental model]. Rev Bras Anestesiol 2018; 69:48-57. [PMID: 30459087 DOI: 10.1016/j.bjan.2018.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 06/29/2018] [Accepted: 07/13/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Anesthetic pre-conditioning attenuates inflammatory response during ischemia-reperfusion lung injury. The molecular mechanisms to explain it are not fully understood. The aim of our investigation was to analyze the molecular mechanism that explain the anti-inflammatory effects of anesthetic pre-conditioning with sevoflurane focusing on its effects on MAPKs (mitogen-activated protein kinases), NF-κB (nuclear factor kappa beta) pathways, and apoptosis in an experimental lung autotransplant model. METHODS Twenty large white pigs undergoing pneumonectomy plus lung autotransplant were divided into two 10-member groups on the basis of the anesthetic received (propofol or sevoflurane). Anesthetic pre-conditioning group received sevoflurane 3% after anesthesia induction and it stopped when one-lung ventilation get started. Control group did not receive sevoflurane in any moment during the whole study period. Intracellular signal-transduction pathways (MAPK family), transcription factor (NF-κB), and apoptosis (caspases 3 and 9) were analyzed during experiment. RESULTS Pigs that received anesthetic pre-conditioning with sevoflurane have shown significant lower values of MAPK-p38, MAPK-P-p38, JNK (c-Jun N-terminal kinases), NF-κB p50 intranuclear, and caspases (p<0.05) than pigs anesthetized with intravenous propofol. CONCLUSIONS Lung protection of anesthetic pre-conditioning with sevoflurane during experimental lung autotransplant is, at least, partially associated with MAPKs and NF κB pathways attenuation, and antiapoptotic effects.
Collapse
Affiliation(s)
- Ignacio Garutti
- Hospital General Universitario Gregorio Marañon, Departamento de Anestesiologia, Madri, Espanha; Universidad Complutense de Madrid, Departamento de Farmacologia, Madri, Espanha.
| | | | | | - Javier Casanova
- Hospital General Universitario Gregorio Marañon, Departamento de Anestesiologia, Madri, Espanha
| | | | - Lisa Rancan
- Universidad Complutense de Madrid, Facultad de Medicina, Departamento de Bioquímica, Madri, Espanha
| | - Carlos Simón
- Hospital General Universitario Gregorio Marañon, Departamento de Cirugía Torácica, Madri, Espanha; Universidad Complutense de Madrid, Departamento de Cirugía, Madri, Espanha
| | - Elena Vara
- Universidad Complutense de Madrid, Facultad de Medicina, Departamento de Bioquímica, Madri, Espanha
| |
Collapse
|
33
|
Li X, Xu Y, Cheng Y, Wang R. α7 Nicotinic acetylcholine receptor contributes to the alleviation of lung ischemia-reperfusion injury by transient receptor potential vanilloid type 1 stimulation. J Surg Res 2018; 230:164-174. [PMID: 30100034 DOI: 10.1016/j.jss.2018.05.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/06/2018] [Accepted: 05/23/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Activation of transient receptor potential vanilloid type 1 (TRPV1) decreases lung ischemia-reperfusion injury (LIRI) in rabbits and rats. Stimulation of α7 nicotinic acetylcholine receptors (α7nAChRs) protects against lung injury. Here we examined whether α7nAChRs contribute to TRPV1-mediated protection against LIRI. METHODS Wild-type (WT) and TRPV1-knockout (KO) mice were subjected to 1-h lung ischemia by clamping left hilum, followed by 2-h reperfusion. WT or KO mice were pretreated with vehicle, TRPV1 agonist capsaicin, TRPV1 antagonist capsazepine, α7nAChR antagonist methyllycaconitine, or α7nAChR agonist PNU-282987. Arterial blood and lung tissues were obtained for blood gas, lung wet-to-dry weight ratio, interleukin (IL)1β, IL6, tumor necrosis factor-α (TNF-α), apoptosis-related proteins (caspases, Bax, Fas), and pathologic scoring. RESULTS Capsaicin pretreatment reduced wet-to-dry ratio, pathologic score, alveolar-arterial oxygen gradient (A-aDO2), and IL1β, IL6, and TNFα levels in WT mice, with no effects in KO mice. This reduction was reversed by TRPV1 blockade. Furthermore, α7nAChR blockade before capsaicin exacerbated LIRI as evidenced by enhanced alveolar-arterial oxygen gradient, pathologic score, and IL1β, IL6, and TNFα levels, while α7nAChR agonist pretreatment under TRPV1 blockade showed opposite changes. Capsaicin also decreased cleaved caspase-3, caspase-3/9, and Bax protein expression, effects abolished by TRPV1 blockade. Similarly, α7nAChR blockade diminished capsaicin-induced downregulation of apoptotic proteins, and α7nAChR activation decreased expression levels even under TRPV1 blockade. CONCLUSIONS TRPV1 activation alleviates LIRI, partially dependent on α7nAChR activity. The α7nAChR stimulation with or without existence of TRPV1 alleviates LIRI. Thus, α7nAChR is involved in the pathway of TRPV1-mediated protection against LIRI and the specific mechanism remains to be revealed.
Collapse
Affiliation(s)
- Xuehan Li
- Department of Anesthesiology, and Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yi Xu
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Cheng
- Department of Anesthesiology, and Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Rurong Wang
- Department of Anesthesiology, and Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
34
|
Pak O, Sydykov A, Kosanovic D, Schermuly RT, Dietrich A, Schröder K, Brandes RP, Gudermann T, Sommer N, Weissmann N. Lung Ischaemia-Reperfusion Injury: The Role of Reactive Oxygen Species. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:195-225. [PMID: 29047088 DOI: 10.1007/978-3-319-63245-2_12] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Lung ischaemia-reperfusion injury (LIRI) occurs in many lung diseases and during surgical procedures such as lung transplantation. The re-establishment of blood flow and oxygen delivery into the previously ischaemic lung exacerbates the ischaemic injury and leads to increased microvascular permeability and pulmonary vascular resistance as well as to vigorous activation of the immune response. These events initiate the irreversible damage of the lung with subsequent oedema formation that can result in systemic hypoxaemia and multi-organ failure. Alterations in the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS) have been suggested as crucial mediators of such responses during ischaemia-reperfusion in the lung. Among numerous potential sources of ROS/RNS within cells, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, xanthine oxidases, nitric oxide synthases and mitochondria have been investigated during LIRI. Against this background, we aim to review here the extensive literature about the ROS-mediated cellular signalling during LIRI, as well as the effectiveness of antioxidants as treatment option for LIRI.
Collapse
Affiliation(s)
- Oleg Pak
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Akylbek Sydykov
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Djuro Kosanovic
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Alexander Dietrich
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Goethestraße 33, 80336, Munich, Germany
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Thomas Gudermann
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Goethestraße 33, 80336, Munich, Germany
| | - Natascha Sommer
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany.
| |
Collapse
|
35
|
Wan L, Zhang D, Zhang J, Ren L. TT-1, an analog of melittin, triggers apoptosis in human thyroid cancer TT cells via regulating caspase, Bcl-2 and Bax. Oncol Lett 2018; 15:1271-1278. [PMID: 29387245 DOI: 10.3892/ol.2017.7366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 06/22/2017] [Indexed: 01/25/2023] Open
Abstract
Melittin is a 26 amino acid residue antimicrobial peptide with known antitumor activity. In the present study, a novel peptide TT-1, derived from melittin and contained only 11 amino acids, was designed, and its antitumor effect was investigated. The present study is aimed to elucidate the effects and relative mechanisms of TT-1 on a human thyroid cancer cell line (TT) in vitro and in vivo. Cell viability assays, Annexin V/propidium iodide assays, western blotting and quantitative reverse transcription polymerase chain reaction were performed. Furthermore, a tumor-xenograft model was established to investigate the apoptotic mechanisms of TT-1 on TT cells. The results obtained indicated that TT-1 was able to suppress the proliferation of TT cells and exhibited low cytotoxicity to normal thyroid cells in vitro. The apoptotic rates of TT cells were also increased following TT-1 treatment. Additionally, TT-1 stimulated caspase-3, caspase-9 and Bax, and inhibited B-cell lymphoma 2 mRNA and protein expression. Finally, it was also demonstrated that TT-1 is able to markedly suppress tumor growth in a TT-bearing nude mouse model. In summary, TT-1 may inhibit the proliferation of TT cells by inducing apoptosis in vitro and in vivo, indicating that TT-1 may be a potential candidate for the treatment of thyroid cancer.
Collapse
Affiliation(s)
- Lanlan Wan
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, Jilin 130033, P.R. China.,Department of Pharmacology and Toxicology, Jilin University School of Pharmaceutical Sciences, Changchun, Jilin 130021, P.R. China
| | - Daqi Zhang
- Department of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Jilin Provincial Key Laboratory of Surgical Translational Medicine, Changchun, Jilin 130033, P.R. China
| | - Jinnan Zhang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Liqun Ren
- Department of Pharmacology and Toxicology, Jilin University School of Pharmaceutical Sciences, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
36
|
Targeting Circulating Leukocytes and Pyroptosis During Ex Vivo Lung Perfusion Improves Lung Preservation. Transplantation 2017; 101:2841-2849. [PMID: 28452921 DOI: 10.1097/tp.0000000000001798] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
37
|
Brunner SN, Bogert NV, Schnitzbauer AA, Juengel E, Moritz A, Werner I, Kornberger A, Beiras-Fernandez A. Levosimendan protects human hepatocytes from ischemia-reperfusion injury. PLoS One 2017; 12:e0187839. [PMID: 29145424 PMCID: PMC5690693 DOI: 10.1371/journal.pone.0187839] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 10/26/2017] [Indexed: 01/28/2023] Open
Abstract
Background Ischemia-reperfusion injury (IRI) is a major challenge in liver transplantation. The mitochondrial pathway plays a pivotal role in hepatic IRI. Levosimendan, a calcium channel sensitizer, was shown to attenuate apoptosis after IRI in animal livers. The aim of this study was to investigate the effect of levosimendan on apoptosis in human hepatocytes. Methods Primary human hepatocytes were either exposed to hypoxia or cultured under normoxic conditions. After the hypoxic phase, reoxygenation was implemented and cells were treated with different concentrations of levosimendan (10ng/ml, 100ng/ml, 1000ng/ml). The overall metabolic activity of the cells was measured using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and aspartate aminotransferase (AST) levels were determined in order to quantify hepatic injury. Fluorescence-activated cell sorting (FACS) analysis was applied to measure necrosis and apoptosis. Finally, Western blotting was performed to analyze apoptotic pathway proteins. Results Administration of levosimendan during reperfusion increases the metabolic activity of human hepatocytes and decreases AST levels. Moreover, apoptosis after IRI is reduced in treated vs. untreated hepatocytes, and levosimendan prevents down-regulation of the anti-apoptotic protein Bcl-2 as well as up-regulation of the pro-apoptotic protein BAX. Conclusion The present study suggests a protective effect of levosimendan on human hepatocytes. Our findings suggest that treatment with levosimendan during reperfusion attenuates apoptosis of human hepatocytes by influencing BAX and Bcl-2 levels.
Collapse
Affiliation(s)
- Stefanie N. Brunner
- Department of Thoracic and Cardiovascular Surgery, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany
| | - Nicolai V. Bogert
- Department of Cardiology, University Hospital Heidelberg, Ruprecht-Karls-University, Heidelberg, Germany
| | - Andreas A. Schnitzbauer
- Clinic for General and Visceral Surgery, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany
| | - Eva Juengel
- Department of Urology, University Hospital Mainz, Johannes Gutenberg University, Mainz, Germany
| | - Anton Moritz
- Department of Thoracic and Cardiovascular Surgery, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany
| | - Isabella Werner
- Department of Thoracic and Cardiovascular Surgery, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany
| | - Angela Kornberger
- Department of Thoracic and Cardiovascular Surgery, University Hospital Mainz, Johannes Gutenberg University, Mainz, Germany
- * E-mail:
| | - Andres Beiras-Fernandez
- Department of Thoracic and Cardiovascular Surgery, University Hospital Mainz, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
38
|
Gao S, Zhang Z, Brunelli A, Chen C, Chen C, Chen G, Chen H, Chen JS, Cassivi S, Chai Y, Downs JB, Fang W, Fu X, Garutti MI, He J, He J, Hu J, Huang Y, Jiang G, Jiang H, Jiang Z, Li D, Li G, Li H, Li Q, Li X, Li Y, Li Z, Liu CC, Liu D, Liu L, Liu Y, Ma H, Mao W, Mao Y, Mou J, Ng CSH, Petersen RH, Qiao G, Rocco G, Ruffini E, Tan L, Tan Q, Tong T, Wang H, Wang Q, Wang R, Wang S, Xie D, Xue Q, Xue T, Xu L, Xu S, Xu S, Yan T, Yu F, Yu Z, Zhang C, Zhang L, Zhang T, Zhang X, Zhao X, Zhao X, Zhi X, Zhou Q. The Society for Translational Medicine: clinical practice guidelines for mechanical ventilation management for patients undergoing lobectomy. J Thorac Dis 2017; 9:3246-3254. [PMID: 29221302 PMCID: PMC5708473 DOI: 10.21037/jtd.2017.08.166] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Patients undergoing lobectomy are at significantly increased risk of lung injury. One-lung ventilation is the most commonly used technique to maintain ventilation and oxygenation during the operation. It is a challenge to choose an appropriate mechanical ventilation strategy to minimize the lung injury and other adverse clinical outcomes. In order to understand the available evidence, a systematic review was conducted including the following topics: (I) protective ventilation (PV); (II) mode of mechanical ventilation [e.g., volume controlled (VCV) versus pressure controlled (PCV)]; (III) use of therapeutic hypercapnia; (IV) use of alveolar recruitment (open-lung) strategy; (V) pre-and post-operative application of positive end expiratory pressure (PEEP); (VI) Inspired Oxygen concentration; (VII) Non-intubated thoracoscopic lobectomy; and (VIII) adjuvant pharmacologic options. The recommendations of class II are non-intubated thoracoscopic lobectomy may be an alternative to conventional one-lung ventilation in selected patients. The recommendations of class IIa are: (I) Therapeutic hypercapnia to maintain a partial pressure of carbon dioxide at 50-70 mmHg is reasonable for patients undergoing pulmonary lobectomy with one-lung ventilation; (II) PV with a tidal volume of 6 mL/kg and PEEP of 5 cmH2O are reasonable methods, based on current evidence; (III) alveolar recruitment [open lung ventilation (OLV)] may be beneficial in patients undergoing lobectomy with one-lung ventilation; (IV) PCV is recommended over VCV for patients undergoing lung resection; (V) pre- and post-operative CPAP can improve short-term oxygenation in patients undergoing lobectomy with one-lung ventilation; (VI) controlled mechanical ventilation with I:E ratio of 1:1 is reasonable in patients undergoing one-lung ventilation; (VII) use of lowest inspired oxygen concentration to maintain satisfactory arterial oxygen saturation is reasonable based on physiologic principles; (VIII) Adjuvant drugs such as nebulized budesonide, intravenous sivelestat and ulinastatin are reasonable and can be used to attenuate inflammatory response.
Collapse
Affiliation(s)
- Shugeng Gao
- Department of Thoracic Surgical Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center, Beijing 100021, China
| | - Zhongheng Zhang
- Department of Emergency Medicine, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | | | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Shanghai 200433, China
| | - Chun Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fujian 350001, China
| | - Gang Chen
- Department of Thoracic Surgery, Guangdong General Hospital, Guangzhou 510080, China
| | | | - Jin-Shing Chen
- Department of Anesthesiology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 10002, Taiwan
| | | | - Ying Chai
- Second Affiliated Hospital, Medical College of Zhejiang University, Hangzhou 310009, China
| | - John B. Downs
- Department of Anesthesiology and Critical Care Medicine, University of Florida, Gainesville, FL, USA
| | - Wentao Fang
- Shanghai Chest Hospital, Shanghai 200030, China
| | - Xiangning Fu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Martínez I. Garutti
- Department of Anaesthesia and Postoperative Care, Hospital General Universitario Gregorio Marañon, Madrid, Spain
| | - Jianxing He
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510000, China
- Guangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou 510000, China
| | - Jie He
- Department of Thoracic Surgical Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center, Beijing 100021, China
| | - Jian Hu
- First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou 310003, China
| | - Yunchao Huang
- Department of Thoracic Surgery, Yunnan Cancer Hospital, Kunming 650100, China
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Shanghai 200433, China
| | - Hongjing Jiang
- Department of Esophageal Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Zhongmin Jiang
- Department of Thoracic Surgery, Shandong Qianfoshan Hospital, Jinan 250014, China
| | - Danqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Beijing 100032, China
| | - Gaofeng Li
- Department of Thoracic Surgery, Yunnan Cancer Hospital, Kunming 650100, China
| | - Hui Li
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Beijing 100049, China
| | - Qiang Li
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institute, Chengdu 610041, China
| | - Xiaofei Li
- Department of Thoracic Surgery, Tangdu Hospital Fourth Military Medical University, Xi’an 710038, China
| | - Yin Li
- Department of Thoracic Surgery, Henan Cancer Hospital, Zhengzhou 450008, China
| | - Zhijun Li
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Chia-Chuan Liu
- Division of Thoracic Surgery, Department of Surgery, Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Deruo Liu
- Department of Thoracic Surgery, China and Japan Friendship Hospital, Beijing 100029, China
| | - Lunxu Liu
- Department of Cardiovascular and Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yongyi Liu
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute, Shengyang 110042, China
| | - Haitao Ma
- Department of Thoracic Surgery, The First Hospital Affiliated to Soochow University, Suzhou 215000, China
| | - Weimin Mao
- Department of Thoracic Surgery, Zhejiang Cancer Hospital, Hangzhou 310000, China
| | - Yousheng Mao
- Department of Thoracic Surgical Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center, Beijing 100021, China
| | - Juwei Mou
- Department of Thoracic Surgical Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center, Beijing 100021, China
| | - Calvin Sze Hang Ng
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T., Hong Kong, China
| | - René H. Petersen
- Department of Cardiothoracic Surgery, Rigshospitalet, Copenhagen, Denmark
| | - Guibin Qiao
- Department of Thoracic Surgery, Guangzhou General Hospital of Guangzhou Military Area Command, Guangzhou 510000, China
| | - Gaetano Rocco
- Department of Thoracic Surgery and Oncology, National Cancer Institute, Pascale Foundation, Naples, Italy
| | - Erico Ruffini
- Thoracic Surgery Unit, University of Torino, Torino, Italy
| | - Lijie Tan
- Department of Thoracic Surgery, Shanghai Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Qunyou Tan
- Department of Thoracic Surgery, Daping Hospital, Research Institute of Surgery Third Military Medical University, Chongqing 400042, China
| | - Tang Tong
- Department of Thoracic Surgery, Second Affiliated Hospital of Jilin University, Changchun 130041, China
| | - Haidong Wang
- Department of Thoracic Surgery, Southwest Hospital, Third Millitary Medical University, Chongqing 400038, China
| | - Qun Wang
- Department of Thoracic Surgery, Shanghai Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Ruwen Wang
- Department of Thoracic Surgery, Daping Hospital, Research Institute of Surgery Third Military Medical University, Chongqing 400042, China
| | - Shumin Wang
- Department of Thoracic Surgery, General Hospital of Shenyang Military Area, Shenyang 110015, China
| | - Deyao Xie
- Department of Cardiovascular and Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Qi Xue
- Department of Thoracic Surgical Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Cancer Center, Beijing 100021, China
| | - Tao Xue
- Department of Thoracic Surgery, Zhongda Hospital Southeast University, Nanjing 210009, China
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Nanjing 210008, China
| | - Shidong Xu
- Department of Thoracic Surgery, Heilongjiang Cancer Hospital, Harbin 150049, China
| | - Songtao Xu
- Department of Thoracic Surgery, Shanghai Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Tiansheng Yan
- Department of Thoracic Surgery, Peking University Third Hospital, Beijing 100083, China
| | - Fenglei Yu
- Department of Cardiovascular Surgery, Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhentao Yu
- Department of Esophageal Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lanjun Zhang
- Cancer Center, San Yat-sen University, Guangzhou 510060, China
| | - Tao Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Xun Zhang
- Department of Thoracic Surgery, Tanjin Chest Hospital, Tianjin 300300, China
| | - Xiaojing Zhao
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200000, China
| | - Xuewei Zhao
- Department of Thoracic Surgery, Shanghai Changzheng Hospital, Shanghai 200000, China
| | - Xiuyi Zhi
- Department of Thoracic Surgery, Xuanwu Hospital of Capital University of Medical Sciences, Beijing 100053, China
| | - Qinghua Zhou
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute, Shengyang 110042, China
| |
Collapse
|
39
|
Cippà PE, Fehr T. Pharmacological modulation of cell death in organ transplantation. Transpl Int 2017; 30:851-859. [PMID: 28480540 DOI: 10.1111/tri.12977] [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: 01/03/2017] [Revised: 02/20/2017] [Accepted: 04/29/2017] [Indexed: 12/22/2022]
Abstract
New options to pharmacologically modulate fundamental mechanisms of regulated cell death are rapidly evolving and found first clinical applications in cancer therapy. Here, we present an overview on how the recent advances in the understanding of the biology and pharmacology of cell death might influence research and clinical practice in solid organ transplantation. Of particular interest are the novel opportunities related to organ preservation and immunomodulation, which might contribute to promote organ repair and to develop more selective ways to modulate allogeneic immune responses to prevent rejection and induce immunological tolerance.
Collapse
Affiliation(s)
- Pietro E Cippà
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Fehr
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland.,Department of Internal Medicine, Cantonal Hospital Graubuenden, Chur, Switzerland
| |
Collapse
|
40
|
Intratracheal Administration of Small Interfering RNA Targeting Fas Reduces Lung Ischemia-Reperfusion Injury. Crit Care Med 2017; 44:e604-13. [PMID: 26963318 DOI: 10.1097/ccm.0000000000001601] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Lung ischemia-reperfusion injury is the main cause of primary graft dysfunction after lung transplantation and results in increased morbidity and mortality. Fas-mediated apoptosis is one of the pathologic mechanisms involved in the development of ischemia-reperfusion injury. We hypothesized that the inhibition of Fas gene expression in lungs by intratracheal administration of small interfering RNA could reduce lung ischemia-reperfusion injury in an ex vivo model reproducing the procedural sequence of lung transplantation. DESIGN Prospective, randomized, controlled experimental study. SETTING University research laboratory. SUBJECTS C57/BL6 mice weighing 28-30 g. INTERVENTIONS Ischemia-reperfusion injury was induced in lungs isolated from mice, 48 hours after treatment with intratracheal small interfering RNA targeting Fas, control small interfering RNA, or vehicle. Isolated lungs were exposed to 6 hours of cold ischemia (4°C), followed by 2 hours of warm (37°C) reperfusion with a solution containing 10% of fresh whole blood and mechanical ventilation with constant low driving pressure. MEASUREMENTS AND MAIN RESULTS Fas gene expression was significantly silenced at the level of messenger RNA and protein after ischemia-reperfusion in lungs treated with small interfering RNA targeting Fas compared with lungs treated with control small interfering RNA or vehicle. Silencing of Fas gene expression resulted in reduced edema formation (bronchoalveolar lavage protein concentration and lung histology) and improvement in lung compliance. These effects were associated with a significant reduction of pulmonary cell apoptosis of lungs treated with small interfering RNA targeting Fas, which did not affect cytokine release and neutrophil infiltration. CONCLUSIONS Fas expression silencing in the lung by small interfering RNA is effective against ischemia-reperfusion injury. This approach represents a potential innovative strategy of organ preservation before lung transplantation.
Collapse
|
41
|
Zhang D, Li C, Song Y, Zhou J, Li Y, Li J, Bai C. Integrin αvβ5 inhibition protects against ischemia-reperfusion-induced lung injury in an autophagy-dependent manner. Am J Physiol Lung Cell Mol Physiol 2017; 313:L384-L394. [PMID: 28522565 DOI: 10.1152/ajplung.00391.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 04/03/2017] [Accepted: 05/16/2017] [Indexed: 11/22/2022] Open
Abstract
Integrin αvβ5 mediates pulmonary endothelial barrier function and acute lung injury (LI), but its roles in cell apoptosis and autophagy are unclear. Thus, the aims of this study were to investigate the significance of αvβ5 in ischemia-reperfusion (I/R)-induced apoptosis and LI and to explore the relationship between αvβ5 and autophagy. Human pulmonary microvascular endothelial cells (HPMVECs) were pretreated with an αvβ5-blocking antibody (ALULA) and challenged with oxygen-glucose deprivation/oxygen-glucose restoration, which mimics I/R; then, cellular autophagy and apoptosis were detected, and cell permeability was assessed. In vivo, mice were pretreated with the autophagy inhibitor chloroquine (CLQ), followed by treatment with ALULA. The mice then underwent operative lung I/R. LI was assessed by performing a pathological examination, calculating the wet/dry lung weight ratio and detecting the bronchial alveolar lavage fluid (BALF) protein concentration. αvβ5 inhibition promoted HPMVEC autophagy under I/R in vitro, alleviated cell permeability, decreased the apoptosis ratio, and activated caspase-3 expression. These outcomes were significantly diminished when autophagy was inhibited with a small-interfering RNA construct targeting autophagy-related gene 7 (siATG7). Moreover, ALULA pretreatment alleviated I/R-induced LI (I/R-LI), which manifested as a decreased wet/dry lung weight ratio, an altered BALF protein concentration, and lung edema. Preinhibiting autophagy with CLQ, however, eliminated the protective effects of ALULA on I/R-LI. Therefore, inhibiting αvβ5 effectively ameliorated I/R-induced endothelial cell apoptosis and I/R-LI. This process was dependent on improved autophagy and its inhibitory effects on activated caspase-3.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Wenzhou City, Zhejiang Province, China
| | - Chichi Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Wenzhou City, Zhejiang Province, China
| | - Yuanlin Song
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Respiratory Research Institute, Shanghai, China; and
| | - Jian Zhou
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Respiratory Research Institute, Shanghai, China; and
| | - Yuping Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Wenzhou City, Zhejiang Province, China
| | - Jing Li
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Respiratory Research Institute, Shanghai, China; and
| | - Chunxue Bai
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; .,Shanghai Respiratory Research Institute, Shanghai, China; and.,State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
42
|
|
43
|
Yen YT, Roan JN, Fang SY, Chang SW, Tseng YL, Lam CF. Autologous endothelial progenitor cells improve allograft survival in porcine lung transplantation with prolonged ischemia. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:277. [PMID: 27570771 DOI: 10.21037/atm.2016.06.22] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND As endothelial progenitor cells (EPCs) attenuated acute lung injury (ALI) in rabbit model, we hypothesized that autologous EPCs preserved lung graft function during the acute reperfusion period of lung transplantation and tested the therapeutic potential of EPCs in a porcine model of lung transplantation with prolonged graft ischemia. METHODS Day-7 EPCs isolated from the recipient subjects or plain culture media were administered into the left pulmonary artery immediately before restoration of pulmonary blood flow in a porcine lung allotransplantation model, with the transplantation surgeons blinded to the content of injection. Hemodynamics and arterial blood gas were recorded, and the right pulmonary artery was occluded 30 min after reperfusion to evaluate the lung graft function. The lung grafts were sectioned for histological examination at the end of experiments. The total ischemic time for lung graft was approximately 14 h. RESULTS All animals receiving plain medium died within 40 min after reperfusion, but 3 out of 5 (60%) piglets receiving EPCs survived up to 4 h after diversion of the entire cardiac output into the lung graft (P<0.01). The donor body weight, recipient body weight, cold ischemic time, and time for anastomosis were comparable between the EPC and control group (P=0.989, 0.822, 0.843, and 0.452, respectively). The mean aortic pressure decreased, and the cardiac output and mean pulmonary artery pressure elevated after right pulmonary artery occlusion. All these parameters were gradually compensated in the EPC group but decompensated in the control group. Better preservation of gas exchange function, reduced thrombi formation in the terminal pulmonary arterioles, and attenuated interstitial hemorrhage of the lung graft were observed in the EPC group. CONCLUSIONS We concluded autologous EPCs significantly enhanced the function of lung allograft and improved survival in a porcine model of lung transplantation with prolonged ischemia.
Collapse
Affiliation(s)
- Yi-Ting Yen
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan;; Institube of Clinical Medicine, College of Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Jun-Neng Roan
- Institube of Clinical Medicine, College of Medical College, National Cheng Kung University, Tainan, Taiwan;; Division of Cardiovascular Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Yuan Fang
- Department of Anesthesiology, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Shi-Wei Chang
- Department of Anesthesiology, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Yau-Lin Tseng
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Chen-Fuh Lam
- Department of Anesthesiology, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan;; Department of Anesthesiology, Buddhist Tzu Chi General Hospital and Tzu Chi University School of Medicine, Hualien, Taiwan
| |
Collapse
|
44
|
The Modulatory Effect of Ischemia and Reperfusion on Arginine Vasopressin-Induced Arterial Reactions. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3679048. [PMID: 27563664 PMCID: PMC4987452 DOI: 10.1155/2016/3679048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/04/2016] [Accepted: 07/10/2016] [Indexed: 11/17/2022]
Abstract
Aim of the Study. The purpose of this study was to investigate the impact of ischemia and reperfusion on the resistance of arteries to AVP (arginine vasopressin), with a particular emphasis on the role of smooth muscle cells in the action of vasopressin receptors and the role of the cGMP-associated signalling pathway. Materials and Methods. Experiment was performed on the perfunded tail arteries from male Wistar rats. The constriction triggered by AVP after 30 minutes of ischemia and 30 and 90 minutes of reperfusion was analysed. Analogous experiments were also carried out in the presence of 8Br-cGMP. Results. Ischemia reduces and reperfusion increases in a time-dependent manner the arterial reaction to AVP. The presence of 8Br-cGMP causes a significant decrease of arterial reactivity under study conditions. Conclusions. Ischemia and reperfusion modulate arterial contraction triggered by AVP. The effect of 8Br-cGMP on reactions, induced by AVP after ischemia and reperfusion, indicates that signalling pathway associated with nitric oxide (NO) and cGMP regulates the tension of the vascular smooth muscle cells.
Collapse
|
45
|
|
46
|
Ranji M, Motlagh MM, Salehpour F, Sepehr R, Heisner JS, Dash RK, Camara AKS. Optical Cryoimaging Reveals a Heterogeneous Distribution of Mitochondrial Redox State in ex vivo Guinea Pig Hearts and Its Alteration During Ischemia and Reperfusion. IEEE JOURNAL OF TRANSLATIONAL ENGINEERING IN HEALTH AND MEDICINE-JTEHM 2016; 4:1800210. [PMID: 27574574 PMCID: PMC4993131 DOI: 10.1109/jtehm.2016.2570219] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/21/2016] [Accepted: 04/26/2016] [Indexed: 12/16/2022]
Abstract
Oxidation of substrates to generate ATP in mitochondria is mediated by redox reactions of NADH and FADH2. Cardiac ischemia and reperfusion (IR) injury compromises mitochondrial oxidative phosphorylation. We hypothesize that IR alters the metabolic heterogeneity of mitochondrial redox state of the heart that is only evident in the 3-D optical cryoimaging of the perfused heart before, during, and after IR. The study involved four groups of hearts: time control (TC: heart perfusion without IR), global ischemia (Isch), global ischemia followed by reperfusion (IR) and TC with PCP (a mitochondrial uncoupler) perfusion. Mitochondrial NADH and FAD autofluorescence signals were recorded spectrofluorometrically online in guinea pig ex vivo-perfused hearts in the Langendorff mode. At the end of each specified protocol, hearts were rapidly removed and snap frozen in liquid N2 for later 3-D optical cryoimaging of the mitochondrial NADH, FAD, and NADH/FAD redox ratio (RR). The TC hearts revealed a heterogeneous spatial distribution of NADH, FAD, and RR. Ischemia and IR altered the spatial distribution and caused an overall increase and decrease in the RR by 55% and 64%, respectively. Uncoupling with PCP resulted in the lowest level of the RR (73% oxidation) compared with TC. The 3-D optical cryoimaging of the heart provides novel insights into the heterogeneous distribution of mitochondrial NADH, FAD, RR, and metabolism from the base to the apex during ischemia and IR. This 3-D information of the mitochondrial redox state in the normal and ischemic heart was not apparent in the dynamic spectrofluorometric data.
Collapse
Affiliation(s)
- Mahsa Ranji
- Department of Electrical EngineeringUniversity of Wisconsin-MilwaukeeMilwaukeeWI53211USA; Biotechnology and Bioengineering CenterMedical College of WisconsinMilwaukeeWI53226USA
| | | | - Fahimeh Salehpour
- Department of Electrical Engineering University of Wisconsin-Milwaukee Milwaukee WI 53211 USA
| | - Reyhaneh Sepehr
- Department of Electrical Engineering University of Wisconsin-Milwaukee Milwaukee WI 53211 USA
| | - James S Heisner
- Department of Anesthesiology Medical College of Wisconsin Milwaukee WI 53226 USA
| | - Ranjan K Dash
- Department of PhysiologyMedical College of WisconsinMilwaukeeWI53226USA; Biotechnology and Bioengineering CenterMedical College of WisconsinMilwaukeeWI53226USA; Cardiovascular Research CenterMedical College of WisconsinMilwaukeeWI53226USA
| | - Amadou K S Camara
- Cardiovascular Research CenterMedical College of WisconsinMilwaukeeWI53226USA; Department of AnesthesiologyMedical College of WisconsinMilwaukeeWI53226USA
| |
Collapse
|
47
|
LI YUEBING, KANG YUQING, WONG KELVINKL, GHISTA DHANJOON, GU MIAONING. INVESTIGATING EXPRESSION OF AUTOPHAGY-ASSOCIATED PROTEINS LEVEL IN RATS WITH ACUTE LUNG INJURY INDUCED BY REMOTE LIMB ISCHEMIA-REPERFUSION. J MECH MED BIOL 2016. [DOI: 10.1142/s0219519416500196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Objective: To explore the early expression of autophagy-associated proteins in lung tissues in acute lung injury (ALI) induced by remote limb ischemia-reperfusion (LIR) by using rats as our test specimens. Method: A total of 48 adult male Sprague-Dawley (SD) rats with weights in the range of 220–250[Formula: see text]g were designated as LIR models, and divided randomly into two groups of 24 each: Sham group and ischemia-reperfusion (I/R) group. Then, each group was divided into four subgroups at the end of 0, 2, 4, 8[Formula: see text]h of reperfusion, after 3[Formula: see text]h of ischemia. The rats were anesthetized by pentobarbital sodium. The serum lactate dehydrogenases (LDH) were detected with enzyme linked immunosorbent assay (ELISA), and the pathological changes of lung tissues were observed by using immunofluorescence techniques. The expression of Beclin1 protein and Atg5 mRNA in the lung tissues were detected by using reverse transcription polymerase chain reaction (RT-PCR), and analyzed by 2[Formula: see text] method; Microtubules associated protein light chain 3 (LC3) in the lung tissues were detected by Western blot test. Result: The levels of serum LDH in I/R groups were much higher than those in Sham groups ([Formula: see text]), which showed that the rats models of LIR were successful. Immunofluorescence examination demonstrated injuries of lung tissues, thickening of alveolar septum and partial consolidation in I/R groups; however, this damage was not observed significantly in Sham groups. The expression of Beclin1 and Atg5 mRNA, LC3-II and the ratio of LC3-II/GAPDH in lung tissues were very much higher at 4 and 8[Formula: see text]h in IR groups ([Formula: see text] or [Formula: see text]), and were significantly higher at the same time compared with Sham groups ([Formula: see text] or [Formula: see text]). Conclusion: LIR causes ALI to induce increased autophagy and high expression of its relevant proteins; while continuous I/R can also cause autophagy inhibition.
Collapse
Affiliation(s)
- YUEBING LI
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, P. R. China
- Southern Hospital of Southern Medical University, Guangzhou 510515, P. R. China
| | - YUQING KANG
- Southern Hospital of Southern Medical University, Guangzhou 510515, P. R. China
| | - KELVIN KL WONG
- Engineering Computational Biology, School of Computer Science and Software Engineering, The University of Western Australia, 35 Stirling Highway, Crawley WA 6000, Australia
| | - DHANJOO N. GHISTA
- Education Committee, Southern Ozarks Alliance for Rural Development, Willow Springs, MO, USA
| | - MIAONING GU
- Southern Hospital of Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
48
|
Inflammatory response and pneumocyte apoptosis during lung ischemia-reperfusion injury in an experimental pulmonary thromboembolism model. J Thromb Thrombolysis 2016; 40:42-53. [PMID: 25677043 PMCID: PMC4445764 DOI: 10.1007/s11239-015-1182-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Lung ischemia-reperfusion injury (LIRI) may occur in the region of the affected lung after reperfusion therapy. The inflammatory response mechanisms related to LIRI in pulmonary thromboembolism (PTE), especially in chronic PTE, need to be studied further. In a PTE model, inflammatory response and apoptosis may occur during LIRI and nitric oxide (NO) inhalation may alleviate the inflammatory response and apoptosis of pneumocytes during LIRI. A PTE canine model was established through blood clot embolism to the right lower lobar pulmonary artery. Two weeks later, we performed embolectomy with reperfusion to examine the LIRI changes among different groups. In particular, the ratio of arterial oxygen partial pressure to fractional inspired oxygen (PaO2/FiO2), serum concentrations of tumor necrosis factor-α (TNF-α), myeloperoxidase concentrations in lung homogenates, alveolar polymorphonuclear neutrophils (PMNs), lobar lung wet to dry ratio (W/D ratio), apoptotic pneumocytes, and lung sample ultrastructure were assessed. The PaO2/FiO2 in the NO inhalation group increased significantly when compared with the reperfusion group 4 and 6 h after reperfusion (368.83 ± 55.29 vs. 287.90 ± 54.84 mmHg, P < 0.05 and 380.63 ± 56.83 vs. 292.83 ± 6 0.34 mmHg, P < 0.05, respectively). In the NO inhalation group, TNF-α concentrations and alveolar PMN infiltration were significantly decreased as compared with those of the reperfusion group, 6 h after reperfusion (7.28 ± 1.49 vs. 8.90 ± 1.43 pg/mL, P < 0.05 and [(19 ± 6)/10 high power field (HPF) vs. (31 ± 11)/10 HPF, P < 0.05, respectively]. The amount of apoptotic pneumocytes in the lower lobar lung was negatively correlated with the arterial blood PaO2/FiO2, presented a positive correlation trend with the W/D ratio of the lower lobar lung, and a positive correlation with alveolar PMN in the reperfusion group and NO inhalation group. NO provided at 20 ppm for 6 h significantly alleviated LIRI in the PTE model. Our data indicate that, during LIRI, an obvious inflammatory response and apoptosis occur in our PTE model and NO inhalation may be useful in treating LIRI by alleviating the inflammatory response and pneumocyte apoptosis. This potential application warrants further investigation.
Collapse
|
49
|
Huang Z, Li Y, Niu L, Xiao Y, Pu X, Zheng H, Qian M. Dynamic expressions of monocyte chemo attractant protein-1 and CC chamomile receptor 2 after balloon injury and their effects in intimal proliferation. Biomed Eng Online 2015; 14:55. [PMID: 26062549 PMCID: PMC4469411 DOI: 10.1186/s12938-015-0030-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/02/2015] [Indexed: 11/10/2022] Open
Abstract
Objective The dynamic expressions of monocyte chemo attractant protein-1 (MCP-1) and CC chamomile receptor 2 (CCR2) after balloon injury and their effects in intimal proliferation were discussed. In this study, the expression of MCP-1 and its receptor during the intimal proliferation in rat artery after balloon injury were studied. Methods Using the model of balloon injury of rats’ arteries, the changes of intimal proliferation were observed with optical microscopy and the expressions of MCP-1 and CCR2 at different times were examined with the methods of RT-PCR and immunohistochemistry. The expressions of MCP-1 and CCR2 in the arterial tissues were detected using reverse transcription polymerase chain reaction (RT-PCR) and analyzed by semi-quantitative method. Results The expressions of MCP-1 and CCR2 mRNA began to gradually increase after balloon injury. The MCP-1 reached to the peak on the first day, but decreased gradually later on. Expressions of CCR2 mRNA began to increase on the first day and reached to the peak on the 7th day, but then started to decrease gradually until 28th day when we can still detect it. The expressions of MCP-1 proteins began to increase gradually after balloon injury and were obviously detected in the VSMC on the 4th and 7th day, until 14th day when we can still detect it clearly in the proliferating intima. Conclusion The dynamic expressions of MCP-1, MCP-1 proteins and CCR2 mRNA after balloon injury were shown to play an important role in intimal proliferation.
Collapse
Affiliation(s)
- Zhigang Huang
- Emergency Department, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| | - Yuebing Li
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| | - Lili Niu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Shenzhen, 518055, China.
| | - Yang Xiao
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Shenzhen, 518055, China.
| | - Xiaodong Pu
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Shenzhen, 518055, China.
| | - Ming Qian
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Shenzhen, 518055, China.
| |
Collapse
|
50
|
Wu C, Geng X, Wan S, Hou H, Yu F, Jia B, Wang L. Cecropin-P17, an analog of Cecropin B, inhibits human hepatocellular carcinoma cell HepG-2 proliferation via regulation of ROS, Caspase, Bax, and Bcl-2. J Pept Sci 2015; 21:661-8. [PMID: 26010398 DOI: 10.1002/psc.2786] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 04/19/2015] [Accepted: 04/20/2015] [Indexed: 12/15/2022]
Abstract
Cecropin-P17 is a peptide derived from Cecropin B. In this study, we investigated the effects and relative mechanisms of Cecropin-P17 in a human liver cancer cell line (HepG-2) in vitro and in vivo. A cell viability assay, Annexin V/propidium iodide assay, western blot, flow cytometry, quantitative real-time polymerase chain reaction, and a tumor-xenograft model were applied to elucidate the mechanism exerted by Cecropin-P17 on HepG-2 cells. Cecropin-P17 significantly inhibited the proliferation of HepG-2 cells and demonstrated low cytotoxicity to normal liver cells in vitro. The apoptotic rate of HepG-2 cells was increased after Cecropin-P17 treatment together with increased production of reactive oxygen species. Moreover, Cecropin-P17 stimulated caspase-3, caspase-9, and Bax and inhibited Bcl-2 on both the transcriptional and translational levels. Finally, Cecropin-P17 significantly suppressed tumor growth in a HepG-2-bearing nude mouse model. All of these results indicated that Cecropin-P17 could be a potential agent for the treatment of liver cancer.
Collapse
Affiliation(s)
- Chunli Wu
- The General Department of the Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Xiaoping Geng
- The General Department of the Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Shengyun Wan
- The General Department of the Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Hui Hou
- The General Department of the Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Fanzong Yu
- The General Department of the Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Benli Jia
- The General Department of the Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Lei Wang
- The General Department of the Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| |
Collapse
|