1
|
Vetter C, Meyer ER, Seidel K, Bervini D, Huber M, Krejci V. Co-administration of dexmedetomidine with total intravenous anaesthesia in carotid endarterectomy reduces requirements for propofol and improves haemodynamic stability: A single-centre, prospective, randomised controlled trial. Eur J Anaesthesiol 2024:00003643-990000000-00243. [PMID: 39529482 DOI: 10.1097/eja.0000000000002099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
BACKGROUND Total intravenous anaesthesia guided by electroencephalography and neurophysiological monitoring may be used for carotid endarterectomy. Reduction of brain metabolic demand during cross-clamping of the internal carotid artery with propofol titrated to burst suppression requires effect-site concentrations that may delay emergence and interfere with intraoperative neurophysiological monitoring. OBJECTIVE To test the hypothesis that dexmedetomidine decreases the effect-site concentration of propofol required for burst-suppression in patients undergoing carotid endarterectomy. DESIGN Randomised controlled trial. PARTICIPANTS Patients undergoing carotid endarterectomy. SETTING University Hospital of Berne, Switzerland, from October 2018 to September 2024. INTERVENTIONS Patients were randomised into a control (n = 23) and a dexmedetomidine groups (n = 22). Total intravenous anaesthesia was administered to both groups. Patients in the dexmedetomidine group received an intravenous bolus of dexmedetomidine (0.4 μg kg-1 over 10 min) before induction, followed by a continuous intravenous infusion (0.4 μg kg-1 h-1). The effect-site concentrations of propofol were titrated against frontal electroencephalography parameters. Burst suppression was induced with propofol during cross-clamping of the internal carotid artery. OUTCOME MEASURES The primary outcome was the effect-site concentration of propofol required for burst-suppression. The secondary outcomes were the requirement for vasoactive substances, neurophysiological monitoring parameters, and postoperative delirium. RESULTS The effect-site concentration of propofol required for burst suppression was 4.0 μg ml-1 [3.50 to 4.90] (median [interquartile range]) in the dexmedetomidine group compared with 6.0 μg ml-1 [5.5 to 7.3] in the control group (P < 0.001). Less norepinephrine was required in the dexmedetomidine group (total 454 μg [246 to 818] compared with 1000 μg [444 to 1326] (P = 0.015) in the control group). Dexmedetomidine did not affect intraoperative neurophysiological monitoring. CONCLUSION Co-administration of dexmedetomidine to total intravenous anaesthesia for carotid endarterectomy decreased the effect-site concentrations of propofol required for burst suppression by 33%. The propofol-sparing effect and peripheral alpha-agonism of dexmedetomidine may explain the reduced requirement for vasopressors. TRIAL REGISTRATION Clinicaltrials.gov identifier: NCT04662177.
Collapse
Affiliation(s)
- Christian Vetter
- From the Department of Anaesthesiology and Pain Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland (CV, ERM, MH, VK), and Department of Neurosurgery, Inselspital, University Hospital, University of Bern, Bern, Switzerland (KS, DB)
| | | | | | | | | | | |
Collapse
|
2
|
Wi W, Kim HJ, Bang S, Kweon OJ, Kim D, Oh EJ. Effect of intravenous versus inhaled anesthetics on blood-brain barrier dysfunction and neuroinflammation in elderly patients undergoing major surgery: study protocol of a randomized controlled trial. Trials 2024; 25:684. [PMID: 39415284 PMCID: PMC11481368 DOI: 10.1186/s13063-024-08515-w] [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: 06/19/2024] [Accepted: 09/27/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is one of the major complications after surgery, with devastating clinical outcomes. Although POCD is a condition with a multifactorial pathophysiology, blood-brain barrier (BBB) dysfunction and neuronal injury have been shown to play a critical role, especially in the elderly. Previous studies have demonstrated that the choice of anesthetics affect BBB permeability and neuroinflammation. However, most studies are carried out on animals, with limited research undertaken on humans. Therefore, we will compare the effect of intravenous anesthetics and inhaled anesthetics on BBB dysfunction and the change of inflammatory markers after surgery. METHODS One hundred and fifty-four patients who are 60 years of age or older undergoing major surgery for more than 2 h will be randomly allocated to two anesthetics groups (intravenous, inhaled) in a 1:1 ratio. In the intravenous anesthetics group (group P), propofol will be infused with a target-controlled infusion (TCI) system throughout the entire surgery. In the inhaled anesthetics group (group G), bolus injection of propofol will be administered for loss of consciousness, and simultaneously sevoflurane will be initiated for the maintenance of anesthesia. The primary outcome is the change in serum S100 calcium binding protein β (S100β) at four time points: before induction of anesthesia, at the end of surgery, 4 h after surgery, postoperative day 1. Secondary outcomes include changes in the inflammatory markers, serum interleukin (IL)-6, IL-8, tumor necrosis factor (TNF)-α, and C-reactive protein; the incidence of delirium; and the change in the cognitive function between groups. In patients pre-scheduled for postoperative intensive care unit admission, the cerebrospinal fluid/serum albumin quotient (Qalb) between the two groups will be compared before and after surgery, and change in inflammatory markers in serum and CSF will be analyzed in relation to the Qalb. DISCUSSION The current study will compare the effect of intravenous versus inhaled anesthetics on blood-brain barrier permeability and, as a result, the difference in neuroinflammation in elderly patients. Also, the study results will provide additional information to develop intraoperative anesthetic strategies to reduce POCD. TRIAL REGISTRATION The trial was prospectively registered at Clinical Trials protocol registration with identifier 2310-117-126 on April 9, 2024.
Collapse
Affiliation(s)
- Wongook Wi
- Department of Anesthesiology and Pain Medicine, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, 110 Deokan-Ro, Gwangmyeong-Si, Gyeonggi-Do, Republic of Korea
| | - Hyo-Jin Kim
- Department of Anesthesiology and Pain Medicine, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, 110 Deokan-Ro, Gwangmyeong-Si, Gyeonggi-Do, Republic of Korea
| | - Sira Bang
- Department of Anesthesiology and Pain Medicine, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, 110 Deokan-Ro, Gwangmyeong-Si, Gyeonggi-Do, Republic of Korea
| | - Oh Joo Kweon
- Department of Laboratory Medicine, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong-Si, Gyeonggi-Do, Republic of Korea
| | - Doyeon Kim
- Department of Anesthesiology and Pain Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Republic of Korea
| | - Eun Jung Oh
- Department of Anesthesiology and Pain Medicine, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, 110 Deokan-Ro, Gwangmyeong-Si, Gyeonggi-Do, Republic of Korea.
| |
Collapse
|
3
|
Wisniewski AM, Chancellor WZ, Young A, Money D, Beller JP, Charlton J, Lunardi N, Yang Z, Laubach VE, Mehaffey JH, Kron IL, Roeser ME. Adenosine 2A Receptor Agonism Improves Survival in Extracorporeal Cardiopulmonary Resuscitation. J Surg Res 2024; 301:404-412. [PMID: 39029264 DOI: 10.1016/j.jss.2024.06.033] [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: 02/21/2024] [Revised: 05/20/2024] [Accepted: 06/22/2024] [Indexed: 07/21/2024]
Abstract
INTRODUCTION Despite resuscitation advances including extracorporeal cardiopulmonary resuscitation (ECPR), freedom from neurologic and myocardial insult after cardiac arrest remains unlikely. We hypothesized that adenosine 2A receptor (A2AR) agonism, which attenuates reperfusion injury, would improve outcomes in a porcine model of ECPR. METHODS Adult swine underwent 20 min of circulatory arrest followed by defibrillation and 6 h of ECPR. Animals were randomized to receive saline vehicle or A2AR agonist (ATL1223 or Regadenoson) infusion during extracorporeal membrane oxygenation. Animals were weaned off extracorporeal membrane oxygenation and monitored for 24 h. Clinical and biochemical end points were compared. RESULTS The administration of A2AR agonists increased survival (P = 0.01) after cardiac arrest compared to vehicle. Markers of neurologic damage including S100 calcium binding protein B and glial fibrillary acidic protein were significantly lower with A2AR agonist treatment. CONCLUSIONS In a model of cardiac arrest treated with ECPR, A2AR agonism increased survival at 24 h and reduced neurologic damage suggesting A2AR activation may be a promising therapeutic target after cardiac arrest.
Collapse
Affiliation(s)
- Alex M Wisniewski
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - William Z Chancellor
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Andrew Young
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Dustin Money
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Jared P Beller
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Jennifer Charlton
- Department of Pediatrics, University of Virginia Health System, Charlottesville, Virginia
| | - Nadia Lunardi
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, Virginia
| | - Zequan Yang
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Victor E Laubach
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - J Hunter Mehaffey
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Irving L Kron
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Mark E Roeser
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia.
| |
Collapse
|
4
|
Wang K, Wang Y, Zhang T, Chang B, Fu D, Chen X. The Role of Intravenous Anesthetics for Neuro: Protection or Toxicity? Neurosci Bull 2024:10.1007/s12264-024-01265-4. [PMID: 39153174 DOI: 10.1007/s12264-024-01265-4] [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: 12/15/2023] [Accepted: 03/15/2024] [Indexed: 08/19/2024] Open
Abstract
The primary intravenous anesthetics employed in clinical practice encompass dexmedetomidine (Dex), propofol, ketamine, etomidate, midazolam, and remimazolam. Apart from their established sedative, analgesic, and anxiolytic properties, an increasing body of research has uncovered neuroprotective effects of intravenous anesthetics in various animal and cellular models, as well as in clinical studies. However, there also exists conflicting evidence pointing to the potential neurotoxic effects of these intravenous anesthetics. The role of intravenous anesthetics for neuro on both sides of protection or toxicity has been rarely summarized. Considering the mentioned above, this work aims to offer a comprehensive understanding of the underlying mechanisms involved both in the central nerve system (CNS) and the peripheral nerve system (PNS) and provide valuable insights into the potential safety and risk associated with the clinical use of intravenous anesthetics.
Collapse
Affiliation(s)
- Kaixin Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China
| | - Bingcheng Chang
- The Second Affiliated Hospital of Guizhou, University of Traditional Chinese Medicine, Guiyang, 550003, China
| | - Daan Fu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China.
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430022, China.
| |
Collapse
|
5
|
Jia Y, Feng Y, Ma Y, Feng G, Xu N, Li M, Liu M, Fan Z, Wang T. Type of anesthesia for endovascular therapy in acute ischemic stroke: A literature review and meta-analysis. Int J Stroke 2024; 19:735-746. [PMID: 38234158 DOI: 10.1177/17474930241228956] [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: 01/19/2024]
Abstract
BACKGROUND Endovascular thrombectomy (EVT) has been proven as the standard treatment for acute ischemic stroke (AIS) patients due to large vessel occlusion (LVO). However, the ideal anesthetic strategy during EVT still remains unclear. Therefore, this systematic review and meta-analysis aimed to determine the optimal anesthetic modality for patients with AIS undergoing EVT based on current randomized controlled trials (RCTs). METHODS The databases Medline (via PubMed), EMBASE, Web of Science, and the Cochrane Library were searched for RCTs comparing general anesthesia (GA) and conscious sedation (CS) in AIS patients undergoing EVT. The primary outcome was a favorable functional outcome at 90 days postintervention. Data analysis was conducted using the Review Manager software (RevMan V.5.3). RESULTS Eight RCTs involving 1199 patients were included. There was no significant difference between GA and CS group in the rate of functional independence (risk ratio (RR) = 1.10, 95% confidence interval (CI) = 0.96 to 1.25; p = 0.17; I2 = 30%). Compared with the CS group, the GA group attained a higher successful recanalization rate (RR = 1.14, 95% CI = 1.08 to 1.20; p < 0.00001; I2 = 17%). In addition, patients in the GA were associated with a higher rate of hypotension (RR = 1.87, 95% CI = 1.44 to 2.41; p < 0.00001; I2 = 66%) and a higher incidence of pneumonia (RR = 1.38, 95% CI = 1.05 to 1.8; p = 0.02; I2 = 37%). CONCLUSION For AIS patients receiving EVT, the choice of anesthetic modality did not influence the 3-month neurological outcome while GA is superior to CS in terms of successful reperfusion rate. Moreover, the patients in the GA group were at a higher risk of developing hypotension and pneumonia. Further studies are required to provide more sufficient evidence.
Collapse
Affiliation(s)
- Yitong Jia
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yao Feng
- Department of Neurology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yanhui Ma
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Guang Feng
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Na Xu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Meng Li
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Miao Liu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhen Fan
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tianlong Wang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Zhang J, Wang S, Zhang H, Yang Y, Yuan M, Yang X, Wen Y. The role of the AMPK/ERK1/2 signaling pathway in neuronal oxidative stress damage following cerebral ischemia-reperfusion. Tissue Cell 2024; 89:102472. [PMID: 39003914 DOI: 10.1016/j.tice.2024.102472] [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: 03/27/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
Cerebral ischemia-reperfusion injury involves a series of pathophysiological processes that occur when blood supply is restored after cerebral vascular obstruction, leading to neuronal damage. The AMPK/ERK1/2 signaling pathway has been identified as crucial in this process, although the exact mechanisms underlying the induction of ischemia-reperfusion injury remain unclear. In this study, we investigated the involvement of the AMPK/ERK1/2 signaling pathway in neuronal oxidative stress damage following cerebral ischemia-reperfusion by establishing animal and cell models. Our experimental results demonstrated that cerebral ischemia-reperfusion leads to oxidative stress damage, including cell apoptosis and mitochondrial dysfunction. Moreover, further experiments showed that inhibition of AMPK and ERK1/2 activity, using U0126 and Compound C respectively, could alleviate oxidative stress-induced cellular injury, improve mitochondrial morphology and function, reduce reactive oxygen species levels, increase superoxide dismutase levels, and suppress apoptosis. These findings clearly indicate the critical role of the AMPK/ERK1/2 signaling pathway in regulating oxidative stress damage and cerebral ischemia-reperfusion injury. The discoveries in this study provide a theoretical basis for further research and development of neuroprotective therapeutic strategies targeting the AMPK/ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Jiejie Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China; Key Laboratory of Clinical Neurology Ministry of Education, Shijiazhuang, Hebei, China
| | - Shan Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China; Key Laboratory of Clinical Neurology Ministry of Education, Shijiazhuang, Hebei, China
| | - Haitao Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China; Key Laboratory of Clinical Neurology Ministry of Education, Shijiazhuang, Hebei, China
| | - Yihan Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China; Key Laboratory of Clinical Neurology Ministry of Education, Shijiazhuang, Hebei, China
| | - Mu Yuan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China; Key Laboratory of Clinical Neurology Ministry of Education, Shijiazhuang, Hebei, China
| | - Xiaotong Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China; Key Laboratory of Clinical Neurology Ministry of Education, Shijiazhuang, Hebei, China
| | - Ya Wen
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China; Key Laboratory of Clinical Neurology Ministry of Education, Shijiazhuang, Hebei, China.
| |
Collapse
|
7
|
Shi Y, Deng H, Zhang Z, Zhu X, Zeng Z. Remimazolam protects the liver from ischemia-reperfusion injury by inhibiting the MAPK/ERK pathway. BMC Anesthesiol 2024; 24:251. [PMID: 39054453 PMCID: PMC11270846 DOI: 10.1186/s12871-024-02641-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Ischemia-reperfusion (I/R) injury is a major factor in liver damage following hepatic resection and liver transplantation, with anesthetics demonstrating the ability to shield organs from this type of injury. METHODS Hypoxia-reoxygenation (H/R) was used to create in vitro I/R hepatocyte cell injury models. The CCK-8 assay, flow cytometer, LDH assay, and ELSIA were utilized to assess hepatocyte injury. The in vivo I/R injury rat model was then built. HE and TUNEL staining were used to assess liver tissue damage. Western-blot was applied to assess the activation of the MAPK/ERK pathway. RESULTS Remimazolam (RMZL) remarkably improved cell viability and decreased apoptosis in H/R-induced hepatocyte injury. RMZL reduced the release of H/R-induced inflammatory mediators (TNF-α and IL-6) as well as LDH levels. We also discovered that RMZL inhibited p38 and ERK1/2 phosphorylation in vivo and in vitro. The stimulation of MAPK/ERK, on the other hand, abolished RMZL's anti-inflammation effects in H/R-induced hepatocyte injury. Furthermore, RMZL reduced liver tissue injury in I/R rats. CONCLUSION RMZL prevented hepatic I/R damage by inhibiting MAPK/ERK signaling.
Collapse
Affiliation(s)
- Yanhua Shi
- Department of Anesthesiology, Chenzhou First People's Hospital, No.102 Luojiajing, Beihu District, Chenzhou City, 423000, Hunan Province, People's Republic of China
| | - Housheng Deng
- Department of Anesthesiology, Chenzhou First People's Hospital, No.102 Luojiajing, Beihu District, Chenzhou City, 423000, Hunan Province, People's Republic of China
| | - Zhiming Zhang
- Department of Anesthesiology, Chenzhou First People's Hospital, No.102 Luojiajing, Beihu District, Chenzhou City, 423000, Hunan Province, People's Republic of China
| | - Xiaoling Zhu
- Department of Anesthesiology, Chenzhou First People's Hospital, No.102 Luojiajing, Beihu District, Chenzhou City, 423000, Hunan Province, People's Republic of China
| | - Zhiqin Zeng
- Department of Anesthesiology, Chenzhou First People's Hospital, No.102 Luojiajing, Beihu District, Chenzhou City, 423000, Hunan Province, People's Republic of China.
| |
Collapse
|
8
|
Gundersen JK, Chakkarapani E, Menassa DA, Walløe L, Thoresen M. The effects of anaesthesia on cell death in a porcine model of neonatal hypoxic-ischaemic brain injury. BJA OPEN 2024; 10:100283. [PMID: 38741692 PMCID: PMC11089311 DOI: 10.1016/j.bjao.2024.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/30/2024] [Indexed: 05/16/2024]
Abstract
Background Hypothermia is neuroprotective after neonatal hypoxic-ischaemic brain injury. However, systemic cooling to hypothermic temperatures is a stressor and may reduce neuroprotection in awake pigs. We compared two experiments of global hypoxic-ischaemic injury in newborn pigs, in which one group received propofol-remifentanil and the other remained awake during post-insult hypothermia treatment. Methods In both studies, newborn pigs were anaesthetised using halothane during a 45-min global hypoxic-ischaemic insult induced by reducing Fio2 and graded hypotension until a low-voltage <7 μV electroencephalogram was achieved. On reoxygenation, the pigs were randomly allocated to receive 24 h of normothermia or hypothermia. In the first study (n=18) anaesthesia was discontinued and the pigs' tracheas were extubated. In the second study (n=14) anaesthesia was continued using propofol and remifentanil. Brain injury was assessed after 72 h by classical global histopathology, Purkinje cell count, and apoptotic cell counts in the hippocampus and cerebellum. Results Global injury was nearly 10-fold greater in the awake group compared with the anaesthetised group (P=0.021). Hypothermia was neuroprotective in the anaesthetised pigs but not the awake pigs. In the hippocampus, the density of cleaved caspase-3-positive cells was increased in awake compared with anaesthetised pigs in normothermia. In the cerebellum, Purkinje cell density was reduced in the awake pigs irrespective of treatment, and the number of cleaved caspase-3-positive Purkinje cells was greatly increased in hypothermic awake pigs. We detected no difference in cleaved caspase-3 in the granular cell layer or microglial reactivity across the groups. Conclusions Our study provides novel insights into the significance of anaesthesia/sedation during hypothermia for achieving optimal neuroprotection.
Collapse
Affiliation(s)
- Julia K. Gundersen
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | - Ela Chakkarapani
- Translational Health Sciences, St. Michael's Hospital, Bristol Medical School, University of Bristol, Bristol, UK
| | - David A. Menassa
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Neuropathology and The Queen's College, University of Oxford, Oxford, UK
- Department of Women's & Children's Health, Karolinska Institutet, Solna, Sweden
| | - Lars Walløe
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Marianne Thoresen
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Translational Health Sciences, St. Michael's Hospital, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
9
|
Ebrahimi M, Dabbagh A, Madadi F. Propofol-induced hippocampal Neurotoxicity: A mitochondrial perspective. Brain Res 2024; 1831:148841. [PMID: 38428475 DOI: 10.1016/j.brainres.2024.148841] [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: 01/19/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
Propofol is a frequently used anesthetic. It can induce neurodegeneration and inhibit neurogenesis in the hippocampus. This effect may be temporary. It can, however, become permanent in vulnerable populations, such as the elderly, who are more susceptible to Alzheimer's disease, and neonates and children, whose brains are still developing and require neurogenesis. Current clinical practice strategies have failed to provide an effective solution to this problem. In addition, the molecular mechanism of this toxicity is not fully understood. Recent advances in molecular research have revealed that apoptosis, in close association with mitochondria, is a crucial mechanism through which propofol contributes to hippocampal toxicity. Preventing the toxicity of propofol on the hippocampus has shown promise in in-vivo, in-vitro, and to a lesser extent human studies. This study seeks to provide a comprehensive literature review of the effects of propofol toxicity on the hippocampus via mitochondria and to suggest translational suggestions based on these molecular results.
Collapse
Affiliation(s)
- Moein Ebrahimi
- Department of Anesthesiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Anesthesiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Dabbagh
- Department of Anesthesiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Anesthesiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Firoozeh Madadi
- Department of Anesthesiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Anesthesiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Huang L, Tuzer F, Murtha A, Green M, Torres C, Liu H, Malaeb S. Differential Susceptibility to Propofol and Ketamine in Primary Cultures of Young and Senesced Astrocytes. Anesthesiol Res Pract 2024; 2024:8876548. [PMID: 38633620 PMCID: PMC11023735 DOI: 10.1155/2024/8876548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/28/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
The adverse effects of general anesthesia on the long-term cognition of young children and senior adults have become of concern in recent years. Previously, mechanistic and pathogenic investigations focused on neurons, and little is known about the effect of commonly used intravenous anesthetics such as propofol and ketamine on astrocytes. Recently, astrocyte dysfunction has been implicated in a wide range of age-related brain diseases. In this study, we examined the survival and viability of both young and senescent astrocytes in culture after adding propofol and ketamine to the media at varying strengths. Oxidative stimulus was applied to commercially available fetal cell lines of human astrocytes in vitro to induce morphological changes in cellular senescence. Our results indicate that propofol reduces the survival of young astrocytes as compared to controls, as well as to ketamine. These effects were seen in comparisons of total cell count and at both high and low dose concentrations. High doses of propofol also significantly reduced cell viability compared to those exposed to baseline controls and ketamine. Senescent astrocytes, on the other hand, demonstrated cell count reductions as compared to baseline controls and ketamine when exposed to either DMSO or propofol. The data show differential susceptibility of young astrocytes to propofol than to ketamine. The observed cell count reduction may be related to the adverse effects of propofol on mitochondrial function and free radical production, as described in previous studies. We speculate that ketamine may have a more favorable safety profile in infants and young children.
Collapse
Affiliation(s)
- Liang Huang
- Department of Anesthesiology and Perioperative Medicine, New York University Langone Health, Grossman School of Medicine, New York, NY, USA
| | - Ferit Tuzer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Abigail Murtha
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Michael Green
- Department of Anesthesiology and Perioperative Medicine, Thomas Jefferson University, Sidney Kimmel Medical College, Philadelphia, PA, USA
| | - Claudio Torres
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Henry Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, Philadelphia, PA, USA
| | - Shadi Malaeb
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
11
|
Everson CA, Szabo A, Plyer C, Hammeke TA, Stemper BD, Budde MD. Sleep loss, caffeine, sleep aids and sedation modify brain abnormalities of mild traumatic brain injury. Exp Neurol 2024; 372:114620. [PMID: 38029810 DOI: 10.1016/j.expneurol.2023.114620] [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: 08/23/2023] [Revised: 11/06/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
Little evidence exists about how mild traumatic brain injury (mTBI) is affected by commonly encountered exposures of sleep loss, sleep aids, and caffeine that might be potential therapeutic opportunities. In addition, while propofol sedation is administered in severe TBI, its potential utility in mild TBI is unclear. Each of these exposures is known to have pronounced effects on cerebral metabolism and blood flow and neurochemistry. We hypothesized that they each interact with cerebral metabolic dynamics post-injury and change the subclinical characteristics of mTBI. MTBI in rats was produced by head rotational acceleration injury that mimics the biomechanics of human mTBI. Three mTBIs spaced 48 h apart were used to increase the likelihood that vulnerabilities induced by repeated mTBI would be manifested without clinically relevant structural damage. After the third mTBI, rats were immediately sleep deprived or administered caffeine or suvorexant (an orexin antagonist and sleep aid) for the next 24 h or administered propofol for 5 h. Resting state functional magnetic resonance imaging (rs-fMRI) and diffusion tensor imaging (DTI) were performed 24 h after the third mTBI and again after 30 days to determine changes to the brain mTBI phenotype. Multi-modal analyses on brain regions of interest included measures of functional connectivity and regional homogeneity from rs-fMRI, and mean diffusivity (MD) and fractional anisotropy (FA) from DTI. Each intervention changed the mTBI profile of subclinical effects that presumably underlie healing, compensation, damage, and plasticity. Sleep loss during the acute post-injury period resulted in dramatic changes to functional connectivity. Caffeine, propofol sedation and suvorexant were especially noteworthy for differential effects on microstructure in gray and white matter regions after mTBI. The present results indicate that commonplace exposures and short-term sedation alter the subclinical manifestations of repeated mTBI and therefore likely play roles in symptomatology and vulnerability to damage by repeated mTBI.
Collapse
Affiliation(s)
- Carol A Everson
- Department of Medicine (Endocrinology and Molecular Medicine) and Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Aniko Szabo
- Division of Biostatistics, Institute for Health & Equity, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Cade Plyer
- Neurology Residency Program, Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
| | - Thomas A Hammeke
- Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian D Stemper
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA; Neuroscience Research, Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA.
| | - Mathew D Budde
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
12
|
Yuhan L, Khaleghi Ghadiri M, Gorji A. Impact of NQO1 dysregulation in CNS disorders. J Transl Med 2024; 22:4. [PMID: 38167027 PMCID: PMC10762857 DOI: 10.1186/s12967-023-04802-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
NAD(P)H Quinone Dehydrogenase 1 (NQO1) plays a pivotal role in the regulation of neuronal function and synaptic plasticity, cellular adaptation to oxidative stress, neuroinflammatory and degenerative processes, and tumorigenesis in the central nervous system (CNS). Impairment of the NQO1 activity in the CNS can result in abnormal neurotransmitter release and clearance, increased oxidative stress, and aggravated cellular injury/death. Furthermore, it can cause disturbances in neural circuit function and synaptic neurotransmission. The abnormalities of NQO1 enzyme activity have been linked to the pathophysiological mechanisms of multiple neurological disorders, including Parkinson's disease, Alzheimer's disease, epilepsy, multiple sclerosis, cerebrovascular disease, traumatic brain injury, and brain malignancy. NQO1 contributes to various dimensions of tumorigenesis and treatment response in various brain tumors. The precise mechanisms through which abnormalities in NQO1 function contribute to these neurological disorders continue to be a subject of ongoing research. Building upon the existing knowledge, the present study reviews current investigations describing the role of NQO1 dysregulations in various neurological disorders. This study emphasizes the potential of NQO1 as a biomarker in diagnostic and prognostic approaches, as well as its suitability as a target for drug development strategies in neurological disorders.
Collapse
Affiliation(s)
- Li Yuhan
- Epilepsy Research Center, Münster University, Münster, Germany
- Department of Breast Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Ali Gorji
- Epilepsy Research Center, Münster University, Münster, Germany.
- Department of Neurosurgery, Münster University, Münster, Germany.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Zhao R, Teng X, Yang Y. Calpain as a Therapeutic Target for Hypoxic-Ischemic Encephalopathy. Mol Neurobiol 2024; 61:533-540. [PMID: 37642934 DOI: 10.1007/s12035-023-03594-3] [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: 03/23/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a complex pathophysiological process with multiple links and factors. It involves the interaction of inflammation, oxidative stress, and glucose metabolism, and results in acute and even long-term brain damage and impairment of brain function. Calpain is a family of Ca2+-dependent cysteine proteases that regulate cellular function. Calpain activation is involved in cerebral ischemic injury, and this involvement is achieved by the interaction among Ca2+, substrates, organelles, and multiple proteases in the neuronal necrosis and apoptosis pathways after cerebral ischemia. Many calpain inhibitors have been developed and tested in the biochemical and biomedical fields. This study reviewed the potential role of calpain in the treatment of HIE and related mechanism, providing new insights for future research on HIE.
Collapse
Affiliation(s)
- Ruiyang Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Xiufei Teng
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Yanchao Yang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
14
|
Zhu X, An X, Chen M, Guo D, Xie P, Wang B, Huang Z, Yu W. Seipin overexpression attenuates cerebral ischemia-reperfusion injury via preventing apoptosis and autophagy. Brain Behav 2023; 13:e3195. [PMID: 37897134 PMCID: PMC10726895 DOI: 10.1002/brb3.3195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Ischemic cerebrovascular disease (ICVD) is one of three fatal diseases in humans, along with heart disease and malignant tumors. Cerebral ischemia/reperfusion injury (CI/RI) is the primary cause of ICVD. Recently, seipin was reported to be crucial for lipid droplet formation, hepatic steatosis, and axonal atrophy. However, the function and mechanism of seipin in CI/RI has not been explicitly stated. METHODS Oxygen-glucose deprivation/reoxygenation (OGD/R) hippocampal neuron cell line (HT-22) and middle cerebral artery occlusion (MCAO) in rats were established. The levels of apoptosis- and autophagy-related proteins and seipin were confirmed by western blot. Cell proliferation was assessed with CCK-8, and ischemic infarction and pathological structure were monitored by TTC and H&E staining, and tissue apoptosis was assessed through TUNEL assay. RESULTS The proliferative activity was decreased, and apoptosis and autophagy pathways could also be induced in the OGD/R HT-22 cells. Seipin overexpression accelerated viability and inhibited apoptosis and autophagy in the OGD/R HT-22 cells. Moreover, the data revealed that seipin overexpression could also attenuate cerebral infarction, apoptosis. Autophagy pathways could be repressed by seipin in the MCAO rats. CONCLUSION Seipin has a protective role against CI/RI in rats, and its mechanism might be relevant to the suppression of apoptosis and autophagy, suggesting that seipin might be a latent target for CI/RI.
Collapse
Affiliation(s)
- Xiaoxi Zhu
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
- Cell engineering LaboratoryAffiliated Hospital of Zunyi Medical UniversityZunyi CityChina
| | - Xiaoqiong An
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| | - Ming Chen
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| | - Dongfen Guo
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| | - Peng Xie
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| | - Bi Wang
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| | - Zhi Huang
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| | - Wenfeng Yu
- Key Laboratory of Molecular BiologySchool of Basic Medical Science of Guizhou Medical UniversityGuiyang CityChina
| |
Collapse
|
15
|
Margaritelis NV. Personalized redox biology: Designs and concepts. Free Radic Biol Med 2023; 208:112-125. [PMID: 37541453 DOI: 10.1016/j.freeradbiomed.2023.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/19/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
Personalized interventions are regarded as a next-generation approach in almost all fields of biomedicine, such as clinical medicine, exercise, nutrition and pharmacology. At the same time, an increasing body of evidence indicates that redox processes regulate, at least in part, multiple aspects of human physiology and pathology. As a result, the idea of applying personalized redox treatments to improve their efficacy has gained popularity among researchers in recent years. The aim of the present primer-style review was to highlight some crucial yet underappreciated methodological, statistical, and interpretative concepts within the redox biology literature, while also providing a physiology-oriented perspective on personalized redox biology. The topics addressed are: (i) the critical issue of investigating the potential existence of inter-individual variability; (ii) the importance of distinguishing a genuine and consistent response of a subject from a chance finding; (iii) the challenge of accurately quantifying the effect of a redox treatment when dealing with 'extreme' groups due to mathematical coupling and regression to the mean; and (iv) research designs and analyses that have been implemented in other fields, and can be reframed and exploited in a redox biology context.
Collapse
Affiliation(s)
- Nikos V Margaritelis
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Agios Ioannis, 62122, Serres, Greece.
| |
Collapse
|
16
|
Yang K, Ma Y, Xie C, He L, Zhao H, Dai Z, Wang X. Dexmedetomidine combined with propofol attenuates myocardial ischemia/reperfusion injury by activating the AMPK signaling pathway. Heliyon 2023; 9:e22054. [PMID: 38034796 PMCID: PMC10682120 DOI: 10.1016/j.heliyon.2023.e22054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
Objective Myocardial ischemia/reperfusion (MI/R) injury is a major cause of cardiac tissue damage, with high disability and death rates. Although both dexmedetomidine (Dex) and propofol (PPF) have been indicated to alleviate MI/R injury in rat models, the effects of the combined use of these two drugs remain unclear. This study aimed to investigate the combined effects of Dex and PPF against MI/R injury and related mechanisms. Methods A rat model of MI/R injury was established and used to explore the combined effects of Dex and PPF on MI/R injury. Hematoxylin-eosin (HE) and Masson staining were used for histopathological evaluation. 2,3,5-triphenyltetrazolium chloride (TTC), echocardiography, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining were used to determine myocardial infarction size, cardiac function, and apoptosis, respectively. Enzyme-linked immunosorbent assay (ELISA) was performed to assess myocardial function and oxidative stress (OS). Autophagy was observed through transmission electron microscopy. Moreover, western blotting was conducted to detect autophagy markers and the AMPK pathway. Results The combination of Dex and PPF alleviated histopathological injury, reduced myocardial infarction, and rescued cardiac dysfunction in MI/R rats. Furthermore, Dex combined with PPF decreased the levels of MDA and ROS and increased the SOD level in MI/R rats. Besides, Dex combined with PPF inhibited myocardial apoptosis in MI/R rats. After combined treatment with Dex and PPF, the number of autophagosomes, expression levels of Beclin-1 and LC3II/LC3I were elevated, while the expression levels of p62 were reduced in MI/R rats. The combined use of Dex and PPF activated the AMPK pathway in MI/R rats. Compound C (an AMPK inhibitor) could abolish the combined effects of Dex and PPF on alleviating myocardial injury and enhancing autophagy in MI/R rats. Conclusion The combination of Dex and PPF attenuated MI/R injury in rats, which may be associated with the activation of the AMPK signaling pathway.
Collapse
Affiliation(s)
| | | | - Chunmei Xie
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, 650000, China
| | - Lixian He
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, 650000, China
| | - Haoxing Zhao
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, 650000, China
| | - Zheng Dai
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, 650000, China
| | - Xiaoqi Wang
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, 650000, China
| |
Collapse
|
17
|
Wu X, Deng J, Li X, Yang L, Zhao G, Yin Q, Shi Y, Tong Z. Effects of Propofol on Perioperative Sleep Quality in Patients Undergoing Gastrointestinal Endoscopy: A Prospective Cohort Study. J Perianesth Nurs 2023; 38:787-791. [PMID: 37269278 DOI: 10.1016/j.jopan.2023.02.001] [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: 07/14/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 06/05/2023]
Abstract
PURPOSE Some patients experience sleep disturbances after endoscopy performed under sedation. This study aimed to evaluate the effects of propofol on sleep quality after gastrointestinal endoscopy (GE). DESIGN This study was a prospective cohort study. METHODS This study enrolled 880 patients who underwent GE. Patients who chose to undergo GE under sedation received intravenous propofol, whereas the control group did not. The Pittsburgh Sleep Quality Index (PSQI) was measured before GE (PSQI-1) and 3 weeks (PSQI-2) after GE. The Groningen Sleep Score Scale (GSQS) was used before GE (GSQS-1) and 1 (GSQS-2) and 7 days (GSQS-3) after GE. FINDINGS There was a significant increase in GSQS scores from baseline to days 1 and 7 after GE (GSQS-2 vs GSQS-1, P < .001, GSQS-3 vs GSQS-1, P = .008). However, no significant changes were observed in the control group (GSQS-2 vs GSQS-1, P = .38, GSQS-3 vs GSQS-1, P = .66). On day 21, there were no significant changes in the baseline PSQI scores over time in either group (sedation group, P = .96; control group, P = .95). CONCLUSIONS GE with propofol sedation negatively affected sleep quality for 7 days after GE but not 3 weeks after GE.
Collapse
Affiliation(s)
- Xiaofei Wu
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Jinhe Deng
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaona Li
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Li Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Gaofeng Zhao
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Qing Yin
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Yongyong Shi
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhilan Tong
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
18
|
Troise D, Infante B, Mercuri S, Netti GS, Ranieri E, Gesualdo L, Stallone G, Pontrelli P. Hypoxic State of Cells and Immunosenescence: A Focus on the Role of the HIF Signaling Pathway. Biomedicines 2023; 11:2163. [PMID: 37626660 PMCID: PMC10452839 DOI: 10.3390/biomedicines11082163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/20/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Hypoxia activates hypoxia-related signaling pathways controlled by hypoxia-inducible factors (HIFs). HIFs represent a quick and effective detection system involved in the cellular response to insufficient oxygen concentration. Activation of HIF signaling pathways is involved in improving the oxygen supply, promoting cell survival through anaerobic ATP generation, and adapting energy metabolism to meet cell demands. Hypoxia can also contribute to the development of the aging process, leading to aging-related degenerative diseases; among these, the aging of the immune system under hypoxic conditions can play a role in many different immune-mediated diseases. Thus, in this review we aim to discuss the role of HIF signaling pathways following cellular hypoxia and their effects on the mechanisms driving immune system senescence.
Collapse
Affiliation(s)
- Dario Troise
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy; (D.T.); (B.I.); (S.M.); (G.S.)
| | - Barbara Infante
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy; (D.T.); (B.I.); (S.M.); (G.S.)
| | - Silvia Mercuri
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy; (D.T.); (B.I.); (S.M.); (G.S.)
| | - Giuseppe Stefano Netti
- Clinical Pathology Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (G.S.N.); (E.R.)
| | - Elena Ranieri
- Clinical Pathology Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (G.S.N.); (E.R.)
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy; (D.T.); (B.I.); (S.M.); (G.S.)
| | - Paola Pontrelli
- Nephrology, Dialysis and Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, Policlinico, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| |
Collapse
|
19
|
Li S, Hou Q, Wang R, Hou Y, Wang Q, Zhang B, Ni C, Zheng H. Sevoflurane upregulates neuron death process-related Ddit4 expression by NMDAR in the hippocampus. Aging (Albany NY) 2023; 15:5698-5712. [PMID: 37348034 PMCID: PMC10333074 DOI: 10.18632/aging.204822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/26/2023] [Indexed: 06/24/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is a serious and common complication induced by anesthesia and surgery. Neuronal apoptosis induced by general anesthetic neurotoxicity is a high-risk factor. However, a comprehensive analysis of general anesthesia-regulated gene expression patterns and further research on molecular mechanisms are lacking. Here, we performed bioinformatics analysis of gene expression in the hippocampus of aged rats that received sevoflurane anesthesia in GSE139220 from the GEO database, found a total of 226 differentially expressed genes (DEGs) and investigated hub genes according to the number of biological processes in which the genes were enriched and performed screening by 12 algorithms with cytoHubba in Cytoscape. Among the screened hub genes, Agt, Cdkn1a, Ddit4, and Rhob are related to the neuronal death process. We further confirmed that these genes, especially Ddit4, were upregulated in the hippocampus of aged mice that received sevoflurane anesthesia. NMDAR, the core target receptor of sevoflurane, rather than GABAAR, mediates the sevoflurane regulation of DDIT4 expression. Our study screened sevoflurane-regulated DEGs and focused on the neuronal death process to reveal DDIT4 as a potential target mediated by NMDAR, which may provide a new target for the treatment of sevoflurane neurotoxicity.
Collapse
Affiliation(s)
- Shuai Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qi Hou
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Runjia Wang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu Hou
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qiang Wang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bo Zhang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Cheng Ni
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
20
|
Choo YH, Seo Y, Oh HJ. Deep Sedation in Traumatic Brain Injury Patients. Korean J Neurotrauma 2023; 19:185-194. [PMID: 37431376 PMCID: PMC10329893 DOI: 10.13004/kjnt.2023.19.e19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 07/12/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of mortality and disability in adults. In cases of severe TBI, preventing secondary brain injury by managing intracranial hypertension during the acute phase is a critical treatment challenge. Among surgical and medical interventions to control intracranial pressure (ICP), deep sedation can provide comfort to patients and directly control ICP by regulating cerebral metabolism. However, insufficient sedation does not achieve the intended treatment goals, and excessive sedation can lead to fatal sedative-related complications. Therefore, it is important to continuously monitor and titrate sedatives by measuring the appropriate depth of sedation. In this review, we discuss the effectiveness of deep sedation, techniques to monitor the depth of sedation, and the clinical use of recommended sedatives, barbiturates, and propofol in TBI.
Collapse
Affiliation(s)
- Yoon-Hee Choo
- Department of Neurosurgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Youngbeom Seo
- Department of Neurosurgery, Yeungnam University Hospital, Yeungnam Universtiy College of Medicine, Daegu, Korea
| | - Hyuk-Jin Oh
- Department of Neurosurgery, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| |
Collapse
|
21
|
Jia D, Guo S, Wu X, Zhao M, Luo J, Cheng M, Qin Y. Effect of dexmedetomidine on liver transplantation: a meta-analysis. Front Pharmacol 2023; 14:1188011. [PMID: 37292152 PMCID: PMC10245273 DOI: 10.3389/fphar.2023.1188011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/12/2023] [Indexed: 06/10/2023] Open
Abstract
Background: Dexmedetomidine (DEX), an adjuvant anesthetic, may improve the clinical outcomes of liver transplantation (LT). Methods: We summarized the relevant clinical trials of DEX in patients undergoing LT. As of 30 January 2023, we searched The Cochrane Library, MEDLINE, EMBASE, Clinical Trial.gov and the WHO ICTRP. The main outcomes were postoperative liver and renal function. The random effect model or fixed effect model was used to summarize the outcomes across centers based on the differences in heterogeneity. Results: The meta-analysis included nine studies in total. Compared with the control group, the DEX group had a reduced warm ischemia time (MD-4.39; 95% CI-6.74--2.05), improved postoperative liver (peak aspartate transferase: MD-75.77, 95% CI-112.81--38.73; peak alanine transferase: MD-133.51, 95% CI-235.57--31.45) and renal function (peak creatinine: MD-8.35, 95% CI-14.89--1.80), and a reduced risk of moderate-to-extreme liver ischemia-reperfusion injury (OR 0.28, 95% CI 0.14-0.60). Finally, the hospital stay of these patients was decreased (MD-2.28, 95% CI-4.00--0.56). Subgroup analysis of prospective studies showed that DEX may have better efficacy in living donors and adult recipients. Conclusion: DEX can improve short-term clinical outcomes and shorten the hospital stay of patients. However, the long-term efficacy of DEX and its interfering factors deserves further study. Systematic Review: identifier CRD42022351664.
Collapse
Affiliation(s)
- Degong Jia
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shanshan Guo
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xinyi Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Minjie Zhao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiefu Luo
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingxiang Cheng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajun Qin
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Long Y, Li D, Yu S, Zhang YL, Liu SY, Wan JY, Shi A, Deng J, Wen J, Li XQ, Ma Y, Li N, Yang M. Natural essential oils: A promising strategy for treating cardio-cerebrovascular diseases. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115421. [PMID: 35659628 DOI: 10.1016/j.jep.2022.115421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Essential oils (EO) are volatile compounds obtained from different parts of natural plants, and have been used in national, traditional and folk medicine to treat various health problems all over the world. Records indicate that in history, herbal medicines rich in EO have been widely used for the treatment of CVDs in many countries, such as China. AIM OF THE STUDY This review focused on the traditional application and modern pharmacological mechanisms of herbal medicine EO against CVDs in preclinical and clinical trials through multi-targets synergy. Besides, the EO and anti-CVDs drugs were compared, and the broad application of EO was explained from the properties of drugs and aromatic administration routes. MATERIALS AND METHODS Information about EO and CVDs was collected from electronic databases such as Web of Science, ScienceDirect, PubMed, and China National Knowledge Infrastructure (CNKI). The obtained data sets were sequentially arranged for better understanding of EO' potential. RESULTS The study showed that EO had significant application in CVDs at different countries or regions since ancient times. Aiming at the complex pathological mechanisms of CVDs, including intracellular calcium overload, oxidative stress, inflammation, vascular endothelial cell injury and dysfunction and dyslipidemia, we summarized the roles of EO on CVDs in preclinical and clinical through multi-targets intervention. Besides, EO had the dual properties of drug and excipients. And aromatherapy was one of the complementary therapies to improve CVDs. CONCLUSIONS This paper reviewed the EO on traditional treatment, preclinical mechanism and clinical application of CVDs. As important sources of traditional medicines, EO' remarkable efficacy had been confirmed in comprehensive literature reports, which showed that EO had great medicinal potential.
Collapse
Affiliation(s)
- Yu Long
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuang Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Lu Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Song-Yu Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jin-Yan Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ai Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Wen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao-Qiu Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Ming Yang
- Key Laboratory of Modern Preparation of TCM, Jiangxi University of Traditional Chinese Medicine, Nanchang, China.
| |
Collapse
|
23
|
Arvin S, Yonehara K, Glud AN. Therapeutic Neuromodulation toward a Critical State May Serve as a General Treatment Strategy. Biomedicines 2022; 10:biomedicines10092317. [PMID: 36140418 PMCID: PMC9496064 DOI: 10.3390/biomedicines10092317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/11/2022] [Accepted: 09/14/2022] [Indexed: 11/18/2022] Open
Abstract
Brain disease has become one of this century’s biggest health challenges, urging the development of novel, more effective treatments. To this end, neuromodulation represents an excellent method to modulate the activity of distinct neuronal regions to alleviate disease. Recently, the medical indications for neuromodulation therapy have expanded through the adoption of the idea that neurological disorders emerge from deficits in systems-level structures, such as brain waves and neural topology. Connections between neuronal regions are thought to fluidly form and dissolve again based on the patterns by which neuronal populations synchronize. Akin to a fire that may spread or die out, the brain’s activity may similarly hyper-synchronize and ignite, such as seizures, or dwindle out and go stale, as in a state of coma. Remarkably, however, the healthy brain remains hedged in between these extremes in a critical state around which neuronal activity maneuvers local and global operational modes. While it has been suggested that perturbations of this criticality could underlie neuropathologies, such as vegetative states, epilepsy, and schizophrenia, a major translational impact is yet to be made. In this hypothesis article, we dissect recent computational findings demonstrating that a neural network’s short- and long-range connections have distinct and tractable roles in sustaining the critical regime. While short-range connections shape the dynamics of neuronal activity, long-range connections determine the scope of the neuronal processes. Thus, to facilitate translational progress, we introduce topological and dynamical system concepts within the framework of criticality and discuss the implications and possibilities for therapeutic neuromodulation guided by topological decompositions.
Collapse
Affiliation(s)
- Simon Arvin
- Center for Experimental Neuroscience—CENSE, Department of Neurosurgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark
- Danish Research Institute of Translational Neuroscience—DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Ole Worms Allé 8, 8000 Aarhus C, Denmark
- Department of Neurosurgery, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11 Building A, 8200 Aarhus N, Denmark
- Correspondence: ; Tel.: +45 6083-1275
| | - Keisuke Yonehara
- Danish Research Institute of Translational Neuroscience—DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Ole Worms Allé 8, 8000 Aarhus C, Denmark
- Multiscale Sensory Structure Laboratory, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
- Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Shizuoka 411-8540, Japan
| | - Andreas Nørgaard Glud
- Center for Experimental Neuroscience—CENSE, Department of Neurosurgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark
- Department of Neurosurgery, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11 Building A, 8200 Aarhus N, Denmark
| |
Collapse
|
24
|
Lee CW, Chang YP, Huang YT, Hsing CH, Pang YL, Chuang MH, Wu SZ, Sun CK, Hung KC. General anesthesia but not conscious sedation improves functional outcome in patients receiving endovascular thrombectomy for acute ischemic stroke: A meta-analysis of randomized clinical trials and trial sequence analysis. Front Neurol 2022; 13:1017098. [PMID: 36188372 PMCID: PMC9515609 DOI: 10.3389/fneur.2022.1017098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 08/26/2022] [Indexed: 11/22/2022] Open
Abstract
Background This study aimed at comparing the difference in prognostic outcomes between patients receiving general anesthesia (GA) and conscious sedation (CS) for endovascular thrombectomy after acute ischemic stroke. Methods Databases from Medline, Embase, Google scholar, and Cochrane library were searched for randomized controlled studies (RCTs) comparing patients undergoing GA and CS for endovascular thrombectomy following anterior circulation ischemic stroke. The primary outcome was frequency of 90-day good functional outcome [defined as modified Rankin Scale score of ≤ 2], while secondary outcomes included successful recanalization rate (SRR) [i.e., modified thrombolysis in cerebral infarction = 2b or 3], mortality risk, symptomatic intracranial hemorrhage (ICH), procedure-related complications, hypotension, pneumonia, neurological outcome at post-procedure 24–48 h, and puncture-to-recanalization time. Results Six RCTs including 883 patients published between 2016 and 2022 were included. Merged results revealed a higher SRR [risk ratio (RR) = 1.11, 95% CI: 1.03–1.2, p = 0.007; I2 = 29%] and favorable neurological outcomes at 3-months (RR = 1.2, 95% CI: 1.01–1.41, p = 0.04; I2 = 8%) in the GA group compared to CS group, without difference in the risk of mortality (RR = 0.88), symptomatic ICH (RR = 0.91), procedure-related complications (RR = 1.05), and pneumonia (RR = 1.9) as well as post-procedure neurological outcome (MD = −0.21) and successful recanalization time (MD = 3.33 min). However, GA was associated with a higher risk of hypotension compared with that of CS. Conclusion Patients with acute anterior circulation ischemic stroke receiving GA were associated with a higher successful recanalization rate as well as a better 3-month neurological outcome compared to the use of CS. Further investigations are warranted to verify our findings. Systematic review registration www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022342483, identifier: CRD42022342483.
Collapse
Affiliation(s)
- Chia-Wei Lee
- Department of Neurology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Yang-Pei Chang
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Yen-Ta Huang
- Department of Surgery, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan City, Taiwan
| | - Chung-Hsi Hsing
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
- Department of Medical Research, Chi Mei Medical Center, Tainan City, Taiwan
| | - Yu-Li Pang
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Min-Hsiang Chuang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan City, Taiwan
| | - Su-Zhen Wu
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Cheuk-Kwan Sun
- Department of Emergency Medicine, E-Da Hospital, Kaohsiung City, Taiwan
- College of Medicine, I-Shou University, Kaohsiung City, Taiwan
| | - Kuo-Chuan Hung
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
- *Correspondence: Kuo-Chuan Hung
| |
Collapse
|
25
|
Ceric A, Holgersson J, May T, Skrifvars MB, Hästbacka J, Saxena M, Aneman A, Delaney A, Reade MC, Delcourt C, Jakobsen J, Nielsen N. Level of sedation in critically ill adult patients: a protocol for a systematic review with meta-analysis and trial sequential analysis. BMJ Open 2022; 12:e061806. [PMID: 36691212 PMCID: PMC9462111 DOI: 10.1136/bmjopen-2022-061806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 08/10/2022] [Indexed: 01/28/2023] Open
Abstract
INTRODUCTION It is standard of care to provide sedation to critically ill patients to reduce anxiety, discomfort and promote tolerance of mechanical ventilation. Given that sedatives can have differing effects based on a variety of patient and pharmacological characteristics, treatment approaches are largely based on targeting the level of sedation. The benefits of differing levels of sedation must be balanced against potential adverse effects including haemodynamic instability, causing delirium, delaying awakening and prolonging the time of mechanical ventilation and intensive care stay. This systematic review with meta-analysis aims to investigate the current evidence and compare the effects of differing sedation levels in adult critically ill patients. METHODS AND ANALYSES We will conduct a systematic review based on searches of preidentified major medical databases (eg, MEDLINE, EMBASE, CENTRAL) and clinical trial registries from their inception onwards to identify trials meeting inclusion criteria. We will include randomised clinical trials comparing any degree of sedation with no sedation and lighter sedation with deeper sedation for critically ill patients admitted to the intensive care unit. We will include aggregate data meta-analyses and trial sequential analyses. Risk of bias will be assessed with domains based on the Cochrane risk of bias tool. An eight-step procedure will be used to assess if the thresholds for clinical significance are crossed, and the certainty of the evidence will be assessed using Grades of Recommendations, Assessment, Development and Evaluation. ETHICS AND DISSEMINATION No formal approval or review of ethics is required as individual patient data will not be included. This systematic review has the potential to highlight (1) whether one should believe sedation to be beneficial, harmful or neither in critically ill adults; (2) the existing knowledge gaps and (3) whether the recommendations from guidelines and daily clinical practice are supported by current evidence. These results will be disseminated through publication in a peer-reviewed journal.
Collapse
Affiliation(s)
- Ameldina Ceric
- Department of Clinical Sciences Lund, Anesthesia and Intensive Care, Lund University, Helsingborg Hospital, Helsingborg, Sweden
| | - Johan Holgersson
- Department of Clinical Sciences Lund, Anesthesia and Intensive Care, Lund University, Helsingborg Hospital, Helsingborg, Sweden
| | - Teresa May
- Department of Critical Care, Maine Medical Center, Portland, Maine, USA
| | - Markus B Skrifvars
- Department of Emergency Care and Services, Helsinki University, Helsinki University Hospital, Helsinki, Finland
| | - Johanna Hästbacka
- Department of Anesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Manoj Saxena
- Senior Lecturer, Critical Care Division, The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Anders Aneman
- Intensive Care Unit, South Western Sydney Local Health District, Liverpool Hospital, South Western Sydney Local Health District, South Western Sydney Clinical School, University of New South Wales, and Faculty of Medicine, Health and Human Sciences, Macquarie University, Liverpool, New South Wales, Australia
| | - Anthony Delaney
- The George Institute for Global Health and the University of New South Wales, Sydney, New South Wales, Australia
- Malcolm Fisher Department of Intensive Care, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Michael C Reade
- Department of Regional Health Research, The Faculty of Health Sciences, University of Southern Denmark, Denmark, Denmark
| | - Candice Delcourt
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Janus Jakobsen
- Department of Regional Health Research, The Faculty of Health Sciences, University of Southern Denmark, Denmark, Denmark
| | - Niklas Nielsen
- Department of Clinical Sciences Lund, Anesthesia and Intensive Care, Lund University, Helsingborg Hospital, Helsingborg, Sweden
| |
Collapse
|
26
|
The Potential Role of m6A in the Regulation of TBI-Induced BGA Dysfunction. Antioxidants (Basel) 2022; 11:antiox11081521. [PMID: 36009239 PMCID: PMC9405408 DOI: 10.3390/antiox11081521] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 02/01/2023] Open
Abstract
The brain–gut axis (BGA) is an important bidirectional communication pathway for the development, progress and interaction of many diseases between the brain and gut, but the mechanisms remain unclear, especially the post-transcriptional regulation of BGA after traumatic brain injury (TBI). RNA methylation is one of the most important modifications in post-transcriptional regulation. N6-methyladenosine (m6A), as the most abundant post-transcriptional modification of mRNA in eukaryotes, has recently been identified and characterized in both the brain and gut. The purpose of this review is to describe the pathophysiological changes in BGA after TBI, and then investigate the post-transcriptional bidirectional regulation mechanisms of TBI-induced BGA dysfunction. Here, we mainly focus on the characteristics of m6A RNA methylation in the post-TBI BGA, highlight the possible regulatory mechanisms of m6A modification in TBI-induced BGA dysfunction, and finally discuss the outcome of considering m6A as a therapeutic target to improve the recovery of the brain and gut dysfunction caused by TBI.
Collapse
|
27
|
What Are We Measuring? A Refined Look at the Process of Disrupted Autoregulation and the Limitations of Cerebral Perfusion Pressure in Preventing Secondary Injury after Traumatic Brain Injury. Clin Neurol Neurosurg 2022; 221:107389. [PMID: 35961231 DOI: 10.1016/j.clineuro.2022.107389] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 11/23/2022]
|
28
|
MicroRNA-17-5p Protects against Propofol Anesthesia-Induced Neurotoxicity and Autophagy Impairment via Targeting BCL2L11. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:6018037. [PMID: 35799645 PMCID: PMC9256336 DOI: 10.1155/2022/6018037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022]
Abstract
Background. Propofol (PPF) has been shown in studies to cause cognitive impairment and neuronal cell death in developing animals. PPF has been demonstrated to decrease the expression of microRNA-17-5p (miR-17-5p) in a recent study. Nonetheless, the function of miR-17-5p in PPF-induced neurotoxicity and related mechanisms is uncharacterized. Methods. After the induction of neurotoxicity by treating the SH-SY5Y cells with PPF, qRT-PCR was conducted to evaluate the level of miR-17-5p. Using MTT and flow cytometry, cell viability and apoptosis rate were assessed, respectively. Interaction between miR-17-5p and BCL2 like 11 was (BCL2L11) studied using a Luciferase reporter assay. With the help of western blot analysis, we determined the level of proteins of apoptosis-related genes and autophagy-related markers. Results. In SH-SY5Y cells, PPF treatment induced neurotoxicity and downregulated miR-17-5p expression. In SH-SY5Y cells post-PPF exposure, overexpression of miR-17-5p increased cell viability and decreased apoptosis. Consistently, miR-17-5p mimics mitigated PPF-generated autophagy via inhibition of Atg5, Beclin1, and LC3II/I level and elevation of p62 protein expression. In addition, BCL2L11, which was highly expressed in PPF-treated SH-SY5Y cells, was directly targeted by miR-17-5p. Further, in PPF-treated SH-SY5Y cells, overexpressed BCL2L11 counteracted the suppressing behavior of miR-17-5p elevation on PPF-induced apoptosis. Conclusion. Overexpressed miR-17-5p alleviates PPF exposure-induced neurotoxicity and autophagy in SH-SY5Y cells via binding to BCL2L11, suggesting the possibility that miR-17-5p can serve as a candidate in the treatment of neurotoxicity (caused by PPF).
Collapse
|
29
|
Propofol Prevents the Growth, Migration, Invasion, and Glycolysis of Colorectal Cancer Cells by Downregulating Lactate Dehydrogenase Both In Vitro and In Vivo. JOURNAL OF ONCOLOGY 2022; 2022:8317466. [PMID: 35535311 PMCID: PMC9078837 DOI: 10.1155/2022/8317466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022]
Abstract
Colorectal cancer (CRC) is one of the most frequently diagnosed gastrointestinal malignancies worldwide and has high rates of morbidity and mortality. Propofol has been reported to have certain anticancer properties. However, the role and mechanism of propofol in CRC are not entirely clear. CRC cells were treated with propofol and/or LDH-overexpression plasmids, and a mouse xenograft model of CRC was also established and treated with propofol. Cell viability, migration, and invasion were evaluated by CCK-8, wound healing, and transwell assays; the expression of related proteins was confirmed by western blotting; indexes of the glycolytic pathway were analyzed using specialized kits; tumor growth in mice was measured; pathological tissue structure was assessed by H&E staining; and 8-OHDG expression was determined by an immunochemistry assay. Our results verified that propofol could effectively prevent the malignant behaviors of CRC cells by suppressing cell viability, migration, and invasion and accelerating apoptosis. We also discovered that propofol could attenuate the glycolytic pathway in CRC cells. Moreover, we proved that lactate dehydrogenase (LDH) was required for the inhibitory effects of propofol on the growth of CRC cells, including glycolysis in CRC cells. Furthermore, our results showed that propofol could not only significantly inhibit tumor growth and glycolysis, but also ameliorate the pathological structure of CRC tumors. The current results proved that propofol could attenuate the malignant progression of CRC by preventing LDH activity, suggesting that propofol might be an effective therapeutic agent for CRC.
Collapse
|
30
|
Li H, Li J, Su P, Zhang J, Ma D. A Clinical Study on the Brain Protection Effect of Propofol Anesthesia on Patients Undergoing Acute Craniocerebral Trauma Surgery Based on Blockchain. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:6111543. [PMID: 35437464 PMCID: PMC9013313 DOI: 10.1155/2022/6111543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/24/2022] [Accepted: 03/16/2022] [Indexed: 11/17/2022]
Abstract
In order to explore the effects of propofol anesthesia system based on block chain on brain tissue and brain function protection, 88 patients undergoing craniocerebral trauma surgery are selected for prospective study. The anesthetic dose of propofol during operation in the control group is determined according to general information and personal situation. The team uses blockchain to process the patient's clinical data and general information to calculate the optimal dose of propofol. The changes of hemodynamic indexes of patients in the two groups are compared, including dural incision (T1), 1 h after craniotomy (T2), end of surgery (T3), and 6 h after surgery (T4). The changes of neuron-specific enolase (NSE) and central neuron-specific protein (S100β) at T1, T2, T3, and T4 in 2 groups are observed and recorded. The changes of malondialdehyde (MDA) at T1, T2, T3, and T4 are compared between the two groups. Based on the analysis of clinical data and general information of patients on blockchain, the optimal intraoperative anesthetic dose of propofol for patients can effectively protect the brain function of patients, and the intraoperative anesthesia state is relatively stable, and the changes and improvements of intraoperative indicators are good.
Collapse
Affiliation(s)
- Hu Li
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610032, China
| | - Jinfeng Li
- Department of Anesthesiology, Chengdu Seventh People's Hospital, Chengdu 610041, China
| | - Peng Su
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610032, China
| | - JianJun Zhang
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610032, China
| | - Ding Ma
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610032, China
| |
Collapse
|
31
|
Wang Z, Du X, Yu D, Yang Y, Ma G, Jia X, Cheng L. Sufentanil alleviates cerebral ischemia-reperfusion injury by inhibiting inflammation and protecting the blood-brain barrier in rats. Eur J Histochem 2022; 66:3328. [PMID: 35016494 PMCID: PMC8764464 DOI: 10.4081/ejh.2022.3328] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/19/2021] [Indexed: 11/23/2022] Open
Abstract
Stroke is a brain system disease with a high fatality rate and disability rate. About 80% of strokes are ischemic strokes. Cerebral ischemia-reperfusion injury (CIRI) caused by ischemic stroke seriously affects the prognosis of stroke patients. The purpose of this study is to investigate the effect of sufentanil (SUF) on CIRI model rats. We used middle cerebral artery occlusion (MCAO) to make the CIRI model in rats and monitored region cerebral blood flow (rCBF) to ensure that blood flow was blocked and recanalized. We used ELISA and RT-PCR to detect the expression of inflammatory factors in rat serum and brain tissue. In addition, we detected the expression of metalloproteinase (MMP) 2, MMP9 and collagen IV in brain tissues and performed Evans blue (EB) assay to determine the permeability of the blood-brain barrier (BBB). Finally, we clarified the apoptosis of brain tissue through the TUNEL staining and the detection of caspase3, Bcl2 and Bax. Various concentrations of SUF, especially 5, 10 and 25 μg/kg of SUF, all alleviated the infarct size, neurological function and brain edema of MCAO rats. SUF pretreatment also effectively reduced the expression of inflammatory cytokines in MCAO rats, including interleukin (IL)-1β, IL-4, IL-6, IL-8, IL-10 and tumor necrosis factor (TNF)-α. In addition, SUF also inhibited MMP2 and MMP9 and promoted the expression of collagen IV, indicating that SUF attenuated the destruction of the BBB. SUF also inhibited caspase3 and Bax rats and promoted Bcl2 in MCAO rats, thus inhibiting cell apoptosis. SUF pretreatment effectively improved the neurological function and cerebral infarction of MCAO rats, inhibited excessive inflammation in rats, protected the BBB, and inhibited cell apoptosis in brain tissue.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Anesthesiology, Zhumadian Central Hospital, Zhumadian.
| | - Xiaoyan Du
- Rehabilitation Ward, Zhumadian City Welfare Home for Children, Zhumadian.
| | - Daoyang Yu
- Department of Anesthesiology, Zhumadian Central Hospital, Zhumadian.
| | - Yang Yang
- Department of Neurosurgery, Zhumadian Central Hospital, Zhumadian.
| | - Gaoen Ma
- Department of Ophtalmology, Third Affiliated Hospital of Xinxiang Medical College, Xinxiang.
| | - Xueli Jia
- Department of Anesthesiology, Zhumadian Central Hospital, Zhumadian.
| | - Lulu Cheng
- Laboratory of Microneurosurgery, Zhumadian Central Hospital, Zhumadian.
| |
Collapse
|
32
|
Kayambankadzanja RK, Samwel R, Baker T. Pragmatic sedation strategies to prevent secondary brain injury in low‐resource settings. Anaesthesia 2022; 77 Suppl 1:43-48. [DOI: 10.1111/anae.15621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 01/22/2023]
Affiliation(s)
- R. K. Kayambankadzanja
- School of Public Health Kamuzu University of Health Science Blantyre Malawi
- Department of Anaesthesia and Intensive Care Queen Elizabeth Central Hospital Blantyre Malawi
| | - R. Samwel
- Department of Anaesthesia and Intensive Care Bugando Medical Centre Mwanza Tanzania
| | - T. Baker
- Muhimbili University of Health and Allied Sciences Dar es Salaam Tanzania
- Department of Clinical Research London School of Hygiene and Tropical Medicine London UK
| |
Collapse
|
33
|
Yuan M, Wu H. Astrocytes in the Traumatic Brain Injury: the Good and the Bad. Exp Neurol 2021; 348:113943. [PMID: 34863998 DOI: 10.1016/j.expneurol.2021.113943] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/08/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022]
Abstract
Astrocytes control many processes of the nervous system in health and disease, and respond to injury quickly. Astrocytes produce neuroprotective factors in the injured brain to clear cellular debris and to orchestrate neurorestorative processes that are beneficial for neurological recovery after traumatic brain injury (TBI). However, astrocytes also become dysregulated and produce cytotoxic mediators that hinder CNS repair by induction of neuronal dysfunction and cell death. Hence, we discuss the potential role of astrocytes in neuropathological processes such as neuroinflammation, neurogenesis, synaptogenesis and blood-brain barrier repair after TBI. Thus, an improved understanding of the dual role of astrocytes may advance our knowledge of post-brain injury recovery, and provide opportunities for the development of novel therapeutic strategies for TBI.
Collapse
Affiliation(s)
- Mengqi Yuan
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Haitao Wu
- Beijing Institute of Basic Medical Sciences, 100850 Beijing, China; Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, Jiangsu, China; Chinese Institute for Brain Research (CIBR), 102206 Beijing, China.
| |
Collapse
|
34
|
Firdaus R, Theresia S, Austin R, Tiara R. Propofol effects in rodent models of traumatic brain injury: a systematic review. ASIAN BIOMED 2021; 15:253-265. [PMID: 37551361 PMCID: PMC10321222 DOI: 10.2478/abm-2021-0032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background Traumatic brain injury (TBI) causes high mortality and disability worldwide. Animal models have been developed to explore the complex processes in TBI. Propofol is used to manage head injuries during surgical intervention and mechanical ventilation in patients with TBI. Many studies have investigated the neuroprotective effect of propofol on TBI. However, other studies have shown neurotoxic effects. Objectives To review systematically the literature regarding the neuroprotective and neurotoxic effects of propofol in rodent models of TBI. Methods Data from rodents as models of TBI with propofol as one of the intervention agents, and/or comparing the neuroprotective effects of propofol with the other substances in rodent models of TBI, were obtained from PubMed, EBSCO Host, and ProQuest databases. The PRISMA 2020 statement recommendations were followed and research questions were developed based on PICOS guidelines. Data was extracted from the literature using a standardized Cochrane method. Results We analyzed data from 12 articles on physiological changes of experimental animals before and after trauma, the effects of propofol administration, and the observed neurotoxic effects. The effects of propofol administration were observed in terms of changes in traumatic lesion volume, the release of antioxidants and inflammatory factors, and the neurological function of rodent models of TBI. Conclusion Propofol has neuroprotective and neurotoxic effects via several mechanisms, and various doses have been used in research to determine its effects. The timing of administration, the dose administered, and the duration of administration contribute to determine the effect of propofol in rodent models of TBI. However, the doses that produce neuroprotective and neurotoxic effects are not yet clear and further research is needed to determine them.
Collapse
Affiliation(s)
- Riyadh Firdaus
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta10430, Indonesia
| | - Sandy Theresia
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta10430, Indonesia
| | - Ryan Austin
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta10430, Indonesia
| | - Rani Tiara
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta10430, Indonesia
| |
Collapse
|
35
|
Sousa GC, Fernandes MV, Cruz FF, Antunes MA, da Silva CM, Takyia C, Battaglini D, Samary CS, Robba C, Pelosi P, Rocco PRM, Silva PL. Comparative effects of dexmedetomidine and propofol on brain and lung damage in experimental acute ischemic stroke. Sci Rep 2021; 11:23133. [PMID: 34848804 PMCID: PMC8633001 DOI: 10.1038/s41598-021-02608-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 11/17/2021] [Indexed: 11/09/2022] Open
Abstract
Acute ischemic stroke is associated with pulmonary complications, and often dexmedetomidine and propofol are used to decrease cerebral metabolic rate. However, it is unknown the immunomodulatory actions of dexmedetomidine and propofol on brain and lungs during acute ischemic stroke. The effects of dexmedetomidine and propofol were compared on perilesional brain tissue and lung damage after acute ischemic stroke in rats. Further, the mean amount of both sedatives was directly evaluated on alveolar macrophages and lung endothelial cells primarily extracted 24-h after acute ischemic stroke. In twenty-five Wistar rats, ischemic stroke was induced and after 24-h treated with sodium thiopental (STROKE), dexmedetomidine and propofol. Dexmedetomidine, compared to STROKE, reduced diffuse alveolar damage score [median(interquartile range); 12(7.8–15.3) vs. 19.5(18–24), p = 0.007)], bronchoconstriction index [2.28(2.08–2.36) vs. 2.64(2.53–2.77), p = 0.006], and TNF-α expression (p = 0.0003), while propofol increased VCAM-1 expression compared to STROKE (p = 0.0004). In perilesional brain tissue, dexmedetomidine, compared to STROKE, decreased TNF-α (p = 0.010), while propofol increased VCAM-1 compared to STROKE (p = 0.024). In alveolar macrophages and endothelial cells, dexmedetomidine decreased IL-6 and IL-1β compared to STROKE (p = 0.002, and p = 0.040, respectively), and reduced IL-1β compared to propofol (p = 0.014). Dexmedetomidine, but not propofol, induced brain and lung protection in experimental acute ischemic stroke.
Collapse
Affiliation(s)
- Giselle C Sousa
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Department of Anesthesiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos Vinicius Fernandes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Rio de Janeiro Network on Neuroinflammation, Carlos Chagas Filho Foundation for Supporting Research in the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Rio de Janeiro Network on Neuroinflammation, Carlos Chagas Filho Foundation for Supporting Research in the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| | - Mariana A Antunes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Carla M da Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Laboratory of Imunopathology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Christina Takyia
- Laboratory of Imunopathology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Denise Battaglini
- San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, University of Genoa, Genoa, Italy
| | - Cynthia S Samary
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Rio de Janeiro Network on Neuroinflammation, Carlos Chagas Filho Foundation for Supporting Research in the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| | - Chiara Robba
- San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, University of Genoa, Genoa, Italy.,Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Paolo Pelosi
- San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, University of Genoa, Genoa, Italy.,Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Rio de Janeiro Network on Neuroinflammation, Carlos Chagas Filho Foundation for Supporting Research in the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, 373, Bloco G-014, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil. .,Rio de Janeiro Network on Neuroinflammation, Carlos Chagas Filho Foundation for Supporting Research in the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil.
| |
Collapse
|
36
|
Liu J, Ai P, Sun Y, Yang X, Li C, Liu Y, Xia X, Zheng JC. Propofol Inhibits Microglial Activation via miR-106b/Pi3k/Akt Axis. Front Cell Neurosci 2021; 15:768364. [PMID: 34776870 PMCID: PMC8581742 DOI: 10.3389/fncel.2021.768364] [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: 08/31/2021] [Accepted: 10/06/2021] [Indexed: 12/05/2022] Open
Abstract
Propofol is an established intravenous anesthetic agent with potential neuroprotective effects. In this study, we investigated the roles and the underlying mechanisms of propofol in inhibiting the pro-inflammatory responses of microglia. Propofol significantly reduced the messenger RNA (mRNA) levels of Tnf, Nos2, and NF-κB pathway related genes Ticam1, Myd88, Irf3, and Nfkb1 in lipopolysaccharide (LPS)-treated primary microglia. After screening the miRNA profiles in microglia under LPS and propofol treatment conditions, we found propofol abrogated the LPS-induced misexpression of miRNAs including miR-106b, miR-124, miR-185, and miR-9. Perturbation of function approaches suggested miR-106b as the core miRNA that mediated the anti-inflammatory effects of propofol on microglial activation. RNA sequencing (RNA-seq) analysis further identified Pi3k/Akt signaling as one of the most affected pathways after miR-106b perturbation of function. The treatment of Pi3k/Akt signaling agonist 740Y-P elevated miR-106b-reduced Akt phosphorylation and abolished the inhibitory effects of miR-106b on the pro-inflammatory responses of microglia. Our results suggest propofol inhibits microglial activation via miR-106b/Pi3k/Akt axis, shedding light on a novel molecular mechanism of propofol-mediated immunomodulatory effects and implying propofol as potential therapeutics for treating neuroinflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Jianhui Liu
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Pu Ai
- Wuxi Clinical College of Anhui Medical University, Hefei, China
| | - Yiyan Sun
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xiaoyu Yang
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Chunhong Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yihan Liu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaohuan Xia
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Jialin C Zheng
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
| |
Collapse
|
37
|
Cheng W, Sun X, Liu Y, Han S, Ren W. The Chronotropic Function of Propofol and the Underlying Mechanism in Rabbits. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:5222745. [PMID: 34820077 PMCID: PMC8608509 DOI: 10.1155/2021/5222745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 11/24/2022]
Abstract
The report of bradycardia caused by propofol is increasing. In the experiment, we investigated the chronotropic function of propofol and the underlying mechanism. Rabbits of both sexes were randomly divided into 4 groups: propofol 5 mg/kg group, 10 mg/kg group, 15 mg/kg group, and sham group. Heart rate and frequency of vagal efferent discharge were recorded before the injection and 0, 0.5, 1, 2, and 10 min after the injection through intravenous mode. Then, their hearts were removed, and sinoatrial nodes were dissected. The action potentials of the sinus node pacemaker cells were recorded by the intracellular glass microelectrode technique, and the sinoatrial (SA) node was exposed to propofol 1, 3, 5, and 10 µM respectively. The action potentials were recorded after the sinoatrial nodes were exposed to each concentration of propofol for 15 min. Our results show that the heart rate significantly decreased, and the vagal efferent discharge was significantly increased at 0, 0.5, 1, and 2 min after the injection, respectively. Besides, as the dose increases, the magnitude of change shows a dose-dependent manner. Propofol exerts a negative chronotropic action on sinoatrial node pacemaker cells. The drug significantly decreased APA, VDD, RPF, and prolonged APD90 in a concentration-dependent manner. These effects may be the main mechanism of propofol-induced bradycardia in clinical study.
Collapse
Affiliation(s)
- Wenjie Cheng
- Department of Anesthesiology, Tianjin Hospital, Tianjin 300211, China
| | - Xiaohua Sun
- Department of Anesthesiology, Tianjin Hospital, Tianjin 300211, China
| | - Yanfang Liu
- Department of Anesthesiology, Tianjin Hospital, Tianjin 300211, China
| | - Shiqi Han
- Department of Anesthesiology, Tianjin Hospital, Tianjin 300211, China
| | - Wanlu Ren
- Department of Anesthesiology, Tianjin Hospital, Tianjin 300211, China
| |
Collapse
|
38
|
Bourg P, Salottolo K, Klein J, Bar-Or D. Can a biomarker for oxidative stress and antioxidant reserves identify frailty in geriatric trauma patients? Injury 2021; 52:2908-2913. [PMID: 33573809 DOI: 10.1016/j.injury.2021.01.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Frailty is a state of systematic physiologic decline and reduced ability to recover from illness. There are no rapid quantitative biological measures to assess frailty. The study objective was to determine whether oxidation-reduction potential (ORP) is correlated with frailty score. METHODS This prospective, observational cohort study was performed using plasma samples of geriatric trauma patients (≥65 years) admitted to a level I trauma center. Frailty was measured with the Canadian Study of Health and Aging (CSHA) Clinical Frailty Scale (7-point scale; 1 = robust health and 7 = severely frail). Plasma ORP was determined using the RedoxSYS™ system to measure static ORP (aggregate measure of oxidative stress) and capacity ORP (antioxidant reserves; log transformed). Spearman rank correlation (presented as rs) and ordinal logistic regression (presented as adjusted odds ratios, AOR) were used to examine the unadjusted and adjusted relationship between frailty score and ORP values. RESULTS There were 93 geriatric trauma patients in our study. The majority (86%) had frailty scores 1-5, 11% were moderately frail and 3% were severely frail. There was a u-shaped relationship between ORP and frailty scale that became monotonic for scores 1-5. Each increase in frailty score demonstrated significant decreases in antioxidant reserves (log cORP rs = -0.26, p = 0.02) and nonsignificant increases in oxidative stress (sORP rs = 0.17, p = 0.15). After adjustment, variables significantly associated with frailty included log cORP (e.g., fewer antioxidant reserves, AOR: 0.70), age (AOR: 1.82), injury severity score (AOR: 0.50), admission lactate ≥2.5 mMol (AOR: 4.31), and alcohol use (AOR: 0.34). CONCLUSIONS The amount of antioxidant reserves (cORP) appears to be a quantitative marker to differentiate the degree of frailty ranging from robust health to mild frailty in geriatric trauma patients. We propose that direct quantification of frailty by way of a biomarker for oxidative reserves could have application in emergent trauma situations.
Collapse
Affiliation(s)
| | | | | | - David Bar-Or
- Trauma Research Department, St Anthony Hospital, Lakewood, CO; Rocky Vista University, Parker, CO.
| |
Collapse
|
39
|
Jing H, Wang C, Zhao L, Cheng J, Qin P, Lin H. Propofol protects cardiomyocytes from hypoxia/reoxygenation injury via regulating MALAT1/miR-206/ATG3 axis. J Biochem Mol Toxicol 2021; 35:e22880. [PMID: 34383354 DOI: 10.1002/jbt.22880] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 06/06/2021] [Accepted: 07/29/2021] [Indexed: 01/24/2023]
Abstract
Previous studies have shown that propofol (PPF) plays a protective role in ischemia-reperfusion (I/R) in multiple organs and tissues. This study was aimed to explore the mechanism of PPF in ameliorating myocardial ischemia-reperfusion injury (MIRI). MIRI model was established with Sprague-Dawley rats, and PPF pretreatment was performed before reperfusion. Creatine kinase isoform (CK-MB), lactate dehydrogenase (LDH), and hematoxylin and eosin stain were used to evaluate the severity of MIRI. H9c2 cells were treated with hypoxia/reoxygenation (H/R) to simulate I/R injury in vitro. Real-time quantitative polymerase chain reaction (qPCR) was employed to assess MALAT1 and microRNA (miR)-206 expressions. Autophagy-related 3 (ATG3), LC3BⅡ/LC3BⅠ, and Beclin-1 expression were examined by western blot. Apoptosis was monitored using flow cytometry. Interaction between MALAT1 and miR-206 was determined by bioinformatics analysis, dual-luciferase reporter gene assay, RIP assay, and RNA pull-down assay. PPF pretreatment remarkably reduced CK-MB level, LDH level, myocardial infarct size, and LC3BⅡ/LC3BⅠ ratio and Beclin-1 expression in the rats with MIRI, and repressed the apoptosis of H9c2 cells exposed to H/R. PPF pretreatment markedly suppressed MALAT1 expression and enhanced miR-206 expression in both in vivo and in vitro models. MiR-206 was identified as a target of MALAT1 in cardiomyocytes, and MALAT1 could increase the expression of ATG3. Additionally, the upregulation of MALAT1 partially reversed the protective effect of PPF on cardiomyocytes in vitro. PPF modulated MALAT1/miR-206/ATG3 axis to protect cardiomyocytes against I/R injury.
Collapse
Affiliation(s)
- Haijuan Jing
- Department of Anesthesiology, Henan Provincial People's Hospital (Central China Fuwai Hospital of Zhengzhou University), Zhengzhou, Henan, China.,Department of Anesthesiology, Central China Fuwai Hospital (Central China Fuwai Hospital of Zhengzhou University), Zhengzhou, Henan, China
| | - Chuan Wang
- Department of Anesthesiology, Hebei North University, Zhangjiakou, Hebei, China
| | - Liang Zhao
- Department of Anesthesiology, Henan Provincial People's Hospital (Central China Fuwai Hospital of Zhengzhou University), Zhengzhou, Henan, China
| | - Jing Cheng
- Department of Anesthesiology, Henan Provincial People's Hospital (Central China Fuwai Hospital of Zhengzhou University), Zhengzhou, Henan, China
| | - Pengyu Qin
- Department of Anesthesiology, Henan Provincial People's Hospital (Central China Fuwai Hospital of Zhengzhou University), Zhengzhou, Henan, China
| | - Hongqi Lin
- Department of Anesthesiology, Henan Provincial People's Hospital (Central China Fuwai Hospital of Zhengzhou University), Zhengzhou, Henan, China.,Department of Anesthesiology, Central China Fuwai Hospital (Central China Fuwai Hospital of Zhengzhou University), Zhengzhou, Henan, China
| |
Collapse
|
40
|
Girnar GA, Mahajan HS. Cerebral ischemic stroke and different approaches for treatment of stroke. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00289-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Cerebral ischemia can be considered a lethal disease as it is a leading cause of death worldwide with no prompt line of treatment. The factors which make this disease more fatal are failure of drugs while crossing BBB, very low availability of the drug in the brain, inefficiency of drug molecule in the clinical studies, limited availability of clinical data, lack of awareness about this disease, and many more.
Main body
This review focuses on reasons and mechanisms of stroke, classification of brain ischemia; it also reveals the current scenario of stroke in India. Very few drugs are effective for the treatment of stroke. This compilation furnishes conventional and recent treatments of stroke along with their hurdles like the gap between preclinical and clinical studies. This review also suggests effective routes of administration of drugs for the treatment of brain ischemia specifically nose-to-brain route and effectiveness of different dosage forms precisely nanoformulations, as the most effective dosage form.
Conclusion
By following different guidelines and treatments, the risk of brain ischemia can be minimized as well as some advanced techniques for the treatment of this disease proving their efficiency. One of the important aspects in the success of the treatment for this disease is the route of administration of the drug. Among all routes, intranasal drug delivery presents a potential approach and is supposed to be the next-generation therapy for brain disorders. The nose-to-brain route is very effective, and it shows some promising results in case of stroke treatment. The strategy is still under investigation despite various successful lab-scale studies; there are numerous challenges to reach the product in the market. Research is going on to get a better understanding of this strategy. We believe that detailed studies to resolve pitfalls will lead to the successful development of an intranasal formulation for the management of ischemic brain injury such as stroke.
Collapse
|
41
|
Jia L, Hao H, Wang C, Wei J. Etomidate attenuates hyperoxia-induced acute lung injury in mice by modulating the Nrf2/HO-1 signaling pathway. Exp Ther Med 2021; 22:785. [PMID: 34055084 PMCID: PMC8145798 DOI: 10.3892/etm.2021.10217] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
The present study aimed to investigate the protective effects of etomidate on hyperoxia-induced acute lung injury in mice, particularly on the nuclear factor-erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. Fifty specific pathogen-free mice were randomly divided into the blank control, model, high oxygen exposure + low etomidate dose (0.3 mg·kg-1), a high oxygen exposure + moderate etomidate dose (3 mg·kg-1), and a high oxygen exposure + high etomidate dose (10 mg·kg-1) groups, with ten mice allotted per group. After 72 h, the mice were sacrificed and the lung tissues were harvested, and the wet-to-dry (W/D) ratio of the tissues was calculated. Hematoxylin-eosin staining was performed to observe the pathological changes in the lung tissues, and the lung injury score (LIS) was calculated. The mRNA and protein expression levels of Nrf2 and HO-1 were measured. The malondialdehyde (MDA), myeloperoxidase (MPO), superoxide dismutase (SOD) and catalase (CAT) levels were also measured, and interleukin (IL)-1β, IL-6, tumor necrosis factor alpha (TNF-α) and IL-10 concentrations in the bronchoalveolar lavage fluid were determined. At low and moderate doses, etomidate decreased pathological damage in the lung tissue, decreased the LIS and W/D ratio, upregulated Nrf2 and HO-1 mRNA and protein expression, decreased IL-1β, IL-6, and TNF-α concentrations, increased MPO activity and IL-10 levels, suppressed the production of the oxidation product MDA, and enhanced the activities of the antioxidant enzymes CAT and SOD. Within a certain dose range, etomidate enhanced antioxidant and anti-inflammatory effects in mice, thereby decreasing lung injury induced by the chronic inhalation of oxygen at high concentrations. Furthermore, the underlying mechanism may be associate with the upregulation of the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Liming Jia
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Hongzhong Hao
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Chunyu Wang
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Jianfeng Wei
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| |
Collapse
|
42
|
Propofol Protects Against Hepatic Ischemia Reperfusion Injury via Inhibiting Bnip3-Mediated Oxidative Stress. Inflammation 2021; 44:1288-1301. [PMID: 33496895 DOI: 10.1007/s10753-021-01416-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/30/2020] [Accepted: 01/05/2021] [Indexed: 12/18/2022]
Abstract
Propofol (PRO) protects against hepatic ischemia/reperfusion (I/R) injury. Bnip3 is involved in the I/R-induced injury. This study investigated whether the effect of PRO on hepatic hypoxia/reoxygenation (H/R) injury was realized through regulating Bnip3. After establishing a hepatic ischemia reperfusion (I/R ) injury model in mice, the serum levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were determined by an automatic biochemical analyzer. The histopathology and apoptosis of liver tissues were detected by hematoxylin-eosin and TUNEL staining. After the H/R liver cells were cultured and treated with PRO, the viability, apoptosis, reactive oxygen species (ROS) production, and the levels of lactate dehydrogenase (LDH), malondialdehyde (MDA), TNF-α, and IL-6 were detected by MTT, flow cytometry, colorimetry, and ELISA. The expressions of Bnip3 and apoptosis-related factors in I/R mouse liver tissues and H/R cells were determined by immunohistochemical assay, immunofluorescence, Western blot, or RT-qPCR. PRO ameliorated the abnormal histopathology, reduced cell apoptosis and the levels of AST, ALT, Bnip3, Cleaved Caspase-3, and Bax, but upregulated the Bcl-2 level in the liver tissues of I/R mice. In H/R liver cells, PRO promoted the cell viability, downregulated the levels of LDH, MDA, TNF-α, IL-6, and reduced ROS production. Moreover, PRO promoted the downregulated expressions of cytosolic Bnip3, total Bni3p, Cleaved Caspase-3, and Bax and upregulated the Bcl-2 level. siBnip3 reversed the effect of H/R on the liver cells, and its overexpression also reversed the effect of PRO on H/R-induced liver cells. PRO protects against hepatic I/R injury via inhibiting Bnip3.
Collapse
|
43
|
Li X, Yu J, Ma D, Weng X. Edaravone Improves the Post-traumatic Brain Injury Dysfunction in Learning and Memory by Modulating Nrf2/ARE Signal Pathway. Clinics (Sao Paulo) 2021; 76:e3131. [PMID: 34878029 PMCID: PMC8610218 DOI: 10.6061/clinics/2021/e3131] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/16/2021] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES To investigate the molecular mechanism of edaravone (EDA) in improving the post-traumatic brain injury (TBI) dysfunction in learning and memory. METHODS In vitro and in vivo TBI models were established using hydrogen peroxide (H2O2) treatment for hippocampal nerve stem cells (NSCs) and surgery for rats, followed by EDA treatment. WST 1 measurement, methylthiazol tetrazolium assay, and flow cytometry were performed to determine the activity, proliferation, and apoptosis of NSCs, and malondialdehyde (MDA), lactic dehydrogenase (LDH), and reactive oxygen species (ROS) detection kits were used to analyze the oxides in NSCs. RESULTS Following EDA pretreatment, NSCs presented with promising resistance to H2O2-induced oxidative stress, whereas NSCs manifested significant increases in activity and proliferation and a decrease in apoptosis. Meanwhile, for NSCs, EDA pretreatment reduced the levels of MDA, LDH, and ROS, with a significant upregulation of Nrf2/antioxidant response element (ARE) signaling pathway, whereas for EDA-treated TBI rats, a significant reduction was observed in the trauma area and injury to the hippocampus, with improvement in memory and learning performance and upregulation of Nrf2/ARE signaling pathway. CONCLUSIONS EDA, by regulating the activity of Nrf2/ARE signal pathway, can improve the TBI-induced injury to NSCs and learning and memory dysfunction in rats.
Collapse
Affiliation(s)
| | - Jing Yu
- Corresponding author. E-mail:
| | | | | |
Collapse
|
44
|
Wang Z, Wu J, Hu Z, Luo C, Wang P, Zhang Y, Li H. Dexmedetomidine Alleviates Lipopolysaccharide-Induced Acute Kidney Injury by Inhibiting p75NTR-Mediated Oxidative Stress and Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5454210. [PMID: 33194004 PMCID: PMC7648709 DOI: 10.1155/2020/5454210] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/22/2020] [Accepted: 10/07/2020] [Indexed: 02/07/2023]
Abstract
Oxidative stress and apoptosis play a key role in the pathogenesis of sepsis-associated acute kidney injury (AKI). Dexmedetomidine (DEX) may present renal protective effects in sepsis. Therefore, we studied antioxidant effects and the mechanism of DEX in an inflammatory proximal tubular epithelial cell model and lipopolysaccharide- (LPS-) induced AKI in mice. Methods. We assessed renal function (creatinine, urea nitrogen), histopathology, oxidative stress (malondialdehyde (MDA) and superoxide dismutase (SOD)), and apoptosis (TUNEL staining and Cleaved caspase-3) in mice. In vitro experiments including Cleaved caspase-3 and p75NTR/p38MAPK/JNK signaling pathways were evaluated using western blot. Reactive oxidative species (ROS) production and apoptosis were determined using flow cytometry. Results. DEX significantly improved renal function and kidney injury and also revert the substantially increased level of MDA concentrations as well as the reduction of the SOD enzyme activity found in LPS-induced AKI mice. In parallel, DEX treatment also reduced the apoptosis and Cleaved caspase-3 expression evoked by LPS. The expression of p75NTR was increased in kidney tissues of mice with AKI but decreased after treatment with DEX. In cultured human renal tubular epithelial cell line (HK-2 cells), DEX inhibited LPS-induced apoptosis and generation of ROS, but this was reversed by overexpression of p75NTR. Furthermore, pretreatment with DEX significantly downregulated phosphorylation of JNK and p38MAPK in LPS-stimulated HK-2 cells, and this effect was abolished by overexpression of p75NTR. Conclusion. DEX ameliorated AKI in mice with sepsis by partially reducing oxidative stress and apoptosis through regulation of p75NTR/p38MAPK/JNK signaling pathways.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiali Wu
- Department of Laboratory Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaolan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Cong Luo
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Pengfei Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yanling Zhang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hui Li
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
45
|
Liao Z, Sun H, Chang Y, Chen H. The expression and clinical significance of miRNA-183 in cerebral ischemia-reperfusion injury patients with cerebral small vessel disease. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1005. [PMID: 32953805 PMCID: PMC7475471 DOI: 10.21037/atm-20-5335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background To investigate the expression and clinical significance of micro (mi)RNA-183 in cerebral ischemia-reperfusion injury (CIRI) in patients with cerebral small vessel disease (CSVD). Methods A total of 138 patients with CSVD complicated with CIRI admitted to our hospital from May 2018 to May 2019 were selected and divided into the CIRI group (138 cases of patients with cerebral vascular disease complicated with CIRI) and the control group [60 cases with no abnormalities in cranial magnetic resonance imaging (MRI) in healthy volunteers]; the results of craniocerebral MRI were subsequently recorded. The serum levels of miRNA-183 were detected by quantitative real-time polymerase chain (RT-qPCR), and the levels of interleukin-6 (IL-6), IL-8, IL-1β, and tumor necrosis factor-α (TNF-α) were determined by enzyme-linked immunosorbent assay (ELISA). A correlation analysis of serum miRNA-183 level and imaging lesion characteristics in patients with CSVD was also conducted. Results RT-qPCR showed that the peripheral blood miRNA-183 level in the CIRI group was increased compared to that in the control group; the level of miRNA-183 in the control group was 30.03±6.32, while the level of miRNA-183 in the CIRI group was 36.78±10.11, which was a statistically significant difference (t=2.475, P<0.05). Compared with the control group, the patient levels of TNF-α, IL-6, IL-8, and IL-1β in the CIRI group were significantly increased (P<0.05). Correlation analysis showed that the serum miRNA-183 level in the CIRI group was positively correlated with an increase of imaging lesions (r=0.997, P<0.05). Conclusions The level of miRNA-183 in CIRI patients with CSVD was higher than that of controls, and the level of miRNA-183 was positively correlated with the increase of imaging lesions.
Collapse
Affiliation(s)
- Zigen Liao
- Department of Neurology, Second Affiliated Hospital, University of South China, Hengyang, China
| | - Hui Sun
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Yanqun Chang
- Department of Pediatrics, Guangdong Women and Children Hospital, Guangzhou, China
| | - Hui Chen
- Department of Emergency, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|