1
|
Junior ERB, Wang A, Ribeiro RVP, Beroncal EL, Ramadan K, Michaelsen VS, Chen M, Ali A, Zhang Y, Pal P, Abdelnour E, Siebiger G, Pinto BM, Waddell T, Andreazza AC, Keshavjee S, Cypel M. The combination of postmortem sevoflurane ventilation and in situ topical cooling provides improved 6 hours lung preservation in an uncontrolled DCD porcine model. J Heart Lung Transplant 2025; 44:780-792. [PMID: 39368680 DOI: 10.1016/j.healun.2024.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Recent clinical series on donation after uncontrolled cardiovascular death (uDCD) reported successful transplantation of lungs preserved by pulmonary inflation up to 3 hours postmortem. This study aims to investigate the additive effects of in situ lowering of intrathoracic temperature and sevoflurane preconditioning on lung grafts in a porcine uDCD model. METHODS After uDCD induction, donor pigs were allocated to one of the following groups: control-static lung inflation only (SLI); TC - SLI + continuous intrapleural topical cooling (TC); or TC+Sevo - SLI + TC + sevoflurane. Lungs were retrieved 6 hours postasystole and evaluated via ex vivo lung perfusion (EVLP) for 6 hours. A left single lung transplant was performed using lungs from the best performing group, followed by 4 hours of graft evaluation. RESULTS Animals that received TC achieved intrathoracic temperature <15°C within 1 hour after chest filling of coolant. Only lungs from donors that received TC and TC+Sevo completed the planned postpreservation 6 hours EVLP assessment. Despite similar early performance of the 2 groups on EVLP, the TC+Sevo group was superior-associated with overall lower airway pressures, higher pulmonary compliances, less edema development, and less inflammation. Transplantation was performed using lungs from the TC+Sevo group, and excellent graft function was observed postreperfusion. CONCLUSIONS Preservation of uDCD lungs with a combination of static lung inflation, TC and sevoflurane treatment maintains good pulmonary function up to 6 hours postmortem with excellent early post lung transplant function. These interventions may significantly expand the clinical utilization of uDCD donor lungs.
Collapse
Affiliation(s)
- Edson Ricardo Brambate Junior
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Aizhou Wang
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Rafaela Vanin Pinto Ribeiro
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Erika L Beroncal
- Departments of Pharmacology & Toxicology and Psychiatry, Mitochondrial Innovation Initiative, University of Toronto, Toronto, Ontario, Canada
| | - Khaled Ramadan
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Vinicius Schenk Michaelsen
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Manyin Chen
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Aadil Ali
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Yu Zhang
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Prodipto Pal
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Etienne Abdelnour
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Gabriel Siebiger
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Bruno Maineri Pinto
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Tom Waddell
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Ana C Andreazza
- Departments of Pharmacology & Toxicology and Psychiatry, Mitochondrial Innovation Initiative, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Research Laboratories, Toronto Lung Transplant Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Division of Thoracic Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Park I, Min EK, Park JH, Oh AY, Ryu JH. The Effects of Volatile Anesthetics on Early Clinical Outcomes in Liver Transplantation: A Systematic Review and Meta-Analysis. Transplant Proc 2024; 56:1984-1994. [PMID: 39472225 DOI: 10.1016/j.transproceed.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 08/27/2024] [Accepted: 09/24/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND The aim of this systematic review and meta-analysis was to evaluate the effects of volatile anesthesia (VA) on early clinical outcomes in liver transplantation. METHODS We searched electronic databases to identify relevant studies comparing VA to non-VA in liver transplant recipients. The primary outcome assessed was early allograft dysfunction (EAD), and secondary outcomes were postoperative peak liver function tests (LFT) including aspartate transaminase (AST) and alanine transaminase (ALT) levels, and hospitalization time. RESULTS Six relevant studies involving 919 patients were analyzed. In meta-analysis of prospective studies, VA was associated with fewer incidence of EAD than non-VA (RR: 0.45; 95% CI: 0.25, 0.84; P = .012; I2 = 0%; Ph = 0.334) but this association was not significant in meta-analysis of retrospective studies (OR: 0.83; 95% CI: 0.58, 1.19; P = .310; I2 = 0%; Ph = 0.624). No significant difference in peak AST (SMD: -0.14 U/L; 95% CI: -0.65, 0.37 U/L; P = .594; I2 = 69.9%; Ph = 0.036) and ALT (SMD: -0.16 U/L; 95% CI: -0.65, 0.33 U/L; P = .529; I2 = 67.0%; Ph = 0.048) were found between VA and non-VA. The hospitalization time also did not differ between the two groups (SMD: -0.09 days; 95% CI: -0.29, 0.10 days; P = .350; I2 = 0%; Ph = 0.864). CONCLUSIONS While there is potential protective effect of VA against EAD in liver transplant recipients, certainty remains low, whereas VA was not associated with postoperative LFT or hospitalization time.
Collapse
Affiliation(s)
- Insun Park
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, Republic of Korea; Department of Anesthesiology and Pain Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun-Ki Min
- Department of General Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Hyon Park
- Department of Radiology, The Armed Forces Daejeon Hospital, Daejeon, Republic of Korea
| | - Ah-Young Oh
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, Republic of Korea; Department of Anesthesiology and Pain Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jung-Hee Ryu
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi, Republic of Korea; Department of Anesthesiology and Pain Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Oliveira-Melo P, Nepomuceno NA, Ruiz LM, Correia AT, Vilela VS, de Oliveira Braga KA, Manzuti GM, Feitosa DDM, Kennedy-Feitosa E, Wang A, Cypel M, Fernandes PMP. Angiotensin-converting enzyme 2 activation attenuates inflammation and oxidative stress in brain death donor followed by rat lung transplantation. Sci Rep 2024; 14:23567. [PMID: 39384890 PMCID: PMC11464679 DOI: 10.1038/s41598-024-75043-7] [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: 05/02/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024] Open
Abstract
Brain death (BD) provides most of the donor organs destined for lung transplantation (LTx). However, the organs may be affected by inflammatory and oxidative processes. Based on this, we hypothesize that the angiotensin-converting enzyme 2 (ACE2) activation can reduce the lung injury associated with LTx. 3 h after BD induction, rats were injected with saline (BD group) or an ACE2 activator (ACE2a group; 15 mg/kg-1) and kept on mechanical ventilation for additional 3 h. A third group included a control ventilation (Control group) prior to transplant. After BD protocol, left LTx were performed, followed by 2 h-reperfusion. ACE2 activation was associated with better oxygenation after BD management (p = 0.01), attenuating edema (p = 0.05) followed by the reduction in tissue resistance (p = 0.01) and increase of respiratory compliance (p = 0.02). Nrf2 expression was also upregulated in the ACE2a group (p = 0.03). After transplantation, ACE2a group showed lower levels of TNF-α (p = 0.02), IL-6 (p = 0.001), IL-1β (p = 0.01), ROS (p = 0.004) and MDA (p = 0.002), in addition to higher CAT activity (p = 0.04). In conclusion, our study suggests that ACE2 activation improves anti-inflammatory and antioxidant activity in a model of LTx.
Collapse
Affiliation(s)
- Paolo Oliveira-Melo
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| | - Natalia Aparecida Nepomuceno
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Liliane Moreira Ruiz
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Aristides Tadeu Correia
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Vanessa Sana Vilela
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Karina Andrighetti de Oliveira Braga
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Giovana Maria Manzuti
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Emanuel Kennedy-Feitosa
- Departamento de Ciências da Saúde, Laboratório de Morfofisiofarmacologia, Universidade Federal Rural do Semi-Árido, Mossoró, RN, Brazil
| | - Aizhou Wang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Marcelo Cypel
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Paulo Manuel Pêgo Fernandes
- Departamento de Cardiopneumologia, Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Instituto do Coração, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
4
|
Ding X, Gao X, Ren A, Xu J, Jiang X, Liang X, Xie K, Zhou Y, Hu C, Huang D. Sevoflurane enhances autophagy via Rac1 to attenuate lung ischaemia‒reperfusion injury. Chem Biol Interact 2024; 397:111078. [PMID: 38815668 DOI: 10.1016/j.cbi.2024.111078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Sevoflurane can attenuate lung ischaemia‒reperfusion injury (LIRI). However, the protective mechanism is unclear. In this study, we developed a LIRI model in vivo that animals (SD, n = 15) were subjected to the administration of 2.2 % sevoflurane 30 min before the onset of left pulmonary artery clamping for 45 min, which was then followed by 60 min of reperfusion treatment. Then, transcriptome sequencing was used to analyse lung tissues. Autophagy inhibition (3-MA) and Rac1-overexpression transfection plasmids were used in BEAS-2B cells, and BEAS-2B cells were subjected to hypoxia reoxygenation (H/R) and sevoflurane treatment. In both animal tissue and cells, inflammatory cytokines and apoptotic and autophagy molecules were measured by quantitative real-time PCR, western blotting and immunostaining. As a result, decreased arterial partial oxygen and damage to the histological structure of lung tissues were observed in LIRI model rats, and these effects were reversed by sevoflurane treatment. Activation of inflammation (elevated IL-1β, IL-6, and TNF-α) and apoptosis (elevated cleaved caspase3/caspase3 and Bax, degraded expression of Bcl2) and inhibition of autophagy (elevated P62, degraded expression of Beclin1 and LC3-II/LC3I) in the model group were ameliorated by sevoflurane. Transcriptome sequencing indicated that the PI3K/Akt pathway regulated by Rac1 plays an important role in LIRI. Furthermore, overexpression of Rac1 in a cell line inhibited the protective effect of sevoflurane in LIRI. Autophagy inhibition (3-MA) also prevented the protective effect of sevoflurane on inflammation and apoptosis. As shown in the present study, sevoflurane enhances autophagy via Rac1/PI3K/AKT signalling to attenuate lung ischaemia‒reperfusion injury.
Collapse
Affiliation(s)
- Xian Ding
- Department of Anesthesiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Xiang Gao
- Department of Anesthesiology, The Affiliated Fujian Maternity and Child Health Hospital of Fujian Medical University, 350001, China
| | - Aolin Ren
- Department of Anesthesiology and Pain Medicine, Jiangnan University Medical Center, Wuxi No.2 People's Hospital, 214002, China
| | - Jingjing Xu
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 214023, China
| | - Xuliang Jiang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, 200030, China
| | - Xiao Liang
- Department of Anesthesiology and Pain Medicine, Jiangnan University Medical Center, Wuxi No.2 People's Hospital, 214002, China
| | - Kangjie Xie
- Department of Anesthesiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Research Center for Neuro-Oncology Interaction, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, China
| | - Yan Zhou
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 214023, China
| | - Chunxiao Hu
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 214023, China
| | - Dongxiao Huang
- Department of Anesthesiology and Pain Medicine, Jiangnan University Medical Center, Wuxi No.2 People's Hospital, 214002, China.
| |
Collapse
|
5
|
Li Y, Gao W, Lei S, Wu X, Yuan T, Ma K, Chi K. Sevoflurane blocks KLF5-mediated transcriptional activation of ITGB2 to inhibit macrophage infiltration in hepatic ischemia/reperfusion injury. J Gene Med 2024; 26:e3692. [PMID: 38745073 DOI: 10.1002/jgm.3692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/01/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Sevoflurane (Sevo) preconditioning and postconditioning play a protective role against injury induced by hepatic ischemia/reperfusion (I/R). At the same time, the involvement of macrophage infiltration in this process and the precise mechanisms are unclear. Here, we designed this research to elucidate the protective effects of Sevo against hepatic I/R injury and the molecules involved. METHODS The alleviating effect of Sevo on the liver injury was analyzed by liver function analysis, hematoxylin and eosin staining, Masson trichrome staining, terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate nick end labeling, western blot analysis and an enzyme-linked immunosorbent assay. An in vitro cell model was developed using alpha mouse liver 12 (AML12) cells, and the cell model was treated with oxygen-glucose deprivation and reoxygenation and Sevo. Multiple bioinformatics databases were used to screen transcriptional regulators related to hepatic I/R injury and the targets of Krueppel-like factor 5 (KLF5). KLF5 expression was artificially upregulated alone or with integrin beta-2 (ITGB2) knockdown to substantiate their involvement in Sevo-mediated hepatoprotection. RESULTS Sevo protected the liver against I/R injury by reducing cell apoptosis and inflammatory response. KLF5 was upregulated in liver tissues following I/R injury, whereas KLF5 overexpression aggravated macrophage infiltration and liver injury induced by I/R injury. KLF5 bound to the promoter of ITGB2 to enhance ITGB2 transcription. Knockdown of ITGB2 reversed the aggravation of injury caused by KLF5 overexpression in mice and AML12 cells. CONCLUSIONS Sevo blocked KLF5-mediated transcriptional activation of ITGB2, thereby inhibiting macrophage infiltration in hepatic I/R injury.
Collapse
Affiliation(s)
- Ye Li
- Department of Anaesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Weinian Gao
- Department of Cardiovascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shuyan Lei
- Department of Anaesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoning Wu
- Department of Anaesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tao Yuan
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Kai Ma
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Kui Chi
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
6
|
Polat Y, Şengel N, Küçük A, Özdemir Ç, Yığman Z, Balcı AB, Ergörün Aİ, Kavutçu M, Arslan M. Effects of sevoflurane and fullerenol C60 on lower limb ischemia-reperfusion injury in streptozocin-induced diabetic mice. Sci Prog 2024; 107:368504241239444. [PMID: 38614462 PMCID: PMC11016234 DOI: 10.1177/00368504241239444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2024]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) poses a significant challenge for physicians, necessitating the management of cell damage and the preservation of organ functions. Various surgical procedures, such as vascular surgery on extremities, temporary cross-clamping of the abdominal aorta in aortic surgery, and the use of a tourniquet in extremity surgeries, may induce lower limb IRI. The susceptibility to IRI is heightened in individuals with diabetes. This study aimed to investigate the effects of fullerenol C60 and sevoflurane on mouse muscle tissue in a lower limb IRI model and to assess their potential in preventing complications arising from ischemia-reperfusion in mice with streptozocin-induced diabetes. METHODS A total of 36 adult Swiss albino mice were randomly divided into six groups, each consisting of six mice: control group (group C), diabetes group (group D), diabetes-ischemia/reperfusion group (group DIR), diabetes-ischemia/reperfusion-fullerenol C60 group (group DIR-FC60), diabetes-ischemia/reperfusion-sevoflurane group (group DIR-S), and diabetes-ischemia/reperfusion-sevoflurane-fullerenol C60 group (DIR-S-FC60). Streptozocin (55 mg/kg) was intraperitoneally administered to induce diabetes in the relevant groups, with mice displaying blood glucose levels of 250 mg/dL or higher at 72 h were considered diabetic. After 4 weeks, all groups underwent laparotomy under anesthesia. In DIR-FC60 and DIR-S-FC60 groups, fullerenol C60 (100 mg/kg) was intraperitoneally administrated 30 min before the ischemia period. Sevoflurane, delivered in 100% oxygen at a rate of 2.3% and 4 L/min, was administered during the ischemia period in DIR-S and DIR-S-FC60 groups. In the IR groups, a microvascular clamp was placed on the infrarenal abdominal aorta for 120 min during the ischemia period, followed by the removal of the clamp and a 120-min reperfusion period. At the end of the reperfusion, gastrocnemius muscle tissues were removed for histopathological and biochemical parameter examinations. RESULTS Histopathological examination revealed a significant reduction in the disorganization and degeneration of muscle cells in the DIR-S-FC60 group compared to the DIR group (p = 0.041). Inflammatory cell infiltration was notably lower in the DIR-S, DIR-FC60, and DIR-S-FC60 groups than in the DIR group (p = 0.031, p = 0.011, and p = 0.013, respectively). The total damage scores in the DIR-FC60 and DIR-S-FC60 groups were significantly lower than in the DIR group (p = 0.018 and p = 0.008, respectively). Furthermore, the levels of malondialdehyde (MDA) in the DIR-S, DIR-FC60, and DIR-S-FC60 groups were significantly lower than in the DIR group (p < 0.001, p < 0.001, and p < 0.001, respectively). Catalase (CAT) enzyme activity in the DIR-S, DIR-FC60, and DIR-S-FC60 groups was higher than in the DIR group (p = 0.001, p = 0.014, and p < 0.001, respectively). Superoxide dismutase (SOD) enzyme activity in the DIR-FC60 and DIR-S-FC60 groups was also higher than in the DIR group (p < 0.001 and p = 0.001, respectively). CONCLUSION Our findings indicate that administering fullerenol C60 30 min prior to ischemia in diabetic mice, in combination with sevoflurane, led to a reduction in oxidative stress and the correction of IR-related damage in muscle tissue histopathology. We believe that the administration of fullerenol C60 before IR, coupled with sevoflurane administration during IR, exerts a protective effect in mice.
Collapse
Affiliation(s)
- Yücel Polat
- Tekirdağ Dr İsmail Fehmi Cumalıoğlu City Hospital, Department of Cardiovascular Surgery, Tekirdağ, Turkey
| | - Necmiye Şengel
- Gazi University Faculty of Dentistry, Department of Oral and Maxillofacial Surgery, (As a specialist in Anesthesiology and Reanimation), Ankara, Turkey
| | - Ayşegül Küçük
- Kutahya Health Sciences University Faculty of Medicine, Department of Physiology, Kutahya, Turkey
| | - Çağrı Özdemir
- Mamak State Hospital, Department of Anesthesiology and Reanimation, Ankara, Turkey
| | - Zeynep Yığman
- Gazi University Faculty of Medicine, Department of Histology and Embryology, Ankara, Turkey
- Gazi University Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Turkey
| | | | - Aydan İremnur Ergörün
- Gazi University Faculty of Medicine, Department of Anesthesiology and Reanimation, Ankara, Turkey
| | - Mustafa Kavutçu
- Gazi University Faculty of Medicine, Department of Medical Biochemistry, Ankara, Turkey
| | - Mustafa Arslan
- Gazi University Faculty of Medicine, Department of Anesthesiology and Reanimation, Ankara, Turkey
- Gazi University, Life Sciences Application and Research Center, Ankara, Turkey
- Gazi University, Laboratory Animal Breeding and Experimental Researches Center (GÜDAM), Ankara, Turkey
| |
Collapse
|
7
|
Steinkühler T, Yang S, Hu MA, Jainandunsing JS, Jager NM, Erasmus ME, Struys MMRF, Bosch DJ, van Meurs M, Jabaudon M, Richard D, Timens W, Leuvenink HGD, Nieuwenhuijs-Moeke GJ. Ex Vivo Optimization of Donor Lungs with Inhaled Sevoflurane during Normothermic Ex Vivo Lung Perfusion (VITALISE): A Pilot and Feasibility Study in Sheep. Int J Mol Sci 2024; 25:2413. [PMID: 38397090 PMCID: PMC10888671 DOI: 10.3390/ijms25042413] [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: 12/21/2023] [Revised: 02/09/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Volatile anesthetics have been shown in different studies to reduce ischemia reperfusion injury (IRI). Ex vivo lung perfusion (EVLP) facilitates graft evaluation, extends preservation time and potentially enables injury repair and improvement of lung quality. We hypothesized that ventilating lungs with sevoflurane during EVLP would reduce lung injury and improve lung function. We performed a pilot study to test this hypothesis in a slaughterhouse sheep DCD model. Lungs were harvested, flushed and stored on ice for 3 h, after which EVLP was performed for 4 h. Lungs were ventilated with either an FiO2 of 0.4 (EVLP, n = 5) or FiO2 of 0.4 plus sevoflurane at a 2% end-tidal concentration (Cet) (S-EVLP, n = 5). Perfusate, tissue samples and functional measurements were collected and analyzed. A steady state of the target Cet sevoflurane was reached with measurable concentrations in perfusate. Lungs in the S-EVLP group showed significantly better dynamic lung compliance than those in the EVLP group (p = 0.003). Oxygenation capacity was not different in treated lungs for delta partial oxygen pressure (PO2; +3.8 (-4.9/11.1) vs. -11.7 (-12.0/-3.2) kPa, p = 0.151), but there was a trend of a better PO2/FiO2 ratio (p = 0.054). Perfusate ASAT levels in S-EVLP were significantly reduced compared to the control group (198.1 ± 93.66 vs. 223.9 ± 105.7 IU/L, p = 0.02). We conclude that ventilating lungs with sevoflurane during EVLP is feasible and could be useful to improve graft function.
Collapse
Affiliation(s)
- Timo Steinkühler
- Department of Anesthesiology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Shuqi Yang
- Department of Anesthesiology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Michiel A. Hu
- Department of Thoracic Surgery, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Jayant S. Jainandunsing
- Department of Anesthesiology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Neeltina M. Jager
- Department of Anesthesiology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Michiel E. Erasmus
- Department of Thoracic Surgery, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Michel M. R. F. Struys
- Department of Anesthesiology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Department of Basic and Applied Medical Sciences, Ghent University, 9000 Ghent, Belgium
| | - Dirk J. Bosch
- Department of Anesthesiology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Matijs van Meurs
- Department of Critical Care, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Matthieu Jabaudon
- Department of Perioperative Medicine, University Hospital Clermont-Ferrand, 63001 Clermont-Ferrand, France
- Institute of Genetics, Reproduction & Development, University Clermont Auvergne, 63001 Clermont-Ferrand, France
- National Institute of Health and Medical Research (INSERM), National Center for Scientific Research (CNRS), 75794 Paris, France
| | - Damien Richard
- Department of Pharmacology and Toxicology, University Hospital Clermont-Ferrand, University Clermont Auvergne, 63001 Clermont-Ferrand, France
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Henri G. D. Leuvenink
- Department of Surgery, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | | |
Collapse
|
8
|
Şengel N, Küçük A, Özdemir Ç, Sezen ŞC, Kip G, Er F, Dursun AD, Polat Y, Kavutçu M, Arslan M. The Effect of Sevoflurane and Fullerenol C 60 on the Liver and Kidney in Lower Extremity Ischemia-Reperfusion Injury in Mice with Streptozocin-Induced Diabetes. Int J Nanomedicine 2023; 18:7543-7557. [PMID: 38111848 PMCID: PMC10725837 DOI: 10.2147/ijn.s432924] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/30/2023] [Indexed: 12/20/2023] Open
Abstract
Objective This study aimed to demonstrate whether fullerenol C60, sevoflurane anesthesia, or a combination of both had protective effects on the liver and kidneys in lower extremity ischemia-reperfusion injury (IRI) in mice with streptozocin-induced diabetes. Methods A total of 46 Swiss albino mice were divided into six groups as follows: control group (group C, n=7), diabetes group (group D, n=7), diabetes-ischemia/reperfusion (group DIR, n=8), diabetes-ischemia/reperfusion-fullerenol C60 (group DIR-FC60, n=8), diabetes-ischemia/reperfusion-sevoflurane (group DIR-S, n=8), and the diabetes-ischemia/reperfusion-fullerenol C60-sevoflurane (group DIR-S-FC60, n=8). Fullerenol C60 (100mg/kg) was administered intraperitoneally 30 min before the ischemia-reperfusion procedure to the fullerenol groups (DIR-FC60 and DIR-S-FC60). In the DIR groups, 2 hours (h) ischemia-2h reperfusion periods were performed. In the sevoflurane groups, sevoflurane was applied during the ischemia-reperfusion period with 100% O2. Liver and kidney tissues were removed at the end of the reperfusion procedure for biochemical and histopathological examinations. Results In liver tissue, hydropic degeneration, sinusoidal dilatation, pycnotic nuclei, prenecrotic cells, and mononuclear cell infiltration in parenchyma were significantly more frequent in group DIR than in groups D and group C. In terms of the histopathologic criteria examined, more positive results were seen in group DIR-FC60, and when group DIR-FC60 was compared with group DIR, the difference was significant. The best results in AST, ALT, glucose, TBARS levels, and SOD enzyme activities in liver tissue were in group DIR-FC60 compared with group DIR, followed by groups DIR-S-FC60 and DIR-S, respectively. Regarding TBARS levels and SOD enzyme activities in kidney tissue, the best results were in groups DIR-FC60, DIR-S-FC60, and DIR-S, respectively. Conclusion According to our findings, it is clear that fullerenol C60 administered intraperitoneally 30 min before ischemia, alone or together with sevoflurane, reduces oxidative stress in distant organ damage caused by lower extremity IRI, and reduces liver and kidney tissue damage in histopathologic examinations.
Collapse
Affiliation(s)
- Necmiye Şengel
- Department of Oral and Maxillofacial Surgery, (As a Specialist in Anesthesiology and Reanimation), Gazi University Faculty of Dentistry, Ankara, Turkey
| | - Ayşegül Küçük
- Department of Physiology, Kutahya Health Sciences University Faculty of Medicine, Kutahya, Turkey
| | - Çağrı Özdemir
- Department of Anesthesiology and Reanimation, Mamak State Hospital, Ankara, Turkey
| | - Şaban Cem Sezen
- Department of Histology and Embryology, Kırıkkale University Faculty of Medicine, Kırıkkale, Turkey
| | - Gülay Kip
- Department of Anesthesiology and Reanimation, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Fatma Er
- Department of Medical Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ali Doğan Dursun
- Department of Physiology, Atılım University Faculty of Medicine, Ankara, Turkey
| | - Yücel Polat
- Cardiovascular Surgery, Tekirdağ Dr. Ismail Fehmi Cumalıoğlu City Hospital, Tekirdağ, Turkey
| | - Mustafa Kavutçu
- Department of Medical Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Mustafa Arslan
- Department of Anesthesiology and Reanimation, Gazi University Faculty of Medicine, Ankara, Turkey
- Life Sciences Application and Research Center, Gazi University, Ankara, Turkey
- Laboratory Animal Breeding and Experimental Researches Center (GÜDAM), Gazi University, Ankara, Turkey
| |
Collapse
|
9
|
Yang C, Deng L, Bao F, Jiang H, Zhang L. Sevoflurane with Low Concentration Decrease DNA Methylation on Chronic Obstructive Pulmonary Disease (COPD)-Related Gene Promoter in COPD Rat. COPD 2023; 20:348-356. [PMID: 38010369 DOI: 10.1080/15412555.2023.2278282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/28/2023] [Indexed: 11/29/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a difficult-to-cure disease that mainly affects the respiratory system. Inhaled anesthetic drug such as sevoflurane plays a controversial role in COPD by different concentration, but the underlying epigenetic mechanism remains unclear. Here, we prepared lipopolysaccharide (LPS)-induced COPD rat model, and isolated Alveolar type II (ATII) cells. We mainly focused DNA methylation on the promoter of COPD-related genes including Sftpa1, Napsa, Ca2, Sfta2, Lamp3, Wif1, Pgc, and Etv5. We observed COPD rat treated by sevoflurane with low (0.5%) and high (2%) concentrations displayed an opposite DNA methylation pattern. These six genes' promoter were all hypomethylated by 0.5% sevoflurane whereas hypermethylated by 2% sevoflurane, accompanied with the opposite transcriptional activity. We further verified that the DNMT1 binding ability contributed to DNA methylation these six genes' promoter. Moreover, we also captured DNMT1 and identified REC8 meiotic recombination protein (REC8) as the specific binding protein only existed in ATII cells treated with 0.5% sevoflurane rather than 2% and control. The binding ability of REC8 on these target genes' promoter showed highly positive correlation with DNMT1. In summary, we uncovered a potential epigenetic role of sevoflurane with low concentration in ATII cells of COPD that may help us deeply understand the pathogenesis and treatment mechanism of inhaled anesthesia drugs in COPD via a dose-dependent manner.
Collapse
Affiliation(s)
- Chuanxin Yang
- Department of Anesthesiology, Qingpu Branch of Zhongshan, Fudan University, Shanghai, China
| | - Libing Deng
- Department of Anesthesiology, Qingpu Branch of Zhongshan, Fudan University, Shanghai, China
| | - Fang Bao
- Department of Anesthesiology, Qingpu Branch of Zhongshan, Fudan University, Shanghai, China
| | - Hui Jiang
- Department of Anesthesiology, Qingpu Branch of Zhongshan, Fudan University, Shanghai, China
| | - Long Zhang
- Department of Anesthesiology, Qingpu Branch of Zhongshan, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Martínez-Castro S, Monleón B, Puig J, Ferrer Gomez C, Quesada M, Pestaña D, Balvis A, Maseda E, de la Rica AS, Feijoo AM, Badenes R. Sedation with Sevoflurane versus Propofol in COVID-19 Patients with Acute Respiratory Distress Syndrome: Results from a Randomized Clinical Trial. J Pers Med 2023; 13:925. [PMID: 37373914 DOI: 10.3390/jpm13060925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/08/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) related to COVID-19 (coronavirus disease 2019) led to intensive care units (ICUs) collapse. Amalgams of sedative agents (including volatile anesthetics) were used due to the clinical shortage of intravenous drugs (mainly propofol and midazolam). METHODS A multicenter, randomized 1:1, controlled clinical trial was designed to compare sedation using propofol and sevoflurane in patients with ARDS associated with COVID-19 infection in terms of oxygenation and mortality. RESULTS Data from a total of 17 patients (10 in the propofol arm and 7 in the sevoflurane arm) showed a trend toward PaO2/FiO2 improvement and the sevoflurane arm's superiority in decreasing the likelihood of death (no statistical significance was found). CONCLUSIONS Intravenous agents are the most-used sedative agents in Spain, even though volatile anesthetics, such as sevoflurane and isoflurane, have shown beneficial effects in many clinical conditions. Growing evidence demonstrates the safety and potential benefits of using volatile anesthetics in critical situations.
Collapse
Affiliation(s)
- Sara Martínez-Castro
- Department Anesthesiology, Surgical-Trauma Intensive Care and Pain Clinic, Hospital Clínic Universitari, University of Valencia, 46010 Valencia, Spain
| | - Berta Monleón
- Department Anesthesiology, Surgical-Trauma Intensive Care and Pain Clinic, Hospital Clínic Universitari, University of Valencia, 46010 Valencia, Spain
| | - Jaume Puig
- Anesthesiology and Intensive Care Department, Consorcio Hospital General Universitario, 46014 Valencia, Spain
| | - Carolina Ferrer Gomez
- Anesthesiology and Intensive Care Department, Consorcio Hospital General Universitario, 46014 Valencia, Spain
| | - Marta Quesada
- Anesthesiology and Intensive Care Department, Consorcio Hospital General Universitario, 46014 Valencia, Spain
| | - David Pestaña
- Anesthesiology and Intensive Care Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Alberto Balvis
- Anesthesiology and Intensive Care Department, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Emilio Maseda
- Surgical Critical Care Department, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Alejandro Suárez de la Rica
- Anesthesiology and Surgical Critical Care Department, Hospital Universitario De La Princesa, 28006 Madrid, Spain
| | - Ana Monero Feijoo
- Surgical Critical Care Department, Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Rafael Badenes
- Department Anesthesiology, Surgical-Trauma Intensive Care and Pain Clinic, Hospital Clínic Universitari, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
11
|
Yamanashi K, Ohsumi A, Oda H, Tanaka S, Yamada Y, Nakajima D, Date H. Reduction of donor mononuclear phagocytes with clodronate-liposome during ex vivo lung perfusion attenuates ischemia-reperfusion injury. J Thorac Cardiovasc Surg 2023; 165:e181-e203. [PMID: 36404143 DOI: 10.1016/j.jtcvs.2022.10.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Clodronate-liposome is used for depleting mononuclear phagocytes associated with ischemia-reperfusion injury. We hypothesized that administration of clodronate-liposome into the perfusate during ex vivo lung perfusion could reduce mononuclear phagocytes and attenuate ischemia-reperfusion injury. METHODS First, the number of mononuclear phagocytes in flushed grafts (minimum cold ischemic time, 6-hour cold ischemic time, 15-hour cold ischemic time, and 18-hour cold ischemic time; n = 6 each) was determined using flow cytometry. Second, grafts (15-hour cold ischemic time) were allocated to control or clodronate (n = 5 each). In the clodronate group, clodronate-liposome is administered into the perfusate. After 4 hours of ex vivo lung perfusion, the number of mononuclear phagocytes in the perfusate and lung tissues was measured. Third, grafts (15-hour cold ischemic time) were allocated to control or clodronate (n = 6 each). After 4 hours of ex vivo lung perfusion, the left lungs were transplanted and reperfused for 2 hours. Lung function was evaluated, and samples were analyzed. RESULTS First, mononuclear phagocytes remain in flushed grafts after prolonged cold ischemia. Second, the number of mononuclear phagocytes in lung tissues after ex vivo lung perfusion was significantly reduced in the clodronate group (P = .008). Third, lung compliance and vascular resistance during ex vivo lung perfusion were significantly improved in the clodronate group (P < .001 for both). Blood oxygenation and pulmonary edema were significantly improved in the clodronate group after 2 hours of reperfusion (P = .015 and P = .026, respectively). Histological findings showed reduced lung injury in the clodronate group (P = .013). CONCLUSIONS Administration of clodronate-liposome into the perfusate during ex vivo lung perfusion resulted in a significant reduction of mononuclear phagocytes in donor lungs, leading to attenuation of ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Keiji Yamanashi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihiro Ohsumi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Hiromi Oda
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satona Tanaka
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshito Yamada
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daisuke Nakajima
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
12
|
Benoit L, Dieu A, Foguenne M, Bonaccorsi-Riani E. Experimental and Clinical Aspects of Sevoflurane Preconditioning and Postconditioning to Alleviate Hepatic Ischemia-Reperfusion Injury: A Scoping Review. Int J Mol Sci 2023; 24:2340. [PMID: 36768670 PMCID: PMC9916998 DOI: 10.3390/ijms24032340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/21/2022] [Accepted: 01/22/2023] [Indexed: 01/27/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) is an inflammatory process inherent in organ transplantation procedures. It is associated with tissue damage and, depending on its intensity, can impact early graft function. In liver transplantation (LT), strategies to alleviate IRI are essential in order to increase the use of extended criteria donor (ECD) grafts, which are more susceptible to IRI, as well as to improve postoperative graft and patient outcomes. Sevoflurane, a commonly used volatile anesthetic, has been shown to reduce IRI. This scoping review aims to give a comprehensive overview of the existing experimental and clinical data regarding the potential benefits of sevoflurane for hepatic IRI (HIRI) and to identify any gaps in knowledge to guide further research. We searched Medline and Embase for relevant articles. A total of 380 articles were identified, 45 of which were included in this review. In most experimental studies, the use of sevoflurane was associated with a significant decrease in biomarkers of acute liver damage and oxidative stress. Administration of sevoflurane before hepatic ischemia (preconditioning) or after reperfusion (postconditioning) appears to be protective. However, in the clinical setting, results are conflicting. While some studies showed a reduction of postoperative markers of liver injury, the benefit of sevoflurane on clinical outcomes and graft survival remains unclear. Further prospective clinical trials remain necessary to assess the clinical relevance of the use of sevoflurane as a protective factor against HIRI.
Collapse
Affiliation(s)
- Loïc Benoit
- Department of Anesthesiology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Audrey Dieu
- Department of Anesthesiology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Maxime Foguenne
- Abdominal Transplant Unit, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Pôle de Chirurgie Expérimentale et Transplantation-Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Eliano Bonaccorsi-Riani
- Abdominal Transplant Unit, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
- Pôle de Chirurgie Expérimentale et Transplantation-Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| |
Collapse
|
13
|
Zheng F, Wu X, Zhang J, Fu Z, Zhang Y. Sevoflurane reduces lipopolysaccharide-induced apoptosis and pulmonary fibrosis in the RAW264.7 cells and mice models to ameliorate acute lung injury by eliminating oxidative damages. Redox Rep 2022; 27:139-149. [PMID: 35801580 PMCID: PMC9272930 DOI: 10.1080/13510002.2022.2096339] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objectives Sevoflurane is identified as an effective candidate drug for acute lung injury (ALI) treatment, but its curing effects and detailed mechanisms have not been fully disclosed. The present study was designed to resolve this academic issue. Methods The ALI mice models were established, and Hematoxylin-eosin staining assay was performed to examine tissue morphologies. Cell viability was determined by CCK-8 assay, and Annexin V-FITC/PI double staining assay was used to examine cell apoptosis. The expression levels of proteins were determined by performing Western Blot analysis and immunofluorescence staining assay. ROS levels were examined by using DCFH-DA staining assay. Results In this study, we investigated this issue and the ALI models were respectively established by treating the BALB/c mice and the murine macrophage cell line RAW264.7 with different concentrations of lipopolysaccharide (LPS) in vivo and in vitro, which were subsequently subjected to sevoflurane co-treatment. The results showed that sevoflurane reduced LPS-induced ALI in mice and suppressed LPS-triggered oxidative stress and apoptotic cell death in the RAW264.7 cells. Interestingly, we evidenced that the elimination of reactive oxygen species (ROS) by N-acetyl-L-cysteine (NAC) reversed LPS-induced cell apoptosis in RAW264.7 cells. Then, we verified that sevoflurane suppressed oxidative damages to restrain LPS-induced apoptotic cell death in the RAW264.7 cells through activating the anti-oxidant Keap1/Nrf2 pathway. Mechanistically, sevoflurane down-regulated Keap1 and upregulated Nrf2 in nucleus to activate the downstream anti-oxidant signaling cascades, which further ameliorated LPS-induced cell apoptosis and lung injury by eliminating oxidative damages. Discussion Taken together, our study illustrated that the sevoflurane attenuates LPS-induced ALI by inhibiting oxidative stress-mediated apoptotic cell death and inflammation, and the Keap1/Nrf2 pathway played an important role in this process.
Collapse
Affiliation(s)
- Fushuang Zheng
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xiuying Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yan Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
14
|
Jabaudon M, Zhai R, Blondonnet R, Bonda WLM. Inhaled sedation in the intensive care unit. Anaesth Crit Care Pain Med 2022; 41:101133. [PMID: 35907598 DOI: 10.1016/j.accpm.2022.101133] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 11/19/2022]
Abstract
Inhaled sedation with halogenated agents, such as isoflurane or sevoflurane, is now feasible in intensive care unit (ICU) patients through dedicated vaporisers and scavenging systems. Such a sedation strategy requires specific equipment and adequate training of ICU teams. Isoflurane and sevoflurane have ideal pharmacological properties that allow efficient, well-tolerated, and titratable light-to-deep sedation. In addition to their function as sedative agents, these molecules may have clinical benefits that could be especially relevant to ICU patients. Our goal was to summarise the pharmacological basis and practical aspects of inhaled ICU sedation, review the available evidence supporting inhaled sedation as a viable alternative to intravenous sedation, and discuss the remaining areas of uncertainty and future perspectives of development.
Collapse
Affiliation(s)
- Matthieu Jabaudon
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France; GReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France.
| | - Ruoyang Zhai
- GReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | - Raiko Blondonnet
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France; GReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | | |
Collapse
|
15
|
Ultrasound Evaluation of the Diaphragm in Clinical Anesthesia. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:2163225. [PMID: 35281531 PMCID: PMC8913060 DOI: 10.1155/2022/2163225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/10/2021] [Accepted: 06/24/2021] [Indexed: 11/22/2022]
Abstract
When the human body is anesthetized, the human nerve tissue will be greatly affected, which also affects the breathing of the human body. The respiration during anesthesia is a lack of initiative, and the energy efficiency of the diaphragm in the lungs is very important to the safety of anesthesia. In this paper, the application of the ultrasound evaluation of the diaphragm in clinical anesthesia was studied. In this paper, 24 patients who underwent lung examination under medical anesthesia at our hospital were evaluated by the ultrasound vertical mixed echo method. Through patient voluntary selection and consent, 16 patients were examined with B-mode ultrasound and the other 8 patients with M-mode ultrasound to compare the effects of different ultrasounds on diaphragm image quality. In addition, this paper also analyzes the differences between different ultrasounds and the strengths and weaknesses of diaphragmatic ultrasound evaluation in clinical anesthesia. The suggestions of using different ultrasounds in ultrasonic evaluation are given. The study showed that 16 cases of B-mode ultrasound evaluation of the diaphragm obtained ultrasound images which showed a large field of vision, acoustic frequency between 7 and 18 MHz, and thickness difference between 0.35 and 0.52 cm. In 8 patients with the diaphragm evaluated by M-mode ultrasound, the local features of M-mode ultrasound images were clearer than those of B-mode ultrasound images, but the visual field area was smaller, the acoustic frequency was between 10 and 15 MHz, and the thickness difference was between 0.12 and 0.18 cm. Based on the above data, this paper suggests that, in the ultrasonic evaluation of the diaphragm, B-mode ultrasound should be used to check the patients first, and then M-mode ultrasound should be used to check the parts with poor quality so that the accurate diaphragm quality of patients can be obtained in the vast majority of patients.
Collapse
|
16
|
Zhang K, Xu X, Hu L. Sevoflurane attenuates hepatic ischemia reperfusion injury by the miR-122/Nrf2 pathway. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:350. [PMID: 35433991 PMCID: PMC9011315 DOI: 10.21037/atm-22-115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/21/2022] [Indexed: 11/18/2022]
Abstract
Background Sevoflurane can protect organs from ischemia-reperfusion (IR) injury, but the mechanism is still unclear. MicroRNA-122 (miR-122) is a liver-specific microRNA (miRNA) and regulates liver function. Therefore, this study aims to elucidate the relationship between the protective effect of sevoflurane and miR-122 in liver IR injury. Methods Wistar rats were divided into the following groups: sham, IR, IR + sevoflurane, IR + miR-122 antagomir, and IR + miR-122 antagomir + sevoflurane. Hematoxylin and eosin (H&E) staining and Suzuki score were used to evaluate the pathological damage of the liver. The levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), lactate dehydrogenase (LDH), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and IL-10 in the serum and the levels of malondialdehyde (MDA), superoxide dismutase (SOD), and nitric oxide (NO) in the liver homogenate supernatant were detected by using the corresponding kit. Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) and flow cytometry was applied to evaluate the apoptosis of liver tissues. The expression of nuclear factor E2-related factor 2 (Nrf2), miR-122, p53, and HO-1 in liver tissue was evaluated by using immunohistochemistry, qRT-PCR, and western blot as needed. Results Compared to the IR group, the sevoflurane post-treatment or miR-122 antagomir groups showed improved liver injury, decreased Suzuki score, inhibited the levels of AST, ALT, LDH, MDA, NO, TNF-α, IL-1β, and IL-6, increased levels of SOD, IL-10, and inhibited hepatocyte apoptosis. Regarding the molecular mechanism, sevoflurane post-treatment fostered the expression of HO-1, promoted the transport of Nrf2 from cytoplasm to the nucleus, and decreased the expression of miR-122 and p53. The combined use of miR-122 antagomir and sevoflurane enhanced the protective effect of miR-122 antagomir in liver injury in IR rats. Conclusions Sevoflurane protected the liver from IR damage by regulating the miR-122/Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Anesthesiology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Xia Xu
- Department of Anesthesiology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Lihong Hu
- Department of Anesthesiology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| |
Collapse
|
17
|
Jin X, Kaes J, Van Slambrouck J, Inci I, Arni S, Geudens V, Heigl T, Jansen Y, Carlon MS, Vos R, Van Raemdonck D, Zhang Y, Vanaudenaerde BM, Ceulemans LJ. A Comprehensive Review on the Surgical Aspect of Lung Transplant Models in Mice and Rats. Cells 2022; 11:cells11030480. [PMID: 35159289 PMCID: PMC8833959 DOI: 10.3390/cells11030480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/17/2022] [Accepted: 01/27/2022] [Indexed: 12/20/2022] Open
Abstract
Lung transplantation improves the outcome and quality of life of patients with end-stage pulmonary disease. However, the procedure is still hampered by the lack of suitable donors, the complexity of the surgery, and the risk of developing chronic lung allograft dysfunction. Over the past decades, translational experiments in animal models have led to a better understanding of physiology and immunopathology following the lung transplant procedure. Small animal models (e.g., rats and mice) are mostly used in experiments regarding immunology and pathobiology and are preferred over large animal models due to the ethical aspects, the cost-benefit balance, and the high throughput possibility. In this comprehensive review, we summarize the reported surgical techniques for lung transplantation in rodent models and the management of perioperative complications. Furthermore, we propose a guide to help identify the appropriate species for a given experiment and discuss recent experimental findings in small animal lung transplant models.
Collapse
Affiliation(s)
- Xin Jin
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium; (X.J.); (J.K.); (J.V.S.); (V.G.); (T.H.); (Y.J.); (M.S.C.); (R.V.); (D.V.R.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium; (X.J.); (J.K.); (J.V.S.); (V.G.); (T.H.); (Y.J.); (M.S.C.); (R.V.); (D.V.R.); (B.M.V.)
| | - Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium; (X.J.); (J.K.); (J.V.S.); (V.G.); (T.H.); (Y.J.); (M.S.C.); (R.V.); (D.V.R.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Ilhan Inci
- Department of Thoracic Surgery, University Hospital Zürich, 8091 Zürich, Switzerland; (I.I.); (S.A.)
| | - Stephan Arni
- Department of Thoracic Surgery, University Hospital Zürich, 8091 Zürich, Switzerland; (I.I.); (S.A.)
| | - Vincent Geudens
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium; (X.J.); (J.K.); (J.V.S.); (V.G.); (T.H.); (Y.J.); (M.S.C.); (R.V.); (D.V.R.); (B.M.V.)
| | - Tobias Heigl
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium; (X.J.); (J.K.); (J.V.S.); (V.G.); (T.H.); (Y.J.); (M.S.C.); (R.V.); (D.V.R.); (B.M.V.)
| | - Yanina Jansen
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium; (X.J.); (J.K.); (J.V.S.); (V.G.); (T.H.); (Y.J.); (M.S.C.); (R.V.); (D.V.R.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Marianne S. Carlon
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium; (X.J.); (J.K.); (J.V.S.); (V.G.); (T.H.); (Y.J.); (M.S.C.); (R.V.); (D.V.R.); (B.M.V.)
- Department of Pharmaceutical and Pharmacological Sciences, Molecular Virology and Gene Therapy, KU Leuven, 3000 Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium; (X.J.); (J.K.); (J.V.S.); (V.G.); (T.H.); (Y.J.); (M.S.C.); (R.V.); (D.V.R.); (B.M.V.)
- Department of Respiratory Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Dirk Van Raemdonck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium; (X.J.); (J.K.); (J.V.S.); (V.G.); (T.H.); (Y.J.); (M.S.C.); (R.V.); (D.V.R.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Yi Zhang
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Correspondence: (Y.Z.); (L.J.C.); Tel.: +32-16-34-68-20 (L.J.C.)
| | - Bart M. Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium; (X.J.); (J.K.); (J.V.S.); (V.G.); (T.H.); (Y.J.); (M.S.C.); (R.V.); (D.V.R.); (B.M.V.)
| | - Laurens J. Ceulemans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium; (X.J.); (J.K.); (J.V.S.); (V.G.); (T.H.); (Y.J.); (M.S.C.); (R.V.); (D.V.R.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Correspondence: (Y.Z.); (L.J.C.); Tel.: +32-16-34-68-20 (L.J.C.)
| |
Collapse
|
18
|
Effects of Sevoflurane on Apoptosis of Myocardial Cells in IRI Rats. BIOMED RESEARCH INTERNATIONAL 2022; 2021:3347949. [PMID: 35005016 PMCID: PMC8741344 DOI: 10.1155/2021/3347949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 11/18/2022]
Abstract
Background Cardiomyocyte apoptosis functions essentially in ischemia/reperfusion- (I/R-) induced myocardial injury. It is suggested that autophagy is widely implicated in the regulation of cell survival and death. Sevoflurane, as a largely used inhalational general anesthetic, has been shown to have a protective effect on cardiomyocytes. However, it was yet elusive on the underlying mechanisms. Aim The objective of this study is to investigate the association of sevoflurane-mediated cardioprotective effects with autophagy regulation. Methods An in vitro hypoxia model was established in primary cardiomyocytes from fresh myocardial tissue of the rats. The apoptosis rate of myocardial cells treated with hypoxia and treated with sevoflurane was measured. Western blot and immunocytochemical assay were used to measure the protein expression. The cell proliferation rate and cell apoptosis were measured using the MTT assay and flow cytometry, respectively. Results The expression of apoptotic proteins including B cell lymphoma-2 (Bcl-2), CCAAT/enhancer-binding protein homologous protein (CHOP), glucose-regulated protein 78 (GRP78), and Bcl-2-associated X protein (BAX) in myocardium treated with sevoflurane was significantly lower than that in myocardium treated with hypoxia. The expression of adhesion proteins such as intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and E-selectin in myocardium treated with sevoflurane was higher than that in myocardium treated with hypoxia, suggesting better connectivity of the myocardium. Conclusion Sevoflurane treatment reduced the apoptosis of myocardial cells after hypoxia treatment.
Collapse
|
19
|
Yang J, Tang L, Zhang F, Yang T, Lu T, Sun K, Sun N, Ren J, Yan M. Sevoflurane preconditioning promotes mesenchymal stem cells to relieve myocardial ischemia/reperfusion injury via TRPC6-induced angiogenesis. Stem Cell Res Ther 2021; 12:584. [PMID: 34809715 PMCID: PMC8607627 DOI: 10.1186/s13287-021-02649-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/06/2021] [Indexed: 01/09/2023] Open
Abstract
Background Ischemic heart diseases is one of the leading causes of death worldwide. Although revascularization timely is an effective therapeutic intervention to salvage the ischemic myocardium, reperfusion itself causes additional myocardial injury called ischemia/reperfusion (I/R) injury. Bone marrow-derived mesenchymal stem cells (MSCs) is one of the promising cells to alleviate ischemic myocardial injury. However, this cell therapy is limited by poor MSCs survival after transplantation. Here, we investigated whether sevoflurane preconditioning could promote MSCs to attenuate myocardial I/R injury via transient receptor potential canonical channel 6 (TRPC6)-induced angiogenesis. Methods The anti-apoptotic effect of sevoflurane preconditioning on MSCs was determined by Annexin V-FITC/propidium iodide staining. TRPC6, hypoxia-inducible factor-1α (HIF-1α), Chemokine receptor 4 (CXCR4) and vascular endothelial growth factor (VEGF) protein expressions and VEGF release from MSCs were determined after hypoxia and reoxygenation (H/R). Small interfering RNA (siRNA) was used to knock down TRPC6 gene expression in MSCs. The angiogenesis of human umbilical vein endothelial cells (HUVECs) co-cultured with MSCs was determined by Matrigel tube formation. Myocardial I/R mouse model was induced by occluding left anterior descending coronary artery for 30 min and then reperfusion. MSCs or sevoflurane preconditioned MSCs were injected around the ligature border zone 5 min before reperfusion. Left ventricle systolic function, infarction size, serum LDH, cTnI and inflammatory cytokines were determined after reperfusion. Results Sevoflurane preconditioning up-regulated TRPC6, HIF-1α, CXCR4 and VEGF expressions in MSCs and VEGF release from MSCs under H/R, which were reversed by knockdown of TRPC6 gene using siRNA in MSCs. Furthermore, sevoflurane preconditioning promoted the angiogenic and anti-inflammatory effect of HUVECs co-cultured with MSCs. Sevoflurane preconditioned MSCs improved left ventricle systolic function and alleviated myocardial infarction and inflammation in mice subjected to I/R insult. Conclusion The current findings reveal that sevoflurane preconditioned MSCs boost angiogenesis in HUVECs subjected to H/R insult and attenuate myocardial I/R injury, which may be mediated by TRPC6 up-regulated HIF-1α, CXCR4 and VEGF. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02649-3.
Collapse
Affiliation(s)
- Jinting Yang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Lihui Tang
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Fengjiang Zhang
- Clinical Skill Training Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Tingting Yang
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, 710061, China
| | - Ting Lu
- Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Kai Sun
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Na Sun
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Jinxuan Ren
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Min Yan
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
20
|
Ji H, Li H, Zhang H, Cheng Z. Role of microRNA‑218‑5p in sevoflurane‑induced protective effects in hepatic ischemia/reperfusion injury mice by regulating GAB2/PI3K/AKT pathway. Mol Med Rep 2021; 25:1. [PMID: 34726254 PMCID: PMC8600399 DOI: 10.3892/mmr.2021.12517] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 05/20/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatic ischemia/reperfusion (I/R) injury (HIRI) often occurs following tissue resection, hemorrhagic shock or transplantation surgery. Previous investigations showed that sevoflurane (Sevo), an inhalation anesthetic, had protective properties against different organ damage in animal models including HIRI. This study is aimed to investigate the underlying mechanisms involved in the protective effects of Sevo on HIRI. The present study results showed that treatment with Sevo improved histologic damage, inflammatory response, oxidative stress and apoptosis after hepatic I/R, indicating the protective role of Sevo against liver I/R injury. Importantly, in order to determine the molecular mechanism of Sevo in HIRI, the focus of the study was on microRNA (miR) regulation. By retrieving the microarray data in the Gene Expression Omnibus dataset (GSE72315), miR-218-5p was found to be significantly downregulated by Sevo. Moreover, miR-218-5p overexpression using agomiR-218-5p reversed the protective roles of Sevo against HIRI. Furthermore, GAB2, a positive regulator of PI3K/AKT signaling pathway, was found as a target gene of miR-218-5p. It was also found that the Sevo-mediated protective effects may be dependent on the activation of GAB2/PI3K/AKT. Collectively, these data revealed that Sevo alleviated HIRI in mice by restraining apoptosis, relieving oxidative stress and inflammatory response through the miR-218-5p/GAB2/PI3K/AKT pathway, which helps in understanding the novel mechanism of the hepatic-protection of Sevo.
Collapse
Affiliation(s)
- Hui Ji
- Department of Anesthesiology, Xinhua Hospital, Chongming Branch, Shanghai 202150, P.R. China
| | - Hui Li
- Department of Anesthesiology, Xinhua Hospital, Chongming Branch, Shanghai 202150, P.R. China
| | - Haixia Zhang
- Department of Anesthesiology, Xinhua Hospital, Chongming Branch, Shanghai 202150, P.R. China
| | - Zhijun Cheng
- Department of Anesthesiology, Xinhua Hospital, Chongming Branch, Shanghai 202150, P.R. China
| |
Collapse
|
21
|
Michaelsen VS, Ribeiro RVP, Brambate E, Ali A, Wang A, Pires L, Kawashima M, Zhang Y, Gazzalle A, Keshavjee S, Del Sorbo L, Cypel M. A novel pre-clinical strategy to deliver antimicrobial doses of inhaled nitric oxide. PLoS One 2021; 16:e0258368. [PMID: 34644318 PMCID: PMC8513841 DOI: 10.1371/journal.pone.0258368] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/25/2021] [Indexed: 11/26/2022] Open
Abstract
Effective treatment of respiratory infections continues to be a major challenge. In high doses (≥160 ppm), inhaled Nitric Oxide (iNO) has been shown to act as a broad-spectrum antimicrobial agent, including its efficacy in vitro for coronavirus family. However, the safety of prolonged in vivo implementation of high-dose iNO therapy has not been studied. Herein we aim to explore the feasibility and safety of delivering continuous high-dose iNO over an extended period of time using an in vivo animal model. Yorkshire pigs were randomized to one of the following two groups: group 1, standard ventilation; and group 2, standard ventilation + continuous iNO 160 ppm + methylene blue (MB) as intravenous bolus, whenever required, to maintain metHb <6%. Both groups were ventilated continuously for 6 hours, then the animals were weaned from sedation, mechanical ventilation and followed for 3 days. During treatment, and on the third post-operative day, physiologic assessments were performed to monitor lung function and other significative markers were assessed for potential pulmonary or systemic injury. No significant change in lung function, or inflammatory markers were observed during the study period. Both gas exchange function, lung tissue cytokine analysis and histology were similar between treated and control animals. During treatment, levels of metHb were maintained <6% by administration of MB, and NO2 remained <5 ppm. Additionally, considering extrapulmonary effects, no significant changes were observed in biochemistry markers. Our findings showed that high-dose iNO delivered continuously over 6 hours with adjuvant MB is clinically feasible and safe. These findings support the development of investigations of continuous high-dose iNO treatment of respiratory tract infections, including SARS-CoV-2.
Collapse
Affiliation(s)
- Vinicius S. Michaelsen
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Rafaela V. P. Ribeiro
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Edson Brambate
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Aadil Ali
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Aizhou Wang
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Layla Pires
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Mitsuaki Kawashima
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Yu Zhang
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Anajara Gazzalle
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Lorenzo Del Sorbo
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Xie D, Deng H, Feng H. Sevoflurane exerts improved protective effects than propofol on hypoxia-reoxygenation injury by regulating the microRNA-221-5p/ADAM8 axis in cardiomyocytes. Exp Ther Med 2021; 22:893. [PMID: 34257708 PMCID: PMC8243314 DOI: 10.3892/etm.2021.10325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 05/24/2021] [Indexed: 11/10/2022] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury is a leading cause of heart disease and death. Decreasing the detrimental effect of I/R remains an urgent issue in clinical practice. The present study examined the interaction of the anesthetics (sevoflurane and propofol), ADAM8, and microRNA (miR)-221-5p in myocardial tissue protection in the hypoxia-reoxygenation (H/R) model. H9C2 cells were cultured and subjected to H/R stimulation for further verifications in vitro. Reverse transcription-quantitative PCR or western blotting was performed to evaluate mRNA or protein expression levels. Cell Counting Kit-8, BrdU, and caspase-3 activity assays were performed to investigate cell viability, proliferation and apoptosis. A dual-luciferase reporter assay was performed to verify the association between miR-221-5p and ADAM8. Sevoflurane had greater protective effects on the life of cardiomyocytes with H/R injury compared with propofol by promoting cell viability, proliferation and inhibiting apoptosis. Concurrently, compared with propofol-treated H/R injured cardiomyocytes, the expression level of ADAM8 in sevoflurane-treated H/R injured cardiomyocytes was higher. In addition, overexpression of ADAM8 promoted the cell viability and proliferation of sevoflurane-treated cardiomyocytes with H/R injury but inhibited cell apoptosis, while the downregulation of miR-221-5p showed an opposite trend to that of ADAM8 overexpression. The present data provide evidence that sevoflurane can mediate the miR-221-5p/ADAM8 axis, playing a better protective role compared with propofol in cardiomyocytes with H/R injury.
Collapse
Affiliation(s)
- Dan Xie
- Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Huifei Deng
- Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Hao Feng
- Department of Neurosurgery, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
23
|
Yu J, Xu C, Lee JS, Alder JK, Wen Z, Wang G, Gil Silva AA, Sanchez PG, Pilewsky JM, McDyer JF, Wang X. Rapid postmortem ventilation improves donor lung viability by extending the tolerable warm ischemic time after cardiac death in mice. Am J Physiol Lung Cell Mol Physiol 2021; 321:L653-L662. [PMID: 34318693 DOI: 10.1152/ajplung.00011.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Uncontrolled donation after cardiac death (uDCD) contributes little to ameliorating donor lung shortage due to rapidly progressive warm ischemia after circulatory arrest. Here, we demonstrated non-hypoxia improves donor lung viability in a novel uDCD lung transplant model undergoing rapid ventilation after cardiac death and compared the evolution of ischemia-reperfusion injury in mice that underwent pulmonary artery ligation (PAL). The tolerable warm ischemia time at 37ºC was initially determined in mice using a modified PAL model. The donor lung following PAL was also transplanted into syngeneic mice and compared to those that underwent rapid ventilation or no ventilation at 37ºC prior to transplantation. Twenty-four hours following reperfusion, lung histology, PaO2/FIO2 ratio, and inflammatory mediators were measured. Four hours of PAL had little impact on PaO2/FIO2 ratio and acute lung injury score in contrast to significant injury induced by 5 hours of PAL. Four-hour PAL lungs showed an early myeloid-dominant inflammatory signature when compared to naïve lungs and substantially injured five-hour PAL lungs. In the context of transplantation, unventilated donor lungs showed severe injury after reperfusion, whereas ventilated donor lungs showed minimal changes in PaO2/FIO2 ratio, histologic score, and expression of inflammatory markers. Taken together, the tolerable warm ischemia time of murine lungs at 37oC can be extended by maintaining alveolar ventilation for up to 4 hours. Non-hypoxic lung warm ischemia-reperfusion injury shows an early transcriptional signature of myeloid cell recruitment and extracellular matrix proteolysis prior to blood-gas barrier dysfunction and significant tissue damage.
Collapse
Affiliation(s)
- Junyi Yu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, United States.,Hand and Microsurgery Department, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Che Xu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Janet S Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jonathan K Alder
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, United States
| | - Zongmei Wen
- Department of Anesthesia, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guifang Wang
- Department of Respiratory Medicine, Huashan Hospital,Fudan University School of Medicine, Shanghai, China
| | - Agustin Alejandro Gil Silva
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, United States
| | - Pablo G Sanchez
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph M Pilewsky
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, United States
| | - John F McDyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, United States.,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Xingan Wang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, PA, United States.,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
24
|
Sevoflurane protects against ischemia-reperfusion injury in mice after total knee arthroplasty via facilitating RASD1-mediated protein kinase A pathway activation. Aging (Albany NY) 2021; 13:13333-13348. [PMID: 33982674 PMCID: PMC8148473 DOI: 10.18632/aging.103899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/21/2020] [Indexed: 12/12/2022]
Abstract
This study aimed to explore effects of Sevoflurane on ischemia-reperfusion (I/R) injury after total knee arthroplasty (TKA). To explore potential molecular mechanism, Ras related dexamethasone induced 1 (RASD1), a Protein kinase A (PKA) activator, frequently associated with various models of I/R injury, was also investigated. In vivo mouse models with I/R injury after TKA and in vitro cell models with I/R injury were induced. Contents of creatinine kinase (CK), lactic dehydrogenase (LDH), superoxide dismutase (SOD), and malondialdehyde (MDA), serum levels of inflammatory factors, expression of PKA pathway-related genes and cell proliferation and apoptosis were measured. RASD1 was altered and PKA pathway was inhibited in mice and cells to elucidate the involvement of RASD1 and PKA pathway in Sevoflurane treatment on I/R injury. RASD1 was upregulated in I/R injury after TKA. Sevoflurane treatment or silencing RASD1 reduced RASD1 expression, CK, LDH and MDA contents, inflammation, apoptosis, but increased proliferation, SOD content, cAMP expression, and extents of PKA and cAMP responsive element binding protein (CREB) phosphorylation in skeletal muscle cells of I/R injury. Additionally, PKA pathway activation potentiated the therapeutic effect of Sevoflurane on I/R injury after TKA. Altogether, Sevoflurane treatment confines I/R injury after TKA via RASD1-mediated PKA pathway activation.
Collapse
|
25
|
Ma H, Yang B, Yu L, Gao Y, Ye X, Liu Y, Li Z, Li H, Li E. Sevoflurane protects the liver from ischemia-reperfusion injury by regulating Nrf2/HO-1 pathway. Eur J Pharmacol 2021; 898:173932. [PMID: 33631180 DOI: 10.1016/j.ejphar.2021.173932] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023]
Abstract
We aimed to investigate the role and mechanism of sevoflurane (SEV) preconditioning in liver ischemia-reperfusion (I/R) injury. In vivo, rats were randomly divided into Sham group, I/R rat model group, I/R + SEV group and SEV group. In vitro, hypoxia-reoxygenation (H/R) cell model were established. Hematoxylin-Eosin (H&E) and TUNEL assay were used to evaluate the degree of tissue damage and detect apoptosis in rats, respectively. HO-1, nuclear Nrf2 and cytosolic Nrf2 expressions were detected by immunohistochemical staining, Western blot analysis and quantitative real-time PCR (qRT-PCR), respectively. Contents of Lactate dehydrogenase (LDH), malondialdehyde (MDA), and reactive oxygen species (ROS) were determined by corresponding kits. Inflammatory factor levels, cell viability, apoptosis were detected by enzyme-linked immunosorbent assay (ELISA), MTT assay, and flow cytometry, respectively.In the I/R group, liver damage was severe, apoptosis-positive cells were increased, HO-1 and nuclear Nrf2 expressions were increased, and cytosolic Nrf2 expression was decreased. After SEV pretreatment, the degree of liver injury and apoptosis in rats were significantly reduced, HO-1 and nuclear Nrf2 expressions were increased significantly, and cytosolic Nrf2 expression was decreased. 4% SEV had the best mitigating effect on H/R-induced liver cell damage, as evidenced by reduced contents of LDH and MDA, decreased inflammatory factors, a lowered apoptosis rate, inhibited ROS production, effectively promoted Nrf2 nucleation, and activated Nrf/HO-1 pathway. ML385 pretreatment significantly inhibited the effect of SEV on hepatocytes.Sevoflurane protects the liver from ischemia-reperfusion injury by regulating the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Hongyan Ma
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China
| | - Baoyi Yang
- Department of Neursurgery, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, No.26, Heping Road, Dongli District, Harbin, Heilongjiang, 150040, China
| | - Lu Yu
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China
| | - Yang Gao
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China
| | - Xiangmei Ye
- Laboratory of Hemooncology, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China
| | - Ying Liu
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China
| | - Zhengtian Li
- Department of Tumor Endoscopic Surgery, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University, No.194, Xuefu Road, Harbin, Heilongjiang, 150001, China
| | - Enyou Li
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, No.23, Youzheng Street, Nangang District, Harbin, Heilongjiang, 150001, China.
| |
Collapse
|
26
|
Xu L, Ge F, Hu Y, Yu Y, Guo K, Miao C. Sevoflurane Postconditioning Attenuates Hepatic Ischemia-Reperfusion Injury by Limiting HMGB1/TLR4/NF-κB Pathway via Modulating microRNA-142 in vivo and in vitro. Front Pharmacol 2021; 12:646307. [PMID: 33935744 PMCID: PMC8085516 DOI: 10.3389/fphar.2021.646307] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Preconditioning of sevoflurane (Sevo) has been demonstrated to protect the liver from ischemia/reperfusion (I/R) injury. However, it is unknown whether it has hepatoprotective when given at the onset of reperfusion (postconditioning), a protocol with more clinical impact. The present study aimed to explore the hepatoprotective effects of Sevo postconditioning against hepatic IR injury in vivo and in vitro and the possible mechanisms. Using a mouse model of hepatic I/R, Sevo postconditioning significantly improved hepatic injury after reperfusion, as demonstrated by reduced AST, ALT, and LDH serum levels and reduced histologic damage in liver tissues. Furthermore, Sevo postconditioning could suppress the apoptosis, inhibit oxidative stress and inflammatory response in liver tissue of HIRI mice, as well as improve the survival rate of HIRI mice. Through analyzing GSE72314 from the gene expression omnibus (GEO) database, it was demonstrated that microRNA (miR)-142 is downregulated by HIRI, which was reversed by Sevo treatment. Further investigation showed that agomiR-142 injection could enhance the hepatoprotective effects of Sevo postconditioning on I/R injury, while antagomiR-142 reversed these effects in mice. Notably, high mobility group box 1 (HMGB1), an important inflammatory factor, was directly targeted by miR-142 in hepatic cells, and we further found that Sevo could inhibit the expression of HMGB1 through up-regulating miR-142 expression in HIRI mice model. In addition, we found that I/R injury induced the activation of TLR4/NF-κB inflammatory pathway was partially suppressed by Sevo postconditioning, and miR-142 mediated the regulatory role of Sevo postconditioning. In line with the in vivo results, Sevo treatment improved the cell viability, inhibited cell apoptosis, oxidative stress and inflammatory response in vitro HIRI model, while these effects were reversed by antagomiR-142 transfection. Collectively, our findings demonstrated that Sevo postconditioning counteracts the downregulation of miR-142 provoked by I/R, in turn decreased the expression of HMGB1, blocking TLR4/NF-κB pathway activation, thus improving hepatic I/R injury. Our data suggest that Sevo may be a valuable alternative anaesthetic agent in liver transplantation and major liver surgeries.
Collapse
Affiliation(s)
- Liying Xu
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Feng Ge
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yan Hu
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Ying Yu
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
27
|
Liu X, Wang L, Xing Q, Li K, Si J, Ma X, Mao L. Sevoflurane inhibits ferroptosis: A new mechanism to explain its protective role against lipopolysaccharide-induced acute lung injury. Life Sci 2021; 275:119391. [PMID: 33774026 DOI: 10.1016/j.lfs.2021.119391] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 12/22/2022]
Abstract
Sevoflurane (Sev) has protective effects in acute lung injury (ALI), but the relevant mechanisms are still not fully understood. The present study aimed to determine whether Sev exerts a protective effect on lipopolysaccharide (LPS)-induced ALI by regulating ferroptosis. In this study, we found that Sev could protect mice from lung injury caused by LPS stimulation, including extenuating lung histological damage, pulmonary edema and pulmonary vascular permeability, and the content of inflammatory factors in Bronchoalveolar lavage fluid (BALF), as well as improving the survival rate of ALI mice, which was in line with the effects of ferroptosis inhibitor ferrostatin-1. Simultaneously, Sev could eliminate the worsening effects of ferroptosis inducer Fe-citrate on LPS-induced ALI to a certain extent. Additionally, the administration of Sev could inhibit ferroptosis caused by LPS, which was manifested by reducing the accumulation of MDA and Fe2+, and increasing the levels of GSH and GPX4 in the lung tissues of ALI mice. It was also observed in BEAS-2B cells that the increased MDA and Fe2+ levels and the decreased GSH and GPX4 levels caused by LPS could be rescued by ferrostatin-1 and Sev. LPS stimulation compensatory up-regulated heme oxygenase-1 (HO-1) expression in mouse lung tissues and BEAS-2B cells, which could be enhanced by Sev. Moreover, HO-1 depletion could offset the inhibitory effect of Sev on LPS-induced ferroptosis and inflammation in BEAS-2B cells. Taken together, Sev inhibited ferroptosis by up-regulating HO-1 expression to reduce LPS-induced ALI, which may provide a possible mechanism for the application of Sev in clinical anesthesia.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003
| | - Ling Wang
- Department of Anesthesiology, No. 989 Hospital of Joint Logistic Support Force of the Chinese People's Liberation Army, Luoyang 471003, Henan Province, China.
| | - Qunzhi Xing
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003
| | - Kehan Li
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003
| | - Jianluo Si
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003
| | - Xiaowu Ma
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003
| | - Lianjing Mao
- Department of Anesthesiology, No. 989 Hospital of Joint Logistic Support Force of the Chinese People's Liberation Army, Luoyang 471003, Henan Province, China
| |
Collapse
|
28
|
Nieuwenhuijs-Moeke GJ, Bosch DJ, Leuvenink HG. Molecular Aspects of Volatile Anesthetic-Induced Organ Protection and Its Potential in Kidney Transplantation. Int J Mol Sci 2021; 22:ijms22052727. [PMID: 33800423 PMCID: PMC7962839 DOI: 10.3390/ijms22052727] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
Ischemia reperfusion injury (IRI) is inevitable in kidney transplantation and negatively impacts graft and patient outcome. Reperfusion takes place in the recipient and most of the injury following ischemia and reperfusion occurs during this reperfusion phase; therefore, the intra-operative period seems an attractive window of opportunity to modulate IRI and improve short- and potentially long-term graft outcome. Commonly used volatile anesthetics such as sevoflurane and isoflurane have been shown to interfere with many of the pathophysiological processes involved in the injurious cascade of IRI. Therefore, volatile anesthetic (VA) agents might be the preferred anesthetics used during the transplantation procedure. This review highlights the molecular and cellular protective points of engagement of VA shown in in vitro studies and in vivo animal experiments, and the potential translation of these results to the clinical setting of kidney transplantation.
Collapse
Affiliation(s)
- Gertrude J. Nieuwenhuijs-Moeke
- Department of Anesthesiology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
- Correspondence: ; Tel.: +31-631623075
| | - Dirk J. Bosch
- Department of Anesthesiology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
| | - Henri G.D. Leuvenink
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
| |
Collapse
|
29
|
Bertani A, Miceli V, De Monte L, Occhipinti G, Pagano V, Liotta R, Badami E, Tuzzolino F, Arcadipane A. Donor Preconditioning with Inhaled Sevoflurane Mitigates the Effects of Ischemia-Reperfusion Injury in a Swine Model of Lung Transplantation. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6625955. [PMID: 33506025 PMCID: PMC7815409 DOI: 10.1155/2021/6625955] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/18/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
Primary graft dysfunction (PGD) and ischemia-reperfusion injury (IRI) occur in up to 30% of patients undergoing lung transplantation and may impact on the clinical outcome. Several strategies for the prevention and treatment of PGD have been proposed, but with limited use in clinical practice. In this study, we investigate the potential application of sevoflurane (SEV) preconditioning to mitigate IRI after lung transplantation. The study included two groups of swines (preconditioned and not preconditioned with SEV) undergoing left lung transplantation after 24-hour of cold ischemia. Recipients' data was collected for 6 hours after reperfusion. Outcome analysis included assessment of ventilatory, hemodynamic, and hemogasanalytic parameters, evaluation of cellularity and cytokines in BAL samples, and histological analysis of tissue samples. Hemogasanalytic, hemodynamic, and respiratory parameters were significantly favorable, and the histological score showed less inflammatory and fibrotic injury in animals receiving SEV treatment. BAL cellular and cytokine profiling showed an anti-inflammatory pattern in animals receiving SEV compared to controls. In a swine model of lung transplantation after prolonged cold ischemia, SEV showed to mitigate the adverse effects of ischemia/reperfusion and to improve animal survival. Given the low cost and easy applicability, the administration of SEV in lung donors may be more extensively explored in clinical practice.
Collapse
Affiliation(s)
- Alessandro Bertani
- Division of Thoracic Surgery and Lung Transplantation, Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, IRCCS-ISMETT, Palermo, Italy
| | | | - Lavinia De Monte
- Division of Thoracic Surgery and Lung Transplantation, Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, IRCCS-ISMETT, Palermo, Italy
| | - Giovanna Occhipinti
- Department of Anesthesiology and Critical Care, IRCCS-ISMETT, Palermo, Italy
| | | | - Rosa Liotta
- Department of Pathology, IRCCS-ISMETT, Palermo, Italy
| | - Ester Badami
- Fondazione Ri.MED, Palermo, Italy
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT, Palermo, Italy
| | | | - Antonio Arcadipane
- Department of Anesthesiology and Critical Care, IRCCS-ISMETT, Palermo, Italy
| |
Collapse
|
30
|
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: 10] [Impact Index Per Article: 2.0] [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
|
31
|
Zhang J, Yu P, Hua F, Hu Y, Xiao F, Liu Q, Huang D, Deng F, Wei G, Deng W, Ma J, Zhu W, Zhang J, Yu S. Sevoflurane postconditioning reduces myocardial ischemia reperfusion injury-induced necroptosis by up-regulation of OGT-mediated O-GlcNAcylated RIPK3. Aging (Albany NY) 2020; 12:25452-25468. [PMID: 33231560 PMCID: PMC7803485 DOI: 10.18632/aging.104146] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/15/2020] [Indexed: 12/13/2022]
Abstract
Inhalation anesthetics have been demonstrated to have protective effects against myocardial ischemia reperfusion injury (MIRI). O-linked GlcNAcylation (O-GlcNAc) modifications have been shown to protect against MIRI. This study aimed to investigate whether O-GlcNAcylation and necroptosis signaling were important for sevoflurane postconditioning (SPC) induced cardioprotective effects. Apart from rats in the SHAM and sevoflurane (SEVO) group, rats underwent 30 min ischemia followed by 2 h reperfusion. Cardiac hemodynamics and function were determined. In addition, myocardial infarction size, cardiac function parameters, myocardial lactic dehydrogenase (LDH) content, myocardium histopathological changes, necrotic myocardium, O-GlcNAcylation, and protein expression levels of necroptosis biomarkers were measured, together with co-immunoprecipitation experiments using proteins associated with the necroptosis pathway and O-GlcNAcylation. SPC reduced myocardial infarction size, ameliorated cardiac function, restored hemodynamic performance, improved histopathological changes, and reduced receptor-interacting protein kinase 1 (RIPK1)/receptor-interacting protein kinase 3 (RIPK3)/mixed lineage kinase domain-like (MLKL) mediated necroptosis. In addition, SPC up-regulated O-GlcNAc transferase (OGT) mediated O-GlcNAcylation, increased O-GlcNAcylated RIPK3, and inhibited the association of RIPK3 and MLKL. However, OSMI-1, an OGT inhibitor, abolished SPC mediated cardioprotective effects and inhibited OGT mediated up-regulation of O-GlcNAcylation and down-regulation of RIPK3 and MLKL proteins induced by SPC. Our study demonstrated that SPC restrained MIRI induced necroptosis via regulating OGT mediated O-GlcNAcylation of RIPK3 and lessening the formulation of RIPK3/MLKL complex.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Anesthesiology, The Second Affiliate Hospital of Nanchang University, Nanchang 330006, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliate Hospital of Nanchang University, Nanchang 330006, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliate Hospital of Nanchang University, Nanchang 330006, China
| | - Yanhui Hu
- Department of Anesthesiology, The Second Affiliate Hospital of Nanchang University, Nanchang 330006, China
| | - Fan Xiao
- Department of Anesthesiology, The Second Affiliate Hospital of Nanchang University, Nanchang 330006, China
| | - Qin Liu
- Department of Anesthesiology, The Second Affiliate Hospital of Nanchang University, Nanchang 330006, China
| | - Dan Huang
- Department of Anesthesiology, The Second Affiliate Hospital of Nanchang University, Nanchang 330006, China
| | - Fumou Deng
- Department of Anesthesiology, The Second Affiliate Hospital of Nanchang University, Nanchang 330006, China
| | - Gen Wei
- Department of Anesthesiology, The Second Affiliate Hospital of Nanchang University, Nanchang 330006, China
| | - Wei Deng
- Department of Anesthesiology, The Second Affiliate Hospital of Nanchang University, Nanchang 330006, China
| | - Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Wengen Zhu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Jiru Zhang
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University (The Fourth People's Hospital in Wuxi City), Wuxi 214000, China
| | - Shuchun Yu
- Department of Anesthesiology, The Second Affiliate Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
32
|
Tchana-Sato V, Ledoux D, Detry O, Hans G, Ancion A, D'Orio V, Massion PB, Amabili P, Bruls S, Lavigne JP, Monard J, Delbouille MH, Sakalihasan N, Defraigne JO. Successful clinical transplantation of hearts donated after circulatory death using normothermic regional perfusion. J Heart Lung Transplant 2020; 38:593-598. [PMID: 31128600 DOI: 10.1016/j.healun.2019.02.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Heart transplantation (HT) from donation after circulatory death (DCD) has yet to achieve wide clinical application despite the encouraging resultsreported recently. In this study we describe 2 cases of successful adult DCD HT performed at our institution using an original protocol. METHODS Our local abdominal DCD protocol was updated to allow DCD heart procurement, and was accepted by the institutional ethics committee. The main features of the protocol include: pre-mortem insertion of peripheral venoarterial extracorporeal membrane oxygenation cannulas; thoracoabdominal normothermic regional perfusion (NRP) by clamping the 3 aortic arch vessels to exclude cerebral circulation; and in-situ heart resuscitation. The retrieved hearts were directly transplanted into recipients located in an adjoining operating room. RESULTS The procurement warm ischemic time was 25 minutes for the first donor, and 26 minutes for the second donor. The cold ischemic time was 16 minutes for the first recipient and 17 minutes for the second recipient. The suture time was 30 minutes for the first recipient, and 53 minutes for the second recipient. Both recipients were easily weaned off cardiopulmonary bypass in sinus rhythm and inotropic support. Post-operative evaluation of cardiac function was excellent and the patients were subsequently discharged home. CONCLUSIONS Transplantation of hearts from DCD donors is now a clinical reality.NRP is a useful tool for resuscitation, reperfusion, and preservation of transplanted hearts. It also offers the opportunity to assess the function and viability of organs before transplantation. However,due to ethical issues, some may object to ante-mortem intervention.
Collapse
Affiliation(s)
| | | | - Olivier Detry
- Abdominal Surgery and Transplantation, Liege, Belgium
| | - Gregory Hans
- Anesthesiology and Intensive Care, Liege, Belgium
| | | | | | | | | | - Samuel Bruls
- Departments ofCardiothoracic Surgery, Liege, Belgium
| | | | - Josée Monard
- Abdominal Surgery and Transplantation, Liege, Belgium
| | | | | | | |
Collapse
|
33
|
Zhou W, Shao W, Zhang Y, Liu D, Liu M, Jin T. Glucagon-like peptide-1 receptor mediates the beneficial effect of liraglutide in an acute lung injury mouse model involving the thioredoxin-interacting protein. Am J Physiol Endocrinol Metab 2020; 319:E568-E578. [PMID: 32723174 PMCID: PMC7839242 DOI: 10.1152/ajpendo.00292.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Repurposing clinically used drugs is among the important strategies in drug discovery. Glucagon-like peptide-1 (GLP-1) and its diabetes-based drugs, such as liraglutide, possess a spectrum of extra-pancreatic functions, while GLP-1 receptor (GLP-1R) is most abundantly expressed in the lung. Recent studies have suggested that GLP-1-based drugs exert beneficial effects in chronic, as well as acute, lung injury rodent models. Here, we show that liraglutide pretreatment reduced LPS induced acute lung injury in mice. It significantly reduced lung injury score, wet/dry lung weight ratio, bronchoalveolar lavage fluid immune cell count and protein concentration, and cell apoptosis in the lung, and it was associated with reduced lung inflammatory cytokine and chemokine gene expression. Importantly, these effects were virtually absent in GLP-1R-/- mice. A well-known function of GLP-1 and GLP-based drugs in pancreatic β-cells is the attenuation of high-glucose stimulated expression of thioredoxin-interacting protein (TxNIP), a key component of inflammasome. LPS-challenged lungs showed elevated TxNIP mRNA and protein expression, which was attenuated by liraglutide treatment in a GLP-1R-dependent manner. Hence, our observations suggest that GLP-1R is essential in mediating beneficial effects of liraglutide in acute lung injury, with the inflammasome component TxNIP as a potential target.
Collapse
Affiliation(s)
- Wenyong Zhou
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weijuan Shao
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yu Zhang
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Dinghui Liu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Mingyao Liu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tianru Jin
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
34
|
Zheng Y, Lu H, Huang H. Desflurane Preconditioning Protects Against Renal Ischemia-Reperfusion Injury and Inhibits Inflammation and Oxidative Stress in Rats Through Regulating the Nrf2-Keap1-ARE Signaling Pathway. Drug Des Devel Ther 2020; 14:1351-1362. [PMID: 32308368 PMCID: PMC7138619 DOI: 10.2147/dddt.s223742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 03/16/2020] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Kidney is sensitive to ischemia-reperfusion (I/R) injury because of its special structure and function. In this study, we aimed to explore the mechanism of desflurane (DFE) preconditioning effecting on renal I/R injury in rats. METHODS Renal I/R injury rats model was constructed, and the expressions of serum renal function parameters (blood urea nitrogen (BUN) and serum creatinine (SCr)) and lipid peroxidation-related factors were detected using corresponding commercial kits to assess the degrees of renal functional damage and oxidative stress. Hematoxylin--eosin (HE) staining and Masson trichrome staining were applied to measure the renal histologic damage. The expressions of inflammation-related factors were determined by ELISA assay. The cell apoptosis was analyzed using TUNEL, Western blot and immunohistochemistry (IHC). IHC was also used to detect the number of myeloperoxidase (MPO)-positive cells. The expressions of proteins associated with the Nrf2-Keap1-ARE pathway were assessed by Western blot and IHC. RESULTS DFE preconditioning inhibited I/R injury-induced BUN and SCr increase and renal histologic injury in rats. Also, DFE suppressed the inflammation, apoptosis and oxidative stress caused by renal I/R injury in vivo. In addition, DFE preconditioning repressed peroxide-related factors (MDA, MPO and NO) expressions and promoted antioxidant-related factors (GSH, SOD, GPx and CAT) expressions. In addition, DFE promoted Nrf2-Keap1-ARE-related proteins including Nrf2, NQO1, HO-1, γ-GCS, GSR and GCLc expressions. CONCLUSION DFE preconditioning protected the kidney as well as inhibited the inflammation, cell apoptosis and oxidative stress in renal I/R injury rats by activating the Nrf2-Keap1-ARE signaling pathway.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Anesthesiology, Xiamen Haicang Hospital, Xiamen361000, People’s Republic of China
| | - Hui Lu
- Department of Anesthesiology, Xiamen Haicang Hospital, Xiamen361000, People’s Republic of China
| | - Huiqiong Huang
- Department of Anesthesiology, Women and Children’s Hospital Affiliated to Xiamen University, Xiamen361000, People’s Republic of China
| |
Collapse
|
35
|
Qiu S, Liu B, Mo Y, Wang X, Zhong L, Han X, Mi F. MicroRNA-153-3p increases autophagy in sevoflurane-preconditioned mice to protect against ischaemic/reperfusion injury after knee arthroplasty. J Cell Mol Med 2020; 24:5330-5340. [PMID: 32239627 PMCID: PMC7205820 DOI: 10.1111/jcmm.15188] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 01/16/2020] [Accepted: 03/01/2020] [Indexed: 12/17/2022] Open
Abstract
The use of tourniquet during total knee arthroplasty (TKA) can result in ischaemia/reperfusion injury (IRI). Of interest, microRNAs (miRs) are reported to be involved in various kinds of IRI due to their ability in modulating autophagy. Therefore, the study aimed to investigate the effect of miR‐153‐3p on autophagy in IRI in vitro and in vivo under sevoflurane preconditioning. In the in vitro model, chondrocytes from naive mice were treated with 0% FBS alone or in combination with sevoflurane. Additionally, in vivo assays were conducted in mouse models with tourniquet‐induced IRI after TKA under or without sevoflurane preconditioning. The pathological observation in vivo validated that sevoflurane preconditioning protected the knee joint against IRI. Moreover, miR‐153‐3p expression was diminished in chondrocytes of the in vitro model and in cartilage tissue of the in vivo model, but its expression was appreciably up‐regulated in the presence of sevoflurane preconditioning. Mechanistic study showed that miR‐153‐3p disrupted the interaction between Bcl‐2 and Beclin1 by targeting Bcl‐2, thereby facilitating autophagy in chondrocytes under sevoflurane preconditioning. Furthermore, the experiments in human chondrocytes also verified the protective effects of miR‐153‐3p against IRI were realized through inhibiting Bcl‐2. Collectively, miR‐153‐3p overexpression blocks the interaction between Bcl‐2 and Beclin1 via down‐regulation of Bcl‐2 to promote autophagy of chondrocytes, thus protecting knee joint against IRI after TKA under sevoflurane preconditioning.
Collapse
Affiliation(s)
- Shuang Qiu
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| | - Benjuan Liu
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| | - Yanshuai Mo
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| | - Xueqin Wang
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| | - Lina Zhong
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| | - Xiao Han
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| | - Fuli Mi
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| |
Collapse
|
36
|
Ohsumi A, Kanou T, Ali A, Guan Z, Hwang DM, Waddell TK, Juvet S, Liu M, Keshavjee S, Cypel M. A method for translational rat ex vivo lung perfusion experimentation. Am J Physiol Lung Cell Mol Physiol 2020; 319:L61-L70. [PMID: 32233924 DOI: 10.1152/ajplung.00256.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The application of ex vivo lung perfusion (EVLP) has significantly increased the successful clinical use of marginal donor lungs. While large animal EVLP models exist to test new strategies to improve organ repair, there is currently no rat EVLP model capable of maintaining long-term lung viability. Here, we describe a new rat EVLP model that addresses this need, while enabling the study of lung injury due to cold ischemic time (CIT). The technique involves perfusing and ventilating male Lewis rat donor lungs for 4 h before transplanting the left lung into a recipient rat and then evaluating lung function 2 h after reperfusion. To test injury within this model, lungs were divided into groups and exposed to different CITs (i.e., 20 min, 6 h, 12 h, 18 h and 24 h). Experiments involving the 24-h-CIT group were prematurely terminated due to the development of severe edema. For the other groups, no differences in the ratio of arterial oxygen partial pressure to fractional inspired oxygen ([Formula: see text]/[Formula: see text]) were observed during EVLP; however, lung compliance decreased over time in the 18-h group (P = 0.012) and the [Formula: see text]/[Formula: see text] of the blood from the left pulmonary vein 2 h after transplantation was lower compared with 20-min-CIT group (P = 0.0062). This new model maintained stable lung function during 4-h EVLP and after transplantation when exposed to up to 12 h of CIT.
Collapse
Affiliation(s)
- Akihiro Ohsumi
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada.,Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Takashi Kanou
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Aadil Ali
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Zehong Guan
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - David M Hwang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Thomas K Waddell
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Stephen Juvet
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Ex-vivo lung perfusion (EVLP) has been developed to expand the donor pool for lung transplantation recipients. The role of EVLP in organ preservation, evaluation and potential reconditioning is reviewed. RECENT FINDINGS EVLP has been shown to significantly increase the utilization of donor lungs for transplantation. Evidence suggests that patient outcomes from EVLP lungs are comparable to standard procurement technique. Novel strategies are being developed to treat and recondition injured donor lungs. EVLP may also prove to be a tool for translational research of lung diseases. SUMMARY EVLP has been shown to be an effective system to expand donor pool for lung transplantation without detriment to recipients. Future potential ex-vivo developments may further improve patient outcomes as well as increasing availability of donor organs.
Collapse
|
38
|
Abstract
In this paper we will describe anaesthetic management of solid organ and reconstructive transplantation (RT) patients. We will focus on similar underlying principles of reperfusion, ischaemic-reperfusion injury, preconditioning and extracorporeal donor organ preservation. Special concerns for anaesthetic management of these patients need to focus on pre-assessment, pre-operative optimisation, vascular access, fluid management, blood and products replacement, cardiovascular monitoring, use of inotropes and vasoconstrictors, maintaining electrolyte balance and regional anaesthesia. Despite the complexity and long duration of transplant procedures, fast-tracking to the surgical ward after transplantation is becoming more popular and its benefits are well recognised.
Collapse
Affiliation(s)
- Zoka Milan
- Anaesthetic Department, King's College Hospital, Denmark Hill, SE5 9RS London, UK.
| | - Miriam Cortes
- Surgical Department, King's College Hospital, London, UK
| | | |
Collapse
|
39
|
Wang Y, Zhang X, Tian J, Liu G, Li X, Shen D. Sevoflurane alleviates LPS‑induced acute lung injury via the microRNA‑27a‑3p/TLR4/MyD88/NF‑κB signaling pathway. Int J Mol Med 2019; 44:479-490. [PMID: 31173183 PMCID: PMC6605322 DOI: 10.3892/ijmm.2019.4217] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 05/24/2019] [Indexed: 02/07/2023] Open
Abstract
Acute lung injury (ALI) is a critical syndrome that is associated with a high morbidity and mortality in patients. Sevoflurane has a lung protective effect in ALI as it reportedly has anti‑inflammatory and apoptotic‑regulating activity. However, the mechanism is still not entirely understood. The aim of the present study was to explore the effects of sevoflurane on lipopolysaccharide (LPS)‑induced ALI in mice and the possible mechanisms involved. The results revealed that sevoflurane treatment improved LPS‑induced lung injury, as evidenced by the reduction in mortality, lung permeability, lung wet/dry ratio and lung histopathological changes in mice. Total cell counts and the production of pro‑inflammatory cytokines [tumor necrosis factor‑α, interleukin (IL)‑1β and IL‑6] in bronchoalveolar fluid were also decreased following treatment with sevoflurane. Additionally, LPS‑triggered apoptosis in lung tissues, which was eliminated by sevoflurane. Furthermore, a miRCURY™ LNA array was employed to screen for differentially expressed microRNAs (miRs/miRNAs). Among these miRNAs, 6 were differentially expressed and were involved in the inflammatory response, but only miR‑27a‑3p (miR‑27a) was regulated by sevoflurane. Subsequently, the present study investigated whether sevoflurane exerts its function through the modulation of miR‑27a. The results demonstrated that the overexpression of miR‑27a via an injection with agomiR‑27a produced similar protections as sevoflurane, while the inhibition of miR‑27a suppressed the lung protective effects of sevoflurane in ALI mice. In addition, the present study identified that miR‑27a inhibited Toll‑like receptor 4 (TLR4) by binding to its 3'‑untranslated region. Western blot analysis demonstrated that sevoflurane may ameliorate the inflammatory response by blocking the miR‑27a/TLR4/MyD88/NF‑κB signaling pathway. The present results indicate that sevoflurane may be a viable therapeutic option in the treatment of patients with ALI.
Collapse
Affiliation(s)
- Yunfei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Xiaoran Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Jianmin Tian
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Guoze Liu
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Xiaofang Li
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Dan Shen
- Department of Anesthesiology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| |
Collapse
|
40
|
Hopper CP, Wollborn J. Delivery of carbon monoxide via halogenated ether anesthetics. Nitric Oxide 2019; 89:93-95. [PMID: 31125687 DOI: 10.1016/j.niox.2019.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 04/04/2019] [Accepted: 05/21/2019] [Indexed: 01/31/2023]
Affiliation(s)
- Christopher P Hopper
- Department of Medicinal Chemistry, College of Pharmacy, The University of Florida, Gainesville, Florida, USA; Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Germany; Institute for Experimental Biomedicine, University Hospital Wuerzburg, Germany.
| | - Jakob Wollborn
- Department of Anesthesiology and Critical Care, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
41
|
Zhao YB, Zhao J, Zhang LJ, Shan RG, Sun ZZ, Wang K, Chen JQ, Mu JX. MicroRNA-370 protects against myocardial ischemia/reperfusion injury in mice following sevoflurane anesthetic preconditioning through PLIN5-dependent PPAR signaling pathway. Biomed Pharmacother 2019; 113:108697. [PMID: 30856533 DOI: 10.1016/j.biopha.2019.108697] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/16/2022] Open
Abstract
Myocardial ischemia/reperfusion injury (IRI) has long been identified to be a contributor to adverse cardiovascular outcomes following myocardial ischemia, cardiac surgery or circulatory arrest. This study aims to investigate the effects of microRNA (miR-370) targeting perilipin-5 (PLIN5) in mice following sevoflurane anesthetic preconditioning (SAP). A mouse model of left ventricular myocardial IRI was established, followed by the evaluation of myocardial infarction size and cardiac function to determine the effects of SAP. The underlying regulatory mechanisms of miR-370 were analyzed in concert with the treatment of miR-370 mimic, miR-370 inhibitor, or siRNA against PLIN5 in cardiomyocytes isolated from mice with IRI. Also, cardiomyocyte proliferation, cell cycle distribution and apoptosis were evaluated following treatment. Lastly, SAP-treated I/R mice were injected with miR-370 inhibitor to verify the mechanism of SAP. The use of SAP conferred cardioprotective effects on myocardial IRI. MiR-370 was downregulated in mice that exhibited IRI, but SAP elevated the miR-370 expression. Functionally, miR-370 negatively targeted PLIN5 and activated the peroxisome proliferator activated-receptor (PPAR) signaling pathway, leading to decreased PPARγ expression but increased PPARα expression. The results also showed that elevation of miR-370 or the silencing of PLIN5 promoted cardiomyocyte proliferation. miR-370 also inhibited cardiomyocyte apoptosis as reflected by decreased caspase-3 expression and increased Bcl-2 expression. Additionally, SAP also alleviated I/R injury by inhibiting PPARγ. This study demonstrates that SAP induces miR-370 and exerts cardioprotective effects on myocardial IRI, where upregulation of miR-370 alleviates myocardial IRI via inhibiting the PLIN5-dependent PPAR signaling pathway.
Collapse
Affiliation(s)
- Yan-Bin Zhao
- Department of Anesthesiology, the First Hospital of the PLA, Lanzhou 730031, PR China
| | - Juan Zhao
- Department of Anesthesiology, the First Hospital of Lanzhou University, Donggang Courtyard Area, Lanzhou 730030, PR China
| | - Li-Jun Zhang
- Department of Anesthesiology, Lanzhou Time Laser Cosmetic Surgery Hospital, Lanzhou 730030, PR China
| | - Run-Gang Shan
- Department of Anesthesiology, the First Hospital of the PLA, Lanzhou 730031, PR China
| | - Zhen-Zhong Sun
- Department of Anesthesiology, Guangdong Armed Police Corps Hospital, Guangzhou 510507, PR China
| | - Kai Wang
- Department of Anesthesiology, the First Hospital of the PLA, Lanzhou 730031, PR China
| | - Jin-Quan Chen
- Department of Anesthesiology, the First People's Hospital of Xianyang City, Xianyang 712000, PR China.
| | - Ji-Xue Mu
- Lanzhou Minimally Invasive Orthopedic Hospital, Lanzhou 730050, PR China.
| |
Collapse
|
42
|
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: 0.3] [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
|
43
|
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: 0.4] [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
|
44
|
Pang QY, An R, Liu HL. Effects of inhalation and intravenous anesthesia on intraoperative cardiopulmonary function and postoperative complications in patients undergoing thoracic surgery. Minerva Anestesiol 2018; 84:1287-1297. [DOI: 10.23736/s0375-9393.18.12501-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
Persson PB, Bondke Persson A. Oxygen-to little, too much or just right. Acta Physiol (Oxf) 2018; 223:e13076. [PMID: 29675842 DOI: 10.1111/apha.13076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- P B Persson
- Charité - Universitätsmedizin Berlin - corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Vegetative Physiology, Berlin, Germany
| | - A Bondke Persson
- Charité - Universitätsmedizin Berlin - corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
46
|
Chen S, Lotz C, Roewer N, Broscheit JA. Comparison of volatile anesthetic-induced preconditioning in cardiac and cerebral system: molecular mechanisms and clinical aspects. Eur J Med Res 2018; 23:10. [PMID: 29458412 PMCID: PMC5819224 DOI: 10.1186/s40001-018-0308-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 02/12/2018] [Indexed: 12/17/2022] Open
Abstract
Volatile anesthetic-induced preconditioning (APC) has shown to have cardiac and cerebral protective properties in both pre-clinical models and clinical trials. Interestingly, accumulating evidences demonstrate that, except from some specific characters, the underlying molecular mechanisms of APC-induced protective effects in myocytes and neurons are very similar; they share several major intracellular signaling pathways, including mediating mitochondrial function, release of inflammatory cytokines and cell apoptosis. Among all the experimental results, cortical spreading depolarization is a relative newly discovered cellular mechanism of APC, which, however, just exists in central nervous system. Applying volatile anesthetic preconditioning to clinical practice seems to be a promising cardio-and neuroprotective strategy. In this review, we also summarized and discussed the results of recent clinical research of APC. Despite all the positive experimental evidences, large-scale, long-term, more precisely controlled clinical trials focusing on the perioperative use of volatile anesthetics for organ protection are still needed.
Collapse
Affiliation(s)
- Shasha Chen
- Department of Anesthesiology and Critical Care, University of Wuerzburg, Oberduerrbacher Str.6, 97080, Wuerzburg, Germany.
| | - Christopher Lotz
- Department of Anesthesiology and Critical Care, University of Wuerzburg, Oberduerrbacher Str.6, 97080, Wuerzburg, Germany
| | - Norbert Roewer
- Department of Anesthesiology and Critical Care, University of Wuerzburg, Oberduerrbacher Str.6, 97080, Wuerzburg, Germany
| | - Jens-Albert Broscheit
- Department of Anesthesiology and Critical Care, University of Wuerzburg, Oberduerrbacher Str.6, 97080, Wuerzburg, Germany
| |
Collapse
|
47
|
Motayagheni N, Phan S, Eshraghi C, Nozari A, Atala A. A Review of Anesthetic Effects on Renal Function: Potential Organ Protection. Am J Nephrol 2017; 46:380-389. [PMID: 29131005 DOI: 10.1159/000482014] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Renal protection is a critical concept for anesthesiologists, nephrologists, and urologists, since anesthesia and renal function are highly interconnected and can potentially interfere with one another. Therefore, a comprehensive understanding of anesthetic drugs and their effects on renal function remains fundamental to the success of renal surgeries, especially transplant procedures. Some experimental studies have shown that some anesthetics provide protection against renal ischemia/reperfusion (IR) injury, but there is limited clinical evidence. SUMMARY The effects of anesthetic drugs on renal failure are particularly important in the context of kidney transplantation, since the conditions of preservation following removal profoundly influence the recovery of organ function. Currently, preservation procedures are typically based on the usage of a cold-storage solution. Some anesthetic drugs induce anti-inflammatory, anti-necrotic, and anti-apoptotic effects. A more thorough understanding of anesthetic effects on renal function can present a novel approach for developing organ-protective strategies. The aim of this review is to discuss the effects of different anesthetic drugs on renal function, with particular focus on IR injury. Many studies have demonstrated the organ-protective effects of some anesthetic drugs, specifically propofol, which indicate the potential of some anesthetics to introduce novel organ protective targets. This is not surprising, since lipid emulsions are major components of propofol, which accumulating data show provide organ protective effects against IR injury. Key Messages: Thorough understanding of the interaction between anesthetic drugs and renal function remains fundamental to the delivery of safe perioperative care and to optimizing outcomes after renal surgeries, particularly transplant procedures. Anesthetics can be repurposed for organ protection with more information about their effects, especially during transplant procedures. Here, we review the effects of different anesthetic drugs - specifically those that contain lipids in their structure, with special reference to IR injury.
Collapse
Affiliation(s)
- Negar Motayagheni
- Institute for Regenerative Medicine (Wake Forest Institute of Regenerative Medicine), Wake Forest School of Medicine Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - Sheshanna Phan
- Department of Anesthesiology, Division of Molecular Medicine, UCLA David Geffen School of Medicine, Los Angeles, California, USA
| | - Crystal Eshraghi
- Department of Anesthesiology, Division of Molecular Medicine, UCLA David Geffen School of Medicine, Los Angeles, California, USA
| | - Ala Nozari
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anthony Atala
- Institute of Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
48
|
Wu W, Wei N, Wang L, Kong D, Shao G, Qin Y, Wang L, Du Y. Sevoflurane preconditioning ameliorates traumatic spinal cord injury through caveolin-3-dependent cyclooxygenase-2 inhibition. Oncotarget 2017; 8:87658-87666. [PMID: 29152109 PMCID: PMC5675661 DOI: 10.18632/oncotarget.21142] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/26/2017] [Indexed: 01/04/2023] Open
Abstract
Acute traumatic spinal cord injury (tSCI) results in a lifetime of paralysis associated with a host of medical complications. The developing secondary complications of tSCI may result in further chronic neurodegenerative diseases. Sevoflurane preconditioning (SF-PreCon) has shown guaranteed protective effects in myocardial or cerebral ischemic/reperfusion injury. However, the role of SF-PreCon in tSCI still remains to be elucidated. Here, we found that SF-PreCon ameliorated the developing secondary complications through reducing the apoptosis rate and the secretion of inflammatory cytokines in injured spinal cord tissues, and therefore enhancing the recovery after tSCI. Notably, we demonstrated that SF-PreCon ameliorates tSCI through inhibiting Cycloxygenase-2 (COX-2). Importantly, we verified that SF-PreCon inhibits the expression of COX-2 and reduces the apoptosis rate after tSCI via the induction of Caveolin-3 (Cav-3). Taken together, our results suggest that SF-PreCon ameliorates tSCI via Cav-3-dependent COX-2 inhibition and provide an economical and practical method against the secondary injury after tSCI.
Collapse
Affiliation(s)
- Weidong Wu
- Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu, PR China
| | - Ningxian Wei
- Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu, PR China
| | - Lihui Wang
- Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu, PR China
| | - Danhui Kong
- Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu, PR China
| | - Gang Shao
- Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu, PR China
| | - Yingchun Qin
- Danyang People's Hospital of Jiangsu Province, Danyang, Jiangsu, PR China
| | - Lixin Wang
- School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Yansheng Du
- School of Medicine, Indiana University, Indianapolis, Indiana, United States
| |
Collapse
|