1
|
Wu H, Wang S, Dai FB, Tang CL. Research progress in the clinical application of inhaled anesthetic sevoflurane. Med Gas Res 2025; 15:85-92. [PMID: 39436171 PMCID: PMC11515067 DOI: 10.4103/mgr.medgasres-d-23-00003] [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: 06/27/2023] [Revised: 07/15/2024] [Accepted: 08/08/2024] [Indexed: 10/23/2024] Open
Abstract
Sevoflurane has been widely used in clinical anesthesia as an inhalation anesthetic. With the development of medicine, there have been several new applications in recent years, such as daytime surgery, labor analgesia, and combined nerve block for some surgeries. Moreover, as research progresses, it has been found that it not only has potential organ protection effects but can also be used to treat severe asthma and relieve the tracheal spasm state. In addition, local administration can effectively treat vascular ulcers. We briefly review the organ protective effect of sevoflurane, its application in dental treatment, asthma treatment, vascular ulcer treatment and some new progress in clinical application.
Collapse
Affiliation(s)
- Hao Wu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of Science and Technology of China, Hefei, Anhui Province, China
| | - Sheng Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of Science and Technology of China, Hefei, Anhui Province, China
| | - Fei-Biao Dai
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of Science and Technology of China, Hefei, Anhui Province, China
| | - Chao-Liang Tang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of Science and Technology of China, Hefei, Anhui Province, China
- Department of Anesthesiology, Anhui Provincial Cancer Hospital, Hefei, Anhui Province, China
| |
Collapse
|
2
|
Morax L, Beck-Schimmer B, Neff J, Mueller M, Flury-Frei R, Schläpfer M. Sevoflurane Postconditioning Protects From an Early Neurological Deficit After Subarachnoid Hemorrhage: Results of a Randomized Laboratory Study in Rats. Anesth Analg 2024; 139:1075-1085. [PMID: 39437202 DOI: 10.1213/ane.0000000000006829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is associated with neurocognitive impairment. Recent data suggest that sevoflurane attenuates edema formation after SAH in rats. However, so far, no information is available about the long-term repair phase, nor if sevoflurane impacts functionality by increasing vascularity. This study tested whether sevoflurane postconditioning would improve long-term neurologic deficit through increased formation of new vessels close to the hemorrhage area. METHODS Fifty-three animals were subjected to SAH or sham surgery with or without a 2-hour sevoflurane postconditioning (versus propofol anesthesia). Animal survival, including dropout animals due to death or reaching termination criteria, as well as neurologic deficit, defined by the Garcia score, were assessed 2 hours after recovery until postoperative day 14. On day 14, blood samples and brain tissue were harvested. Vessel density was determined by the number of cluster of differentiation 31 (CD31)-positive vessels, and activated glial cells by glial fibrillary acidic protein (GFAP)-positive astrocytes per field of view. RESULTS The survival rate for sham animals was 100%, 69% in the SAH-propofol and 92% in the SAH-sevoflurane groups. According to the log-rank Mantel-Cox test, survival curves were significantly different ( P = .024). The short-term neurologic deficit was higher in SAH-propofol versus SAH-sevoflurane animals 2 hours after recovery and on postoperative day 1 (propofol versus sevoflurane: 14. 6 ± 3.4 vs 15. 9 ± 2.7 points, P = .034, and 16. 2 ± 3.5 vs 17. 8 ± 0.9 points, P = .015). Overall complete recovery from neurologic deficit was observed on day 7 in both SAH groups (18. 0 ± 0.0 vs 18. 0 ± 0.0 points, P = 1.000). Cortical vascular density increased to 80. 6 ± 15.0 vessels per field of view in SAH-propofol animals (vs 71. 4 ± 10.1 in SAH-sevoflurane, P < .001). Activation of glial cells, an indicator of neuroinflammation, was assessed by GFAP-positive astrocytes GFAP per field of view. Hippocampal GFAP-positive cells were 201 ± 68 vs 179 ± 84 cells per field of view in SAH-propofol versus SAH-sevoflurane animals ( P < .001). CONCLUSIONS Sevoflurane postconditioning improves survival by 23% (SAH-sevoflurane versus SAH-propofol). The sevoflurane intervention could attenuate the early neurologic deficit, while the long-term outcome was similar across the groups. A higher vascular density close to the SAH area in the propofol group was not associated with improved outcomes.
Collapse
Affiliation(s)
- Laurent Morax
- From the Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Beatrice Beck-Schimmer
- From the Institute of Physiology, University of Zurich, Zurich, Switzerland
- Institute of Anesthesiology, University Hospital Zurich, Zurich, Switzerland
| | - Jonah Neff
- From the Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Mattia Mueller
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Renata Flury-Frei
- Department of Pathology, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Martin Schläpfer
- From the Institute of Physiology, University of Zurich, Zurich, Switzerland
- Institute of Anesthesiology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Ni W, Zou Z, Jiang P, Wang S. Sevoflurane alleviates inflammation, apoptosis and permeability damage of human umbilical vein endothelial cells induced by lipopolysaccharide by inhibiting endoplasmic reticulum stress via upregulating RORα. Prostaglandins Other Lipid Mediat 2024; 172:106821. [PMID: 38373554 DOI: 10.1016/j.prostaglandins.2024.106821] [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: 12/07/2023] [Revised: 02/08/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
Endothelial dysfunction often accompanies sepsis. Sevoflurane (Sev) is a widely used inhaled anesthetic that has a protective effect on sepsis-associated damage. We aimed to elucidate the role of Sev in endothelial dysfunction by using a model of LPS induced HUVECs. Sev increased the viability and decreased the apoptosis of HUVECs exposed to LPS. Inflammation and endothelial cell adhesion were improved after Sev addition. Besides, Sev alleviated LPS-induced endothelial cell permeability damage in HUVECs. RORα served as a potential protein that bound to Sev. Importantly, Sev upregulated RORα expression and inhibited endoplasmic reticulum (ER) stress in LPS-treated HUVECs. RORα silencing reversed the impacts of Sev on ER stress. Moreover, RORα deficiency or tunicamycin (ER stress inducer) treatment restored the effects of Sev on the viability, apoptosis, inflammation and endothelial permeability damage of HUVECs exposed to LPS. Taken together, Sev ameliorated LPS-induced endothelial cell damage by targeting RORα to inhibit ER stress.
Collapse
Affiliation(s)
- Weiwei Ni
- Department of Anesthesiology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213000, China; Department of Anesthesiology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu 213000, China
| | - Zhiwei Zou
- Department of Anesthesiology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213000, China; Department of Anesthesiology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu 213000, China
| | - Ping Jiang
- Department of Anesthesiology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213000, China; Department of Anesthesiology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu 213000, China
| | - Shuo Wang
- Department of Anesthesiology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213000, China; Department of Anesthesiology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu 213000, China.
| |
Collapse
|
4
|
Cheng J, Wang Z, Yu H, Chen Y, Wang Z, Zhang L, Peng X. The duration-dependent and sex-specific effects of neonatal sevoflurane exposure on cognitive function in rats. Braz J Med Biol Res 2024; 57:e13437. [PMID: 38808889 PMCID: PMC11136479 DOI: 10.1590/1414-431x2024e13437] [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: 10/24/2023] [Accepted: 04/07/2024] [Indexed: 05/30/2024] Open
Abstract
Clinical studies have found that neonatal sevoflurane exposure can increase the risk of cognitive dysfunction. However, recent studies have found that it can exhibit neuroprotective effects in some situations. In this study, we aimed to explore the effects of sevoflurane neonatal exposure in rats. A total of 144 rat pups (72 males and 72 females) were assigned to six groups and separately according to sevoflurane exposure of different times on the seventh day after birth. Blood gas analysis and western blot detection in the hippocampus were conducted after exposure. The Morris water maze test was conducted on the 32nd to 38th days after birth. The expression of PSD95 and synaptophysin in the hippocampus was detected after the Morris water maze test. We found that neonatal exposure to sevoflurane promoted apoptosis in the hippocampus, and Bax and caspase-3 were increased in a dose-dependent manner. The 2-h exposure had the greatest effects on cognitive dysfunction. However, with the extension of exposure time to 6 h, the effects on cognitive function were partly compensated. In addition, sevoflurane exposure decreased synaptogenesis in the hippocampus. However, as the exposure time was extended, the suppression of synaptogenesis was attenuated. In conclusion, neonatal sevoflurane exposure exhibited duration-dependent effects on cognitive function via Bax-caspase-3-dependent apoptosis and bidirectional effects on synaptogenesis in rats.
Collapse
Affiliation(s)
- Jiangxia Cheng
- Department of Anesthesia, Wuhan Fourth Hospital, Wuhan, China
- Department of Anesthesia, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhuo Wang
- Department of Anesthesia, Wuhan Fourth Hospital, Wuhan, China
| | - Hui Yu
- Department of Anesthesia, Wuhan Fourth Hospital, Wuhan, China
| | - Ye Chen
- Department of Anesthesia, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhengchao Wang
- Department of Orthopedics, Wuhan Fourth Hospital, Wuhan, China
| | - Liangcheng Zhang
- Department of Anesthesia, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaohong Peng
- Department of Anesthesia, Wuhan Fourth Hospital, Wuhan, China
| |
Collapse
|
5
|
Wang L, Zhang Q, Zhang Y, Zheng G, Wang K, Wu Z, Zhang J, Jia W, Zhang G. Insufficiency of plasmatic arginine/homoarginine during the initial postoperative phase among patients with tumors affecting the medulla oblongata heightens the likelihood of neurogenic pulmonary oedema following surgery. Int J Surg 2024; 110:1475-1483. [PMID: 38079589 PMCID: PMC10942246 DOI: 10.1097/js9.0000000000000957] [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: 07/15/2023] [Accepted: 11/20/2023] [Indexed: 03/16/2024]
Abstract
BACKGROUND This prospective clinical study aims to investigate the fluctuations of neurotransmitters in peripheral venous blood during the perioperative period and to identify independent predictors for postoperative neurogenic pulmonary oedema (NPE) in patients with medulla oblongata-involved tumours. MATERIALS AND METHODS Peripheral venous blood samples of the enroled patients at seven perioperative time points, as well as their medical records and radiologic data were collected. High-performance liquid chromatography-tandem mass spectrometry was utilized to detect the concentrations of 39 neurotransmitters in these samples. The study applied univariate and multivariate generalized estimating equation (GEE) logistic regression analyses to explore independent predictors of postoperative NPE, and one-way repeated-measures ANOVA to compare the concentrations of the same neurotransmitter at different perioperative time points. RESULTS The study included 36 patients with medulla oblongata-involved tumours from January to December 2019, and found that 13.9% of them experienced postoperative NPE. The absence of intraoperative use of sevoflurane ( P =0.008), decreased concentrations of arginine ( P =0.026) and homoarginine ( P =0.030), and prolonged postoperative tracheal extubation ( P <0.001) were identified as independent risk factors for postoperative NPE in medulla oblongata-involved tumour patients. Pairwise comparison analysis revealed that the perioperative decreases in arginine and homoarginine concentrations mainly occurred within the postoperative 8 h. CONCLUSION This study demonstrates that NPE is not uncommon in patients with medulla oblongata-involved tumours. The absence of intraoperative use of sevoflurane, decreased concentrations of plasmatic arginine and homoarginine, and prolonged postoperative tracheal extubation are independent predictors of postoperative NPE. These two neurotransmitters' concentrations dropped mainly within the early postoperative hours and could serve as potential early warning indicators of postoperative NPE in clinical practice.
Collapse
Affiliation(s)
- Liang Wang
- Department of Neurosurgery
- China National Clinical Research Center for Neurological Diseases
- Center of Brain Tumor, Beijing Institute for Brain Disorders
- Beijing Key Laboratory of Brain Tumor, Beijing, People’s Republic of China
| | - Qing Zhang
- Department of Neurosurgery
- China National Clinical Research Center for Neurological Diseases
- Center of Brain Tumor, Beijing Institute for Brain Disorders
- Beijing Key Laboratory of Brain Tumor, Beijing, People’s Republic of China
| | - Yuan Zhang
- Department of Neurosurgery
- China National Clinical Research Center for Neurological Diseases
- Center of Brain Tumor, Beijing Institute for Brain Disorders
- Beijing Key Laboratory of Brain Tumor, Beijing, People’s Republic of China
| | - Guanghui Zheng
- Laboratory Diagnosis Center, Beijing Tiantan Hospital, Capital Medical University
| | - Ke Wang
- Department of Neurosurgery
- China National Clinical Research Center for Neurological Diseases
- Center of Brain Tumor, Beijing Institute for Brain Disorders
- Beijing Key Laboratory of Brain Tumor, Beijing, People’s Republic of China
| | - Zhen Wu
- Department of Neurosurgery
- China National Clinical Research Center for Neurological Diseases
- Center of Brain Tumor, Beijing Institute for Brain Disorders
- Beijing Key Laboratory of Brain Tumor, Beijing, People’s Republic of China
| | - Junting Zhang
- Department of Neurosurgery
- China National Clinical Research Center for Neurological Diseases
- Center of Brain Tumor, Beijing Institute for Brain Disorders
- Beijing Key Laboratory of Brain Tumor, Beijing, People’s Republic of China
| | - Wang Jia
- Department of Neurosurgery
- China National Clinical Research Center for Neurological Diseases
- Center of Brain Tumor, Beijing Institute for Brain Disorders
- Beijing Key Laboratory of Brain Tumor, Beijing, People’s Republic of China
| | - Guojun Zhang
- Laboratory Diagnosis Center, Beijing Tiantan Hospital, Capital Medical University
| |
Collapse
|
6
|
Sun J, Zhang L, Cheng Q, Wu Y. Aberrant expression and regulatory role of histone deacetylase 9 in vascular endothelial cell injury in intracranial aneurysm. BIOMOLECULES & BIOMEDICINE 2024; 24:61-72. [PMID: 37573538 PMCID: PMC10787617 DOI: 10.17305/bb.2023.9364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/18/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023]
Abstract
Intracranial aneurysm (IA) is one of the most challenging cerebrovascular lesions for clinicians. The aim of this study was to investigate the abnormal expression and role of histone deacetylase 9 (HDAC9) in IA-associated injury of vascular endothelial cells (VECs). First, IA tissue and normal arterial tissue were collected and VECs were isolated from IA patients. The expression levels of HDAC9, microRNA (miR)-34a-5p, and vascular endothelial growth factor-A (VEGFA) were determined. Cell viability, proliferation, apoptosis, and migration were assessed by Cell Counting Kit-8 (CCK-8) assay, EdU staining, TUNEL staining, and transwell assay. The binding of miR-34a-5p to VEGFA was analyzed by the dual-luciferase assay, and the accumulation of HDAC9 and lysine histone acetylation at H3 (H3K9, H3K14, and H3K18) on the miR-34a-5p promoter was detected by the chromatin immunoprecipitation assay. The results showed that HDAC9 and VEGFA were increased and miR-34a-5p was decreased in IA tissues and cells. Silencing of HDAC9 inhibited apoptosis and increased viability, proliferation, and migration of VECs, whereas overexpression of HDAC9 exerted the opposite functions. HDAC9 accumulated at the miR-34a-5p promoter to decrease miR-34a-5p expression by reducing H3 locus-specific acetylation and further promoted VEGFA expression. Knockdown of miR-34a-5p or VEGFA overexpression reversed the protective role of HDAC9 silencing in VECs injury. In conclusion, our study suggests that HDAC9 may be a therapeutic target for IA.
Collapse
Affiliation(s)
- Jingwei Sun
- Department of Neurosurgery, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Langfeng Zhang
- Interventional Treatment Department, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Quanjiang Cheng
- Department of Neurosurgery, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Yajun Wu
- Department of Neurosurgery, Shenzhen Longhua District Central Hospital, Shenzhen, China
| |
Collapse
|
7
|
Scheid S, Goebel U, Ulbrich F. Neuroprotection Is in the Air-Inhaled Gases on Their Way to the Neurons. Cells 2023; 12:2480. [PMID: 37887324 PMCID: PMC10605176 DOI: 10.3390/cells12202480] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/29/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
Cerebral injury is a leading cause of long-term disability and mortality. Common causes include major cardiovascular events, such as cardiac arrest, ischemic stroke, and subarachnoid hemorrhage, traumatic brain injury, and neurodegenerative as well as neuroinflammatory disorders. Despite improvements in pharmacological and interventional treatment options, due to the brain's limited regeneration potential, survival is often associated with the impairment of crucial functions that lead to occupational inability and enormous economic burden. For decades, researchers have therefore been investigating adjuvant therapeutic options to alleviate neuronal cell death. Although promising in preclinical studies, a huge variety of drugs thought to provide neuroprotective effects failed in clinical trials. However, utilizing medical gases, noble gases, and gaseous molecules as supportive treatment options may offer new perspectives for patients suffering neuronal damage. This review provides an overview of current research, potentials and mechanisms of these substances as a promising therapeutic alternative for the treatment of cerebral injury.
Collapse
Affiliation(s)
- Stefanie Scheid
- Department of Anesthesiology and Critical Care, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| | - Ulrich Goebel
- Department of Anesthesiology and Critical Care Medicine, St. Franziskus-Hospital, 48145 Muenster, Germany;
| | - Felix Ulbrich
- Department of Anesthesiology and Critical Care, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| |
Collapse
|
8
|
Zhai R, Lenga Ma Bonda W, Leclaire C, Saint-Béat C, Theilliere C, Belville C, Coupet R, Blondonnet R, Bouvier D, Blanchon L, Sapin V, Jabaudon M. Effects of sevoflurane on lung epithelial permeability in experimental models of acute respiratory distress syndrome. J Transl Med 2023; 21:397. [PMID: 37331963 DOI: 10.1186/s12967-023-04253-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/08/2023] [Indexed: 06/20/2023] Open
Abstract
BACKGROUND Preclinical studies in acute respiratory distress syndrome (ARDS) have suggested that inhaled sevoflurane may have lung-protective effects and clinical trials are ongoing to assess its impact on major clinical outcomes in patients with ARDS. However, the underlying mechanisms of these potential benefits are largely unknown. This investigation focused on the effects of sevoflurane on lung permeability changes after sterile injury and the possible associated mechanisms. METHODS To investigate whether sevoflurane could decrease lung alveolar epithelial permeability through the Ras homolog family member A (RhoA)/phospho-Myosin Light Chain 2 (Ser19) (pMLC)/filamentous (F)-actin pathway and whether the receptor for advanced glycation end-products (RAGE) may mediate these effects. Lung permeability was assessed in RAGE-/- and littermate wild-type C57BL/6JRj mice on days 0, 1, 2, and 4 after acid injury, alone or followed by exposure at 1% sevoflurane. Cell permeability of mouse lung epithelial cells was assessed after treatment with cytomix (a mixture of TNFɑ, IL-1β, and IFNγ) and/or RAGE antagonist peptide (RAP), alone or followed by exposure at 1% sevoflurane. Levels of zonula occludens-1, E-cadherin, and pMLC were quantified, along with F-actin immunostaining, in both models. RhoA activity was assessed in vitro. RESULTS In mice after acid injury, sevoflurane was associated with better arterial oxygenation, decreased alveolar inflammation and histological damage, and non-significantly attenuated the increase in lung permeability. Preserved protein expression of zonula occludens-1 and less increase of pMLC and actin cytoskeletal rearrangement were observed in injured mice treated with sevoflurane. In vitro, sevoflurane markedly decreased electrical resistance and cytokine release of MLE-12 cells, which was associated with higher protein expression of zonula occludens-1. Improved oxygenation levels and attenuated increase in lung permeability and inflammatory response were observed in RAGE-/- mice compared to wild-type mice, but RAGE deletion did not influence the effects of sevoflurane on permeability indices after injury. However, the beneficial effect of sevoflurane previously observed in wild-type mice on day 1 after injury in terms of higher PaO2/FiO2 and decreased alveolar levels of cytokines was not found in RAGE-/- mice. In vitro, RAP alleviated some of the beneficial effects of sevoflurane on electrical resistance and cytoskeletal rearrangement, which was associated with decreased cytomix-induced RhoA activity. CONCLUSIONS Sevoflurane decreased injury and restored epithelial barrier function in two in vivo and in vitro models of sterile lung injury, which was associated with increased expression of junction proteins and decreased actin cytoskeletal rearrangement. In vitro findings suggest that sevoflurane may decrease lung epithelial permeability through the RhoA/pMLC/F-actin pathway.
Collapse
Affiliation(s)
- Ruoyang Zhai
- iGReD, UFR de Médecine et des Professions Paramédicales, Place Henri Dunant, CNRS, INSERM, Université Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Woodys Lenga Ma Bonda
- iGReD, UFR de Médecine et des Professions Paramédicales, Place Henri Dunant, CNRS, INSERM, Université Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Charlotte Leclaire
- iGReD, UFR de Médecine et des Professions Paramédicales, Place Henri Dunant, CNRS, INSERM, Université Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Cécile Saint-Béat
- iGReD, UFR de Médecine et des Professions Paramédicales, Place Henri Dunant, CNRS, INSERM, Université Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Camille Theilliere
- iGReD, UFR de Médecine et des Professions Paramédicales, Place Henri Dunant, CNRS, INSERM, Université Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Corinne Belville
- iGReD, UFR de Médecine et des Professions Paramédicales, Place Henri Dunant, CNRS, INSERM, Université Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Randy Coupet
- iGReD, UFR de Médecine et des Professions Paramédicales, Place Henri Dunant, CNRS, INSERM, Université Clermont Auvergne, 63000, Clermont-Ferrand, France
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Raiko Blondonnet
- iGReD, UFR de Médecine et des Professions Paramédicales, Place Henri Dunant, CNRS, INSERM, Université Clermont Auvergne, 63000, Clermont-Ferrand, France
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Damien Bouvier
- iGReD, UFR de Médecine et des Professions Paramédicales, Place Henri Dunant, CNRS, INSERM, Université Clermont Auvergne, 63000, Clermont-Ferrand, France
- Department of Medical Biochemistry and Molecular Genetics, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Loic Blanchon
- iGReD, UFR de Médecine et des Professions Paramédicales, Place Henri Dunant, CNRS, INSERM, Université Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Vincent Sapin
- iGReD, UFR de Médecine et des Professions Paramédicales, Place Henri Dunant, CNRS, INSERM, Université Clermont Auvergne, 63000, Clermont-Ferrand, France
- Department of Medical Biochemistry and Molecular Genetics, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Matthieu Jabaudon
- iGReD, UFR de Médecine et des Professions Paramédicales, Place Henri Dunant, CNRS, INSERM, Université Clermont Auvergne, 63000, Clermont-Ferrand, France.
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France.
| |
Collapse
|
9
|
Roth Z'graggen B, Urner M, Beck-Schimmer B, Schläpfer M. Effects of sevoflurane and its metabolite hexafluoroisopropanol on hypoxia/reoxygenation-induced injury and mitochondrial bioenergetics in murine cardiomyocytes. BJA OPEN 2023; 5:100116. [PMID: 37587996 PMCID: PMC10430838 DOI: 10.1016/j.bjao.2022.100116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 11/22/2022] [Indexed: 08/18/2023]
Abstract
Background The volatile anaesthetic sevoflurane protects cardiac tissue from reoxygenation/reperfusion. Mitochondria play an essential role in conditioning. We aimed to investigate how sevoflurane and its primary metabolite hexafluoroisopropanol (HFIP) affect necrosis, apoptosis, and reactive oxygen species formation in cardiomyocytes upon hypoxia/reoxygenation injury. Moreover, we aimed to describe the similarities in the mode of action in a mitochondrial bioenergetics analysis. Methods Murine cardiomyocytes were exposed to hypoxia (0.2% O2 for 6 h), followed by reoxygenation (air with 5% CO2 for 2 h) in the presence or absence sevoflurane 2.2% or HFIP 4 mM. Lactate dehydrogenase (LDH) release (necrosis), caspase activation (apoptosis), reactive oxygen species, mitochondrial membrane potential, and mitochondrial function (Seahorse XF analyser) were measured. Results Hypoxia/reoxygenation increased cell death by 44% (+31 to +55%, P<0.001). Reoxygenation in the presence of sevoflurane 2.2% or HFIP 4 mM increased LDH release only by +18% (+6 to +30%) and 20% (+7 to +32%), respectively. Apoptosis and reactive oxygen species formation were attenuated by sevoflurane and HFIP. Mitochondrial bioenergetics analysis of the two substances was profoundly different. Sevoflurane did not influence oxygen consumption rate (OCR) or extracellular acidification rate (ECAR), whereas HFIP reduced OCR and increased ECAR, an effect similar to oligomycin, an adenosine triphosphate (ATP) synthase inhibitor. When blocking the metabolism of sevoflurane into HFIP, protective effects of sevoflurane - but not of HFIP - on LDH release and caspase were mitigated. Conclusion Together, our data suggest that sevoflurane metabolism into HFIP plays an essential role in cardiomyocyte postconditioning after hypoxia/reoxygenation injury.
Collapse
Affiliation(s)
| | - Martin Urner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Interdepartmental Division of Critical Care Medicine and University of Toronto, Toronto, Canada
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Canada
| | - Beatrice Beck-Schimmer
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Institute of Anaesthesiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Martin Schläpfer
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Institute of Anaesthesiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Prolonged anesthesia induces neuroinflammation and complement-mediated microglial synaptic elimination involved in neurocognitive dysfunction and anxiety-like behaviors. BMC Med 2023; 21:7. [PMID: 36600274 PMCID: PMC9814183 DOI: 10.1186/s12916-022-02705-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Perioperative neurocognitive disorders (PND) with a high incidence frequently occur in elderly surgical patients closely associated with prolonged anesthesia-induced neurotoxicity. The neuromorphopathological underpinnings of anesthesia-induced neurotoxicity have remained elusive. METHODS Prolonged anesthesia with sevoflurane was used to establish the sevoflurane-induced neurotoxicity (SIN) animal model. Morris water maze, elevated plus maze, and open field test were employed to track SIN rats' cognitive behavior and anxiety-like behaviors. We investigated the neuropathological basis of SIN through techniques such as transcriptomic, electrophysiology, molecular biology, scanning electron microscope, Golgi staining, TUNEL assay, and morphological analysis. Our work further clarifies the pathological mechanism of SIN by depleting microglia, inhibiting neuroinflammation, and C1q neutralization. RESULTS This study shows that prolonged anesthesia triggers activation of the NF-κB inflammatory pathway, neuroinflammation, inhibition of neuronal excitability, cognitive dysfunction, and anxiety-like behaviors. RNA sequencing found that genes of different types of synapses were downregulated after prolonged anesthesia. Microglial migration, activation, and phagocytosis were enhanced. Microglial morphological alterations were also observed. C1qa, the initiator of the complement cascade, and C3 were increased, and C1qa tagging synapses were also elevated. Then, we found that the "Eat Me" complement pathway mediated microglial synaptic engulfment in the hippocampus after prolonged anesthesia. Afterward, synapses were remarkably lost in the hippocampus. Furthermore, dendritic spines were reduced, and their genes were also downregulated. Depleting microglia ameliorated the activation of neuroinflammation and complement and rescued synaptic loss, cognitive dysfunction, and anxiety-like behaviors. When neuroinflammatory inhibition or C1q neutralization occurred, complement was also decreased, and synaptic elimination was interrupted. CONCLUSIONS These findings illustrated that prolonged anesthesia triggered neuroinflammation and complement-mediated microglial synaptic engulfment that pathologically caused synaptic elimination in SIN. We have demonstrated the neuromorphopathological underpinnings of SIN, which have direct therapeutic relevance for PND patients.
Collapse
|
11
|
Zhao Y, Gan L, Ren L, Lin Y, Ma C, Lin X. Factors influencing the blood-brain barrier permeability. Brain Res 2022; 1788:147937. [PMID: 35568085 DOI: 10.1016/j.brainres.2022.147937] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) is a dynamic structure that protects the brain from harmful blood-borne, endogenous and exogenous substances and maintains the homeostatic microenvironment. All constituent cell types play indispensable roles in the BBB's integrity, and other structural BBB components, such as tight junction proteins, adherens junctions, and junctional proteins, can control the barrier permeability. Regarding the need to exchange nutrients and toxic materials, solute carriers, ATP-binding case families, and ion transporter, as well as transcytosis regulate the influx and efflux transport, while the difference in localisation and expression can contribute to functional differences in transport properties. Numerous chemical mediators and other factors such as non-physicochemical factors have been identified to alter BBB permeability by mediating the structural components and barrier function, because of the close relationship with inflammation. In this review, we highlight recently gained mechanistic insights into the maintenance and disruption of the BBB. A better understanding of the factors influencing BBB permeability could contribute to supporting promising potential therapeutic targets for protecting the BBB and the delivery of central nervous system drugs via BBB permeability interventions under pathological conditions.
Collapse
Affiliation(s)
- Yibin Zhao
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Gan
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Ren
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yubo Lin
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Congcong Ma
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianming Lin
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
12
|
Li MM, Zheng YL, Wang WD, Lin S, Lin HL. Neuropeptide Y: An Update on the Mechanism Underlying Chronic Intermittent Hypoxia-Induced Endothelial Dysfunction. Front Physiol 2021; 12:712281. [PMID: 34512386 PMCID: PMC8430344 DOI: 10.3389/fphys.2021.712281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/02/2021] [Indexed: 12/17/2022] Open
Abstract
Endothelial dysfunction (ED) is a core pathophysiological process. The abnormal response of vascular endothelial (VE) cells to risk factors can lead to systemic consequences. ED caused by intermittent hypoxia (IH) has also been recognized. Neuropeptide Y (NPY) is an important peripheral neurotransmitter that binds to different receptors on endothelial cells, thereby causing ED. Additionally, hypoxia can induce the release of peripheral NPY; however, the involvement of NPY and its receptor in IH-induced ED has not been determined. This review explains the definition of chronic IH and VE function, including the relationship between ED and chronic IH-related vascular diseases. The results showed that that the effect of IH on VE injury is mediated by the VE-barrier structure and endothelial cell dysfunction. These findings offer new ideas for the prevention and treatment of obstructive sleep apnea syndrome and its complications.
Collapse
Affiliation(s)
- Mei-Mei Li
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yan-Li Zheng
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wan-da Wang
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.,Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.,Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Hui-Li Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
13
|
Liang TY, Peng SY, Ma M, Li HY, Wang Z, Chen G. Protective effects of sevoflurane in cerebral ischemia reperfusion injury: a narrative review. Med Gas Res 2021; 11:152-154. [PMID: 34213497 PMCID: PMC8374460 DOI: 10.4103/2045-9912.318860] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Ischemia/reperfusion (I/R) injury is a phenomenon that the reperfusion of ischemic organs or tissues aggravates their damage, which poses a serious health threat and economic burden to the world. I/R gives rise to a series of physiological and pathological world, including inflammatory response, oxidative stress, brain edema, blood-brain barrier destruction, and neuronal death. Therefore, finding effective treatment measures is extremely important to the recovery of I/R patients and the improvement of long-term quality of life. Sevoflurane is an important volatile anesthetic which has been reported to reduce myocardial I/R damage and infarct size. Sevoflurane also has anti-inflammatory and neuroprotective effects. As reported sevoflurane treatment could reduce nerve function injury, cerebral infarction volume and the level of inflammatory factors. At the same time, there is evidence that sevoflurane can reduce neuron apoptosis and antioxidant stress. The protective effect of sevoflurane in brain injury has been proved to be existed in several aspects, so that a comprehensive understanding of its neuroprotective effect is helpful to exploit new treatment paths for I/R, provide clinicians with new clinical treatment decisions, contribute to the effective treatment of I/R patients and the improvement of quality of life after I/R healing.
Collapse
Affiliation(s)
- Tian-Yu Liang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Song-Yang Peng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Mian Ma
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu Province, China
| | - Hai-Ying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
14
|
Lee YC, Kao ST, Cheng CY. Acorus tatarinowii Schott extract reduces cerebral edema caused by ischemia-reperfusion injury in rats: involvement in regulation of astrocytic NKCC1/AQP4 and JNK/iNOS-mediated signaling. BMC Complement Med Ther 2020; 20:374. [PMID: 33298024 PMCID: PMC7726880 DOI: 10.1186/s12906-020-03168-z] [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: 03/31/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022] Open
Abstract
Background This study aimed to evaluate the effects of the Acorus tatarinowii Schott [Shi Chang Pu (SCP)] extract administered at the start of 2 h of middle cerebral artery occlusion (MCAo), followed by 3 d of reperfusion, and to determine mechanisms involved in anti-edema effects in the penumbra of the cerebral cortex. Method Rats were intraperitoneally administered the SCP extract at a dose of 0.25 g/kg (SCP-0.25 g), 0.5 g/kg (SCP-0.5 g), or 1 g/kg (SCP-1 g) at the start of MCAo. Result SCP-0.5 g and SCP-1 g treatments effectively reduced the cerebral infarct size, ameliorated cerebral edema, reduced blood–brain barrier permeability, and restored neurological function. SCP-0.5 g and SCP-1 g treatments markedly downregulated the levels of glial fibrillary acidic protein, Na+-K+-2Cl− cotransporter type 1 (NKCC1), aquaporin 4 (AQP4), phospho-c-Jun N-terminal kinase (p-JNK)/JNK, inducible nitric oxide synthase (iNOS), 3-nitrotyrosine, intercellular adhesion molecule-1 (ICAM-1), matrix metalloproteinase-9 (MMP-9), vascular endothelial growth factor-A (VEGF-A), and zonula occluden-1 (ZO-1) and upregulated ZO-3 expression in the penumbra of the cerebral cortex 3 d after reperfusion. Conclusions SCP-0.5 g and SCP-1 g treatments exert neuroprotective effects against cerebral infarction and cerebral edema partially by mitigating astrocytic swelling and blood–brain barrier disruption. Moreover, the anti-cerebral edema effects of SCP extract treatments are possibly associated with the downregulation of astrocytic NKCC1/AQP4 and JNK/iNOS-mediated ICAM-1/MMP-9 signaling in the penumbra of the cerebral cortex 3 d after reperfusion.
Collapse
Affiliation(s)
- Yu-Chen Lee
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan.,Department of Chinese Medicine, China Medical University Hospital 40447, Taichung, Taiwan.,Research Center for Chinese Medicine & Acupuncture, China Medical University, Taichung, 40402, Taiwan
| | - Shung-Te Kao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Chin-Yi Cheng
- School of Post-baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan. .,Department of Chinese Medicine, Hui-Sheng Hospital 42056, Taichung, Taiwan.
| |
Collapse
|
15
|
Neag MA, Mitre AO, Catinean A, Mitre CI. An Overview on the Mechanisms of Neuroprotection and Neurotoxicity of Isoflurane and Sevoflurane in Experimental Studies. Brain Res Bull 2020; 165:281-289. [DOI: 10.1016/j.brainresbull.2020.10.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022]
|
16
|
Abstract
Central nervous system injuries are a leading cause of death and disability worldwide. Although the exact pathophysiological mechanisms of various brain injuries vary, central nervous system injuries often result in an inflammatory response, and subsequently lead to brain damage. This suggests that neuroprotection may be necessany in the treatment of multiple disease models. The use of medical gases as neuroprotective agents has gained great attention in the medical field. Medical gases include common gases, such as oxygen, hydrogen and carbon dioxide; hydrogen sulphide and nitric oxide that have been considered toxic; volatile anesthetic gases, such as isoflurane and sevoflurane; and inert gases like helium, argon, and xenon. The neuroprotection from these medical gases has been investigated in experimental animal models of various types of brain injuries, such as traumatic brain injury, stroke, subarachnoid hemorrhage, cerebral ischemic/reperfusion injury, and neurodegenerative diseases. Nevertheless, the transition into the clinical practice is still lagging. This delay could be attributed to the contradictory paradigms and the conflicting results that have been obtained from experimental models, as well as the presence of inconsistent reports regarding their safety. In this review, we summarize the potential mechanisms underlying the neuroprotective effects of medical gases and discuss possible candidates that could improve the outcomes of brain injury.
Collapse
Affiliation(s)
- Yue-Zhen Wang
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ting-Ting Li
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Hong-Ling Cao
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wan-Chao Yang
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
17
|
Beck-Schimmer B, Restin T, Muroi C, Roth Z'Graggen B, Keller E, Schläpfer M. Sevoflurane sedation attenuates early cerebral oedema formation through stabilisation of the adherens junction protein beta catenin in a model of subarachnoid haemorrhage: A randomised animal study. Eur J Anaesthesiol 2020; 37:402-412. [PMID: 32068571 DOI: 10.1097/eja.0000000000001161] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Severe neurological impairment is a problem after subarachnoid haemorrhage (SAH). Although volatile anaesthetics, such as sevoflurane, have demonstrated protective properties in many organs, their use in cerebral injury is controversial. Cerebral vasodilation may lead to increased intracranial pressure (ICP), but at the same time volatile anaesthetics are known to stabilise the SAH-injured endothelial barrier. OBJECTIVE To test the effect of sevoflurane on ICP and blood-brain barrier function. DESIGN Randomised study. PARTICIPANTS One hundred male Wistar rats included, 96 analysed. INTERVENTIONS SAH was induced by the endoluminal filament method under ketamine/xylazine anaesthesia. Fifteen minutes after sham surgery or induction of SAH, adult male Wistar rats were randomised to 4 h sedation with either propofol or sevoflurane. MAIN OUTCOME MEASURES Mean arterial pressure (MAP), ICP, extravasation of water (small), Evan's blue (intermediate) and IgG (large molecule) were measured. Zonula occludens-1 (ZO-1) and beta-catenin (β-catenin), as important representatives of tight and adherens junction proteins, were determined by western blot. RESULTS Propofol and sevoflurane sedation did not affect MAP or ICP in SAH animals. Extravasation of small molecules was higher in SAH-propofol compared with SAH-sevoflurane animals (79.1 ± 0.9 vs. 78.0 ± 0.7%, P = 0.04). For intermediate and large molecules, no difference was detected (P = 0.6 and P = 0.2). Both membrane and cytosolic fractions of ZO-1 as well as membrane β-catenin remained unaffected by the injury and type of sedation. Decreased cytosolic fraction of β-catenin in propofol-SAH animals (59 ± 15%) was found to reach values of sham animals (100%) in the presence of sevoflurane in SAH animals (89 ± 21%; P = 0.04). CONCLUSION This experiment demonstrates that low-dose short-term sevoflurane sedation after SAH in vivo did not affect ICP and MAP and at the same time may attenuate early brain oedema formation, potentially by preserving adherens junctions. TRIAL REGISTRATION No 115/2014 Veterinäramt Zürich.
Collapse
Affiliation(s)
- Beatrice Beck-Schimmer
- From the Institute of Physiology and Zurich Centre for Integrative Human Physiology, University of Zurich (BBS, TR, BRZ, MS), Institute of Anaesthesiology, University Hospital Zurich, Zurich, Switzerland (BBS, TR, MS), Department of Anesthesiology, University of Illinois at Chicago, Chicago, USA (BBS) and Neurosurgical Intensive Care Unit, University Hospital Zurich, Zurich, Switzerland (CM, EK)
| | | | | | | | | | | |
Collapse
|
18
|
Yang A, Liu B. May sevoflurane prevent the development of neurogenic pulmonary edema and improve the outcome? Or as a new sedation method for severe brain injury patients. Med Hypotheses 2020; 137:109538. [PMID: 31911369 DOI: 10.1016/j.mehy.2019.109538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 02/05/2023]
Abstract
Neurogenic pulmonary edema (NPE) is a life-threatening complication that develops rapidly and dramatically after injury to the central nervous system (CNS). Severe primary brain injury and subsequent secondary brain injury cascade events are thought to be involved in the development of NPE. Activation of the sympathetic nervous system and release of vasoactive substances are also essential prerequisites for NPE. We hypothesize that sevoflurane may be an effective treatment for preventing the development of NPE. Sevoflurane may play a role in protecting brain and lung tissue after acute brain injury through its sympatholytic, antioxidative, ion channel stabilizing, anti-inflammatory, anti-apoptotic, and pulmonary protection effects. It has the potential to be used as a sedative in the neurosurgical intensive care unit (NICU), which can help maintain nervous system and cardiopulmonary function in patients with acute brain injury to improve prognosis. Sevoflurane also has the advantages of fast induction of anesthesia, rapid drug metabolism, little interference to the cardiovascular system, and controllable depth of anesthesia. If our hypothesis is supported by further experiments, use of sevoflurane may open a new door for the treatment of acute brain injury and NPE.
Collapse
Affiliation(s)
- Aobing Yang
- Department of Neurosurgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China
| | - Bin Liu
- Department of Neurosurgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China.
| |
Collapse
|
19
|
Pan W, Hu L, Chen Y, Zhu Z, Wang Y, Song J, Shan Z. Sphingosine-1-phosphate alleviates irradiation-induced parotid injury in a miniature pig model. Oral Dis 2020; 26:920-929. [PMID: 32034858 DOI: 10.1111/odi.13302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 01/24/2020] [Accepted: 02/04/2020] [Indexed: 01/26/2023]
Abstract
OBJECTIVES Our aim was to verify the alleviation effect of sphingosine-1-phosphate (S1P) in a miniature pig model. MATERIAL AND METHODS Thirty male miniature pigs were randomly separated into 10 groups in our experiment. We administered S1P through the parotid duct in a retrograde fashion 2 hr before irradiation (IR). The salivary flow rate and blood flow rate were tested 20 weeks after IR. The apoptotic level was checked at 12, 24 hr and 7 days post-IR. RESULTS Twenty weeks after IR, the salivary flow rate of the IR-side parotid gland in IR + S1P group can be maintained at about 40% of the non-IR side, while only 20% was maintained in the IR group. The blood flow rate and microvascular density were significantly higher in the IR + S1P group than in the IR group. The apoptotic level and cleaved caspase-3 expression were downregulated in IR + S1P group, and the ratio of Bcl-2/Bax was increased. The blood flow rate and CD31 level were significantly restored at 12, 24 hr and 7 days post-IR. CONCLUSION Sphingosine-1-phosphate may partially alleviate IR-induced parotid dysfunction by decreasing apoptosis of microvascular endothelial cells and maintaining the blood flow rate.
Collapse
Affiliation(s)
- Wen Pan
- Outpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing, China
| | - Liang Hu
- Outpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing, China
| | - Yawen Chen
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhao Zhu
- Outpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing, China.,Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Yingxin Wang
- Outpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing, China
| | - Jiaxin Song
- Outpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing, China
| | - Zhaochen Shan
- Outpatient Department of Oral and Maxillofacial Surgery, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
20
|
Yu Y, Fang H, Qiu Z, Xia Z, Zhou B. DHA Attenuates Hypoxia/Reoxygenation Injury by Activating SSeCKS in Human Cerebrovascular Pericytes. Neurochem Res 2019; 45:310-321. [PMID: 31776970 PMCID: PMC6985071 DOI: 10.1007/s11064-019-02915-0] [Citation(s) in RCA: 2] [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: 06/30/2019] [Revised: 11/07/2019] [Accepted: 11/18/2019] [Indexed: 12/11/2022]
Abstract
Docosahexaenoic acid (DHA) can alleviate cerebral ischemia/reperfusion injury by reducing blood–brain barrier permeability and maintaining its integrity, accompanied by an increased Ang-1/Ang-2 ratio; however, the underlying mechanisms of these effects remain unclear. Src-suppressed C kinase substrates (SSeCKS), a substrate of protein kinase C, plays an important role in maintaining cell junctions and cell morphology and regulating cell permeability. However, whether DHA can increase SSeCKS expression and then mediate the Ang-1/Ang-2 ratio still needs to be studied. Human cerebrovascular pericytes (HBVPs) cultured in vitro were divided into groups, treated with or without DHA along with SSeCKS siRNA to knockdown SSeCKS expression, and then subjected to 24 h of hypoxia followed by 6 h of reoxygenation. Cell viability; lactate dehydrogenase (LDH) release; and Ang-1, Ang-2 and VEGF activity were detected by using ELISA kits. The apoptosis rate was assessed by TUNEL flow cytometry. Expression of the SSeCKS, Ang-1, Ang-2 and VEGF proteins was evaluated by western blotting. Pretreatment with 10 μM or 40 μM DHA efficiently attenuated hypoxia/reoxygenation (H/R) injury by activating SSeCKS to increase the Ang-1/Ang-2 ratio and downregulate VEGF expression in HBVPs, as evidenced by decreased LDH release and apoptotic rates and increased HBVPs viability. Meanwhile, after we used SSeCKS siRNA to knock down SSeCKS protein expression, the protective effect of DHA on HBVPs following H/R injury was reversed. In conclusion, DHA can activate SSeCKS to increase the Ang-1/Ang-2 ratio and downregulate VEGF expression in HBVPs, thus reducing H/R injury.
Collapse
Affiliation(s)
- Yanli Yu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Haibin Fang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhen Qiu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Bin Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
21
|
Wei LK, Quan LS. Biomarkers for ischemic stroke subtypes: A protein-protein interaction analysis. Comput Biol Chem 2019; 83:107116. [PMID: 31561071 DOI: 10.1016/j.compbiolchem.2019.107116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/10/2019] [Accepted: 08/26/2019] [Indexed: 01/21/2023]
Abstract
According to the Trial of Org 10172 in Acute Stroke Treatment, ischemic stroke is classified into five subtypes. However, the predictive biomarkers of ischemic stroke subtypes are still largely unknown. The utmost objective of this study is to map, construct and analyze protein-protein interaction (PPI) networks for all subtypes of ischemic stroke, and to suggest the predominant biological pathways for each subtypes. Through 6285 protein data retrieved from PolySearch2 and STRING database, the first PPI networks for all subtypes of ischemic stroke were constructed. Notably, F2 and PLG were identified as the critical proteins for large artery atherosclerosis (LAA), lacunar, cardioembolic, stroke of other determined etiology (SOE) and stroke of undetermined etiology (SUE). Gene ontology and DAVID analysis revealed that GO:0030193 regulation of blood coagulation and GO:0051917 regulation of fibrinolysis were the important functional clusters for all the subtypes. In addition, inflammatory pathway was the key etiology for LAA and lacunar, while FOS and JAK2/STAT3 signaling pathways might contribute to cardioembolic stroke. Due to many risk factors associated with SOE and SUE, the precise etiology for these two subtypes remained to be concluded.
Collapse
Affiliation(s)
- Loo Keat Wei
- Department of Biological Science, Faculty of Science, Universiti Tunku Abdul Rahman, Bandar Barat, 31900 Kampar, Perak, Malaysia.
| | - Leong Shi Quan
- Department of Biological Science, Faculty of Science, Universiti Tunku Abdul Rahman, Bandar Barat, 31900 Kampar, Perak, Malaysia
| |
Collapse
|
22
|
Ge X, Zhang Y, Zuo Y, Israr M, Li B, Yu P, Gao G, Chang YZ, Shi Z. Transcriptomic analysis reveals the molecular mechanism of Alzheimer-related neuropathology induced by sevoflurane in mice. J Cell Biochem 2019; 120:17555-17565. [PMID: 31134678 DOI: 10.1002/jcb.29020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 04/13/2019] [Accepted: 04/18/2019] [Indexed: 12/18/2022]
Abstract
Anesthetics could induce cognitive dysfunctions, such as Alzheimer's disease in humans or mice. However, the precise molecular mechanism is unclear. Sevoflurane is a common anesthetic widely used in clinical practice. Here, we demonstrated the induction of cognitive dysfunction induced by Sev in mice to corroborate the signaling pathway and the differentially expressed genes (DEGs) followed by analyzing their functions. The cognitive function of mice was measured by the Morris water maze test. Transcriptomic data were annotated with Illumina HiSeq. 2000. Further, the changes in related proteins or genes were analyzed by western blotting and real-time quantitative polymerase chain reaction. Our results showed that Sev could cause a decline in cognitive competence in mice. The transcriptomic data indicated that adding up to 566 genes were upregulated and 1073 genes were downregulated. The genes of Plin4, Lcn2, Lrg1, Foxf1, and Ctla2a were significantly upregulated, while the genes of Arc, Npas4, Egr2, Hes5, and Cdh9 were downregulated dramatically. The Gene Ontology term with the highest enrichment of DEGs are involved in the regulation of cellular and macromolecule metabolism and cation and nucleic acid binding, respectively. The Kyoto encyclopedia of genes and genomes analysis indicated that the mitogen-activated protein kinases (MAPK) pathway was one of the most important metabolic pathways. In addition, the metabolic pathways related to cognitive function, such as the nervous system and neurodegenerative disease showed significant changes. Furthermore, we found that p38, c-Jun N-terminal kinase, and extracellular signal-regulated kinase of the MAPK signaling pathway played important roles in this process. In conclusion, these results provide the first important clues for identifying the DEGs and signaling pathways in the hippocampus due to a Sev-induced cognitive deficiency in mice.
Collapse
Affiliation(s)
- Xing Ge
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Ying Zhang
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Yong Zuo
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Muhammad Israr
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Bowen Li
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Peng Yu
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Guofen Gao
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Yan-Zhong Chang
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Zhenhua Shi
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, China
| |
Collapse
|
23
|
Sub-Toxic Human Amylin Fragment Concentrations Promote the Survival and Proliferation of SH-SY5Y Cells via the Release of VEGF and HspB5 from Endothelial RBE4 Cells. Int J Mol Sci 2018; 19:ijms19113659. [PMID: 30463298 PMCID: PMC6274958 DOI: 10.3390/ijms19113659] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 12/19/2022] Open
Abstract
Human amylin is a 37-residue peptide hormone (hA1-37) secreted by β-cells of the pancreas and, along with insulin, is directly associated with type 2 diabetes mellitus (T2DM). Amyloid deposits within the islets of the pancreas represent a hallmark of T2DM. Additionally, amylin aggregates have been found in blood vessels and/or brain of patients with Alzheimer’s disease, alone or co-deposited with β-amyloid. The purpose of this study was to investigate the neuroprotective potential of human amylin in the context of endothelial-neuronal “cross-talk”. We initially performed dose-response experiments to examine cellular toxicity (quantified by the [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] MTT assay) of different hA17–29 concentrations in endothelial cells (RBE4). In the culture medium of these cells, we also measured heat shock protein B5 (HspB5) levels by ELISA, finding that even a sub-toxic concentration of hA17–29 (3 µM) produced an increase of HspB5. Using a cell medium of untreated and RBE4 challenged for 48 h with a sub-toxic concentration of hA17–29, we determined the potential beneficial effect of their addition to the medium of neuroblastoma SH-SY5Y cells. These cells were subsequently incubated for 48 h with a toxic concentration of hA17–29 (20 µM). We found a complete inhibition of hA17–29 toxicity, potentially related to the presence in the conditioned medium not only of HspB5, but also of vascular endothelial growth factor (VEGF). Pre-treating SH-SY5Y cells with the anti-Flk1 antibody, blocking the VEGF receptor 2 (VEGFR2), significantly decreased the protective effects of the conditioned RBE4 medium. These data, obtained by indirectly measuring VEGF activity, were strongly corroborated by the direct measurement of VEGF levels in conditioned RBE4 media as detected by ELISA. Altogether, these findings highlighted a novel role of sub-toxic concentrations of human amylin in promoting the secretion of proteic factors by endothelial cells (HspB5 and VEGF) that support the survival and proliferation of neuron-like cells.
Collapse
|
24
|
Xu H, Mei XP, Xu LX. The effect of pre- and after-treatment of sevoflurane on central ischemia tolerance and the underlying mechanisms. J Dent Anesth Pain Med 2018; 18:1-8. [PMID: 29556553 PMCID: PMC5858006 DOI: 10.17245/jdapm.2018.18.1.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 01/03/2023] Open
Abstract
In recent years, with continuous research efforts targeted at studying the effects of pre- and after-treatment of inhaled anesthetics, significant progress has been made regarding the common clinical use of low concentrations of inhaled sevoflurane and its effect on induced central ischemia tolerance by pre- and post-treatment. In this study, we collected, analyzed, classified, and summarized recent literature regarding the effect of sevoflurane on central ischemia tolerance and its related mechanisms. In addition, we provide a theoretical basis for the clinical application of sevoflurane to protect the central nervous system and other important organs against ischemic injury.
Collapse
Affiliation(s)
- Hao Xu
- Institution of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Xiao-Peng Mei
- Department of Anesthesiology, the First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, China
| | - Li-Xian Xu
- Department of Anesthesiology, School of Stomatology, the Fourth Military Medical University, Xi'an, China
| |
Collapse
|