1
|
Ballók B, Schranc Á, Tóth I, Somogyi P, Tolnai J, Peták F, Fodor GH. Comparison of the respiratory effects of commonly utilized general anaesthesia regimes in male Sprague-Dawley rats. Front Physiol 2023; 14:1249127. [PMID: 37791348 PMCID: PMC10544940 DOI: 10.3389/fphys.2023.1249127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/05/2023] [Indexed: 10/05/2023] Open
Abstract
Background: Respiratory parameters in experimental animals are often characterised under general anaesthesia. However, anaesthesia regimes may alter the functional and mechanical properties of the respiratory system. While most anaesthesia regimes have been shown to affect the respiratory system, the effects of general anaesthesia protocols commonly used in animal models on lung function have not been systematically compared. Methods: The present study comprised 40 male Sprague-Dawley rats divided into five groups (N = 8 in each) according to anaesthesia regime applied: intravenous (iv) Na-pentobarbital, intraperitoneal (ip) ketamine-xylazine, iv propofol-fentanyl, inhaled sevoflurane, and ip urethane. All drugs were administered at commonly used doses. End-expiratory lung volume (EELV), airway resistance (Raw) and tissue mechanics were measured in addition to arterial blood gas parameters during mechanical ventilation while maintaining positive end-expiratory pressure (PEEP) values of 0, 3, and 6 cm H2O. Respiratory mechanics were also measured during iv methacholine (MCh) challenges to assess bronchial responsiveness. Results: While PEEP influenced baseline respiratory mechanics, EELV and blood gas parameters (p < 0.001), no between-group differences were observed (p > 0.10). Conversely, significantly lower doses of MCh were required to achieve the same elevation in Raw under ketamine-xylazine anaesthesia compared to the other groups. Conclusion: In the most frequent rodent model of respiratory disorders, no differences in baseline respiratory mechanics or function were observed between commonly used anaesthesia regimes. Bronchial hyperresponsiveness in response to ketamine-xylazine anaesthesia should be considered when designing experiments using this regime. The findings of the present study indicate commonly used anaesthetic regimes allow fair comparison of respiratory mechanics in experimental animals undergoing any of the examined anaesthesia protocols.
Collapse
Affiliation(s)
- Bence Ballók
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Álmos Schranc
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Unit for Anaesthesiological Investigations, Department of Anaesthesiology, Pharmacology, Intensive Care, and Emergency Medicine, University of Geneva, Geneva, Switzerland
| | - Ibolya Tóth
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Petra Somogyi
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - József Tolnai
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Ferenc Peták
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Gergely H. Fodor
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| |
Collapse
|
2
|
Benković V, Milić M, Oršolić N, Horvat Knežević A, Brozović G, Borojević N. Brain DNA damaging effects of volatile anesthetics and 1 and 2 Gy gamma irradiation in vivo: Preliminary results. Toxicol Ind Health 2023; 39:67-80. [PMID: 36602468 DOI: 10.1177/07482337221145599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Although both can cause DNA damage, the combined impact of volatile anesthetics halothane/sevoflurane/isoflurane and radiotherapeutic exposure on sensitive brain cells in vivo has not been previously analyzed. Healthy Swiss albino male mice (240 in total, 48 groups) were exposed to either halothane/sevoflurane/isoflurane therapeutic doses alone (2 h); 1 or 2 gray of gamma radiation alone; or combined exposure. Frontal lobe brain samples from five animals were taken immediately and 2, 6, and 24 h after exposure. DNA damage and cellular repair index were analyzed using the alkaline comet assay and the tail intensity parameter. Elevated tail intensity levels for sevoflurane/halothane were the highest at 6 h and returned to baseline within 24 h for sevoflurane, but not for halothane, while isoflurane treatment caused lower tail intensity than control values. Combined exposure demonstrated a slightly halothane/sevoflurane protective and isoflurane protective effect, which was stronger for 2 than for 1 gray. Cellular repair indices and tail intensity histograms indicated different modes of action in DNA damage creation. Isoflurane/sevoflurane/halothane preconditioning demonstrated protective effects in sensitive brain cells in vivo. Owing to the constant increases in the combined use of radiotherapy and volatile anesthetics, further studies should explore the mechanisms behind these effects, including longer and multiple exposure treatments and in vivo brain tumor models.
Collapse
Affiliation(s)
- Vesna Benković
- Faculty of Science, 117036University of Zagreb, Zagreb, Croatia
| | - Mirta Milić
- Mutagenesis Unit, 118938Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Nada Oršolić
- Faculty of Science, 117036University of Zagreb, Zagreb, Croatia
| | | | - Gordana Brozović
- Department of Anesthesiology, Reanimatology and ICU, University Hospital for Tumors, 499232Sestre Milosrdnice University Hospital Centre, Zagreb, Croatia.,Faculty of Dental Medicine and Health, 84992University of Osijek, Osijek, Croatia
| | - Nikola Borojević
- 8256Warrington and Halton Teaching Hospitals NHS Foundation Trust, Warrington, UK
| |
Collapse
|
3
|
Grüßer L, Blaumeiser-Debarry R, Krings M, Kremer B, Höllig A, Rossaint R, Coburn M. Argon attenuates the emergence of secondary injury after traumatic brain injury within a 2-hour incubation period compared to desflurane: an in vitro study. Med Gas Res 2017; 7:93-100. [PMID: 28744361 PMCID: PMC5510299 DOI: 10.4103/2045-9912.208512] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite years of research, treatment of traumatic brain injury (TBI) remains challenging. Considerable data exists that some volatile anesthetics might be neuroprotective. However, several studies have also revealed a rather neurotoxic profile of anesthetics. In this study, we investigated the effects of argon 50%, desflurane 6% and their combination in an in vitro TBI model with incubation times similar to narcotic time slots in a daily clinical routine. Organotypic hippocampal brain slices of 5- to 7-day-old mice were cultivated for 14 days before TBI was performed. Slices were eventually incubated for 2 hours in an atmosphere containing no anesthetic gas, argon 50% or desflurane 6% or both. Trauma intensity was evaluated via fluorescent imagery. Our results show that neither argon 50% nor desflurane 6% nor their combination could significantly reduce the trauma intensity in comparison to the standard atmosphere. However, in comparison to desflurane 6%, argon 50% displayed a rather neuroprotective profile within the first 2 hours after a focal mechanical trauma (P = 0.015). A 2-hour incubation in an atmosphere containing both gases, argon 50% and desflurane 6%, did not result in significant effects in comparison to the argon 50% group or the desflurane 6% group. Our findings demonstrate that within a 2-hour incubation time neither argon nor desflurane could affect propidium iodide-detectable cell death in an in vitro TBI model in comparison to the standard atmosphere, although cell death was less with argon 50% than with desflurane 6%. The results show that within this short time period processes concerning the development of secondary injury are already taking place and may be manipulated by argon.
Collapse
Affiliation(s)
- Linda Grüßer
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | | | - Matthias Krings
- Department of Anesthesiology and Intensive Care, Medizinisches Zentrum StaedteRegion Aachen, Wuerselen, Germany
| | - Benedikt Kremer
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Rolf Rossaint
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Mark Coburn
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
4
|
TASK channels contribute to neuroprotective action of inhalational anesthetics. Sci Rep 2017; 7:44203. [PMID: 28276488 PMCID: PMC5343576 DOI: 10.1038/srep44203] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/06/2017] [Indexed: 01/13/2023] Open
Abstract
Postconditioning with inhalational anesthetics can reduce ischemia-reperfusion brain injury, although the cellular mechanisms for this effect have not been determined. The current study was designed to test if TASK channels contribute to their neuroprotective actions. Whole cell recordings were used to examine effects of volatile anesthetic on TASK currents in cortical neurons and to verify loss of anesthetic-activated TASK currents from TASK−/− mice. A transient middle cerebral artery occlusion (tMCAO) model was used to establish brain ischemia-reperfusion injury. Quantitative RT-PCR analysis revealed that TASK mRNA was reduced by >90% in cortex and hippocampus of TASK−/− mice. The TASK−/− mice showed a much larger region of infarction than C57BL/6 J mice after tMCAO challenge. Isoflurane or sevoflurane administered after the ischemic insult reduced brain infarct percentage and neurological deficit scores in C57BL/6 J mice, these effect were reduced in TASK−/− mice. Whole cell recordings revealed that the isoflurane-activated background potassium current observed in cortical pyramidal neurons from wild type mice was conspicuously reduced in TASK−/− mice. Our studies demonstrate that TASK channels can limit ischemia-reperfusion damage in the cortex, and postconditioning with volatile anesthetics provides neuroprotective actions that depend, in part, on activation of TASK currents in cortical neurons.
Collapse
|
5
|
Wang H, Li P, Xu N, Zhu L, Cai M, Yu W, Gao Y. Paradigms and mechanisms of inhalational anesthetics mediated neuroprotection against cerebral ischemic stroke. Med Gas Res 2016; 6:194-205. [PMID: 28217291 PMCID: PMC5223310 DOI: 10.4103/2045-9912.196901] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cerebral ischemic stroke is a leading cause of serious long-term disability and cognitive dysfunction. The high mortality and disability of cerebral ischemic stroke is urging the health providers, including anesthesiologists and other perioperative professioners, to seek effective protective strategies, which are extremely limited, especially for those perioperative patients. Intriguingly, several commonly used inhalational anesthetics are recently suggested to possess neuroprotective effects against cerebral ischemia. This review introduces multiple paradigms of inhalational anesthetic treatments that have been investigated in the setting of cerebral ischemia, such as preconditioning, proconditioning and postconditioning with a variety of inhalational anesthetics. The pleiotropic mechanisms underlying these inhalational anesthetics-afforded neuroprotection against stroke are also discussed in detail, including the common pathways shared by most of the inhalational anesthetic paradigms, such as anti-excitotoxicity, anti-apoptosis and anti-inflammation. There are also distinct mechanisms involved in specific paradigms, such as preserving blood brain barrier integrity, regulating cerebral blood flow and catecholamine release. The ready availability of these inhalational anesthetics bedside and renders them a potentially translatable stroke therapy attracting great efforts for understanding of the underlying mechanisms.
Collapse
Affiliation(s)
- Hailian Wang
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China; Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peiying Li
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Na Xu
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ling Zhu
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mengfei Cai
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanqin Gao
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China; Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
6
|
Bjørn-Yoshimoto WE, Underhill SM. The importance of the excitatory amino acid transporter 3 (EAAT3). Neurochem Int 2016; 98:4-18. [PMID: 27233497 DOI: 10.1016/j.neuint.2016.05.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 05/09/2016] [Accepted: 05/17/2016] [Indexed: 12/21/2022]
Abstract
The neuronal excitatory amino acid transporter 3 (EAAT3) is fairly ubiquitously expressed in the brain, though it does not necessarily maintain the same function everywhere. It is important in maintaining low local concentrations of glutamate, where its predominant post-synaptic localization can buffer nearby glutamate receptors and modulate excitatory neurotransmission and synaptic plasticity. It is also the main neuronal cysteine uptake system acting as the rate-limiting factor for the synthesis of glutathione, a potent antioxidant, in EAAT3 expressing neurons, while on GABAergic neurons, it is important in supplying glutamate as a precursor for GABA synthesis. Several diseases implicate EAAT3, and modulation of this transporter could prove a useful therapeutic approach. Regulation of EAAT3 could be targeted at several points for functional modulation, including the level of transcription, trafficking and direct pharmacological modulation, and indeed, compounds and experimental treatments have been identified that regulate EAAT3 function at different stages, which together with observations of EAAT3 regulation in patients is giving us insight into the endogenous function of this transporter, as well as the consequences of altered function. This review summarizes work done on elucidating the role and regulation of EAAT3.
Collapse
Affiliation(s)
- Walden E Bjørn-Yoshimoto
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 København Ø, Denmark
| | - Suzanne M Underhill
- National Institute of Mental Health, National Institutes of Health, 35 Convent Drive Room 3A: 210 MSC3742, Bethesda, MD 20892-3742, USA.
| |
Collapse
|
7
|
Krings M, Höllig A, Liu J, Grüsser L, Rossaint R, Coburn M. Desflurane impairs outcome of organotypic hippocampal slices in an in vitro model of traumatic brain injury. Med Gas Res 2016; 6:3-9. [PMID: 27826417 PMCID: PMC5075680 DOI: 10.4103/2045-9912.179338] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Decreased mortality and disability after traumatic brain injury is a significant medical challenge. Desflurane, a widely used volatile anesthetic has proven to be neuroprotective in a variety of in vitro and in vivo models of ischemic brain injury. The aim of this study was to investigate whether desflurane exhibits neuroprotective properties in an in vitro model of traumatic brain injury. Organotypic hippocampal slice cultures were prepared from brains of 5–7-day-old C57/BL6 mouse pups. After 14 days of culture, the slices were subjected to a focal mechanical trauma and thereafter incubated with three different concentrations of desflurane (2, 4 and 6%) for 2, 24 and 72 hours. Cell injury was assessed with propodium iodide uptake. Our results showed that after 2 hours of desflurane exposure, no significant change in trauma intensity was observed. However, 2% and 4% desflurane could reduce the trauma intensity significantly in the no trauma group than in the no desflurane and trauma group. Incubation with 4% desflurane for 24 hours doubled the trauma intensity in comparison to the trauma control group and the trauma intensity further increased after 72 hours of incubation. Furthermore, a dose-dependent increase of trauma intensity after 24 hours exposure was observed. Our results suggest that a general neuroprotective attribute of desflurane in an in vitro model of traumatic brain injury was not observed.
Collapse
Affiliation(s)
- Matthias Krings
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Jingjin Liu
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Linda Grüsser
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Rolf Rossaint
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Mark Coburn
- Department of Anesthesiology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
8
|
Yu H, Deng J, Zuo Z. High-fat diet reduces neuroprotection of isoflurane post-treatment: Role of carboxyl-terminal modulator protein-Akt signaling. Obesity (Silver Spring) 2014; 22:2396-405. [PMID: 25142024 PMCID: PMC4224981 DOI: 10.1002/oby.20879] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 08/02/2014] [Accepted: 08/05/2014] [Indexed: 02/06/2023]
Abstract
OBJECTIVE High-fat diet (HFD) contributes to the increased prevalence of obesity and hyperlipidemia in young adults, a possible cause for their recent increase in stroke. Isoflurane post-treatment provides neuroprotection. Isoflurane post-treatment induced neuroprotection in HFD-fed mice was determined. METHODS Six-week old CD-1 male mice were fed HFD or regular diet (RD) for 5 or 10 weeks. Their hippocampal slices (400 µm) were subjected to oxygen-glucose deprivation (OGD). Some slices were exposed to isoflurane for 30 min immediately after OGD. Some mice had a 90-min middle cerebral arterial occlusion and were post-treated with 2% isoflurane for 30 min. RESULTS OGD time-dependently induced cell injury. This injury was dose-dependently reduced by isoflurane. The effect was apparent at 1% or 2% isoflurane in RD-fed mice but required 3% isoflurane in HFD-fed mice. HFD influenced the isoflurane effects in DG. OGD increased carboxyl-terminal modulator protein (CTMP), an Akt inhibitor, and decreased Akt signaling. Isoflurane reduced these effects. LY294002, an Akt activation inhibitor, attenuated the isoflurane effects. HFD increased CTMP and reduced Akt signaling. Isoflurane improved neurological outcome in the RD-fed mice but not in the HFD-fed mice. CONCLUSIONS HFD attenuated isoflurane post-treatment-induced neuroprotection possibly because of decreased prosurvival Akt signaling.
Collapse
Affiliation(s)
- Hai Yu
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia 22908
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China 610041
| | - Jiao Deng
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia 22908
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China 710032
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia 22908
| |
Collapse
|
9
|
Ren X, Wang Z, Ma H, Zuo Z. Sevoflurane postconditioning provides neuroprotection against brain hypoxia-ischemia in neonatal rats. Neurol Sci 2014; 35:1401-4. [PMID: 24705859 DOI: 10.1007/s10072-014-1726-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 03/15/2014] [Indexed: 10/25/2022]
Abstract
Application of volatile anesthetics after brain ischemia provides neuroprotection in adult animals (anesthetic postconditioning). We tested whether postconditioning with sevoflurane, the most commonly used general anesthetic in pediatric anesthesia, reduced neonatal brain injury in rats. Seven-day-old Sprague-Dawley rats were subjected to brain hypoxia-ischemia (HI). They were postconditioned with sevoflurane in the presence or absence of 5-hydroxydecanoic acid, a mitochondrial KATP channel inhibitor. Sevoflurane postconditioning dose-dependently reduced brain tissue loss observed 7 days after brain HI. This effect was induced by clinically relevant concentrations and abolished by 5-hydroxydecanoic acid. These results suggest that sevoflurane postconditioning protects neonatal brain against brain HI via mitochondrial KATP channels.
Collapse
Affiliation(s)
- Xiaoyan Ren
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | | | | | | |
Collapse
|
10
|
Deng J, Lei C, Chen Y, Fang Z, Yang Q, Zhang H, Cai M, Shi L, Dong H, Xiong L. Neuroprotective gases – Fantasy or reality for clinical use? Prog Neurobiol 2014; 115:210-45. [DOI: 10.1016/j.pneurobio.2014.01.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/03/2014] [Accepted: 01/03/2014] [Indexed: 12/17/2022]
|
11
|
Zhu X, Yin J, Li L, Ma L, Tan H, Deng J, Chen S, Zuo Z. Electroacupuncture preconditioning-induced neuroprotection may be mediated by glutamate transporter type 2. Neurochem Int 2013; 63:302-8. [PMID: 23831620 PMCID: PMC3758789 DOI: 10.1016/j.neuint.2013.06.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 06/21/2013] [Accepted: 06/28/2013] [Indexed: 11/27/2022]
Abstract
Electroacupuncture has been shown to induce a preconditioning effect in the brain. The mechanisms for this protection are not fully elucidated. We hypothesize that this protection is mediated by excitatory amino acid transporters (EAATs) that have been shown to be neuroprotective. To test this hypothesis, two-month old male Sprague-Dawley rats and EAAT type 3 (EAAT3) knockout mice received or did not receive 30-min electroacupuncture once a day for five consecutive days. They were subjected to a 120-min middle cerebral arterial occlusion (MCAO) at 24h after the last electroacupuncture. Neurological outcome was assessed 2days after the MCAO. Brain tissues were harvested at 24h after the last electroacupuncture for Western blotting. Rats subjected to electroacupuncture at the Baihui acupoint had smaller brain infarct volumes and better neurological deficit scores than control rats. Electroacupuncture increased EAAT type 2 (EAAT2) in the cerebral cortex, tended to increase EAAT3 in the hippocampus, and had no effect on EAAT type 1 expression. Dihydrokainate, an EAAT2 inhibitor, worsened the neurological outcome of rats with electroacupuncture pretreatment. Electroacupuncture pretreatment at the Baihui acupoint increased EAAT2 in the cerebral cortex and improved the neurological outcome of EAAT3 knockout mice. Together, our results suggest that EAAT2 may mediate the electroacupuncture preconditioning-induced neuroprotection.
Collapse
Affiliation(s)
- Xiaoling Zhu
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia 22908, USA
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi’an, 710032, China
| | - Jinbo Yin
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia 22908, USA
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University Chongqing 400037, China
| | - Liaoliao Li
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Lei Ma
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi’an, 710032, China
| | - Hongying Tan
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia 22908, USA
- Department of Anesthesiology, Cancer Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Jiao Deng
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia 22908, USA
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi’an, 710032, China
| | - Shaoyang Chen
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi’an, 710032, China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia 22908, USA
| |
Collapse
|
12
|
Li L, Deng J, Zuo Z. Glutamate transporter type 3 mediates isoflurane preconditioning-induced acute phase of neuroprotection in mice. Brain Res Bull 2013; 98:23-9. [PMID: 23827345 DOI: 10.1016/j.brainresbull.2013.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 06/19/2013] [Accepted: 06/24/2013] [Indexed: 11/26/2022]
Abstract
A pre-exposure to isoflurane reduces ischemic brain injury in rodents (isoflurane preconditioning). This neuroprotection has acute and delayed phases. Our previous in vitro studies suggest that the acute phase may involve excitatory amino acid transporters (EAATs). We determine whether this protection involves EAAT3, the major neuronal EAAT. Adult male EAAT3 knockout mice and their wild-type littermates were exposed or were not exposed to 1.5% isoflurane for 30 min. Sixty minutes later, they were subjected to a 90- or 60-min middle cerebral arterial occlusion (MCAO). Their neurological outcomes were evaluated 24h after the MCAO. In another experiment, cerebral cortex was harvested for Western blotting at 30 min after animals were exposed to 1.5% isoflurane for 30 min. Here, we showed that isoflurane reduced brain infarct volumes and improved neurological functions of wild-type mice after a 90-min MCAO. However, isoflurane pre-exposure did not change the neurological outcome of EAAT3 knockout mice no matter whether the MCAO was for 90 min or 60 min. Isoflurane increased phospho-Akt, a survival-promoting protein, in the wild-type mice but not in the EAAT3 knockout mice. The isoflurane-induced neuroprotection in the wild-type mice was abolished by LY294004, an Akt activation inhibitor. LY294004 alone did not affect the neurological outcome of the wild-type or EAAT3 knockout mice after focal brain ischemia. These results suggest that the isoflurane preconditioning-induced acute phase of neuroprotection involves EAAT3. The downstream event includes Akt activation.
Collapse
Affiliation(s)
- Liaoliao Li
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, United States
| | | | | |
Collapse
|
13
|
Hu X, Zhang Y, Li W, Liu J, Li Y. Preconditioning with sevoflurane ameliorates spatial learning and memory deficit after focal cerebral ischemia-reperfusion in rats. Int J Dev Neurosci 2013; 31:328-33. [PMID: 23628555 DOI: 10.1016/j.ijdevneu.2013.04.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 03/31/2013] [Accepted: 04/21/2013] [Indexed: 11/16/2022] Open
Abstract
Previous studies have demonstrated that sevoflurane could attenuate cerebral neuron necrosis and apoptosis in ischemia-reperfusion models in rats. The aim of our study was to investigate the effect of preconditioning with sevoflurane on spatial learning and memory ability after focal cerebral ischemia-reperfusion injury in rats and its potential mechanisms. Focal cerebral ischemia was performed via 1h of middle cerebral artery occlusion (MCAO) followed by reperfusion. Before ischemia, rats were subjected to preconditioning with inhalation of 2.4% sevoflurane for 1h. The spatial learning and memory ability of rats was measured by the Morris water maze. The activity of choline acetyltransferase (ChAT) in hippocampus CA1 region was observed by immunohistochemistry method. We found MCAO elicited a significant decrease of the ability of spatial learning and memory in contrast to the sham surgery controls. However, preconditioning with sevoflurane resulted in significantly ameliorates spatial learning and memory deficit induced by MCAO. Furthermore, the number of ChAT positive cells in hippocampus CA1 region in sevoflurane preconditioning group was striking more than that of ischemia-reperfusion group. All results suggested that preconditioning with 2.4% sevoflurane could ameliorate the ability of spatial learning and memory after focal cerebral ischemia-reperfusion in rats via protecting the cholinergic neurons in hippocampal CA1 region.
Collapse
Affiliation(s)
- Xianwen Hu
- Department of Anesthesiology, The Second Hospital of Anhui Medical University, Hefei, Anhui Province 230601, PR China
| | | | | | | | | |
Collapse
|
14
|
Lujan HL, DiCarlo SE. Cardiac output, at rest and during exercise, before and during myocardial ischemia, reperfusion, and infarction in conscious mice. Am J Physiol Regul Integr Comp Physiol 2013; 304:R286-95. [PMID: 23302959 DOI: 10.1152/ajpregu.00517.2012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Multiple systems and regulatory strategies interact to control cardiac homeostasis. In fact, regulated systems, feedback controls, and redundant control mechanisms dominate in whole animals. Accordingly, molecular and cellular tools and techniques must be utilized in complex models with multiple systems and regulatory strategies to fully appreciate the physiological context. Currently, these techniques are mainly performed under conditions remote from the normal in vivo condition; thus, the extrapolation of molecular changes to the in vivo situation and the facilitation of translational aspect of the findings are limited. A major obstacle has been the reliance on preparations that do not mimic the clinical or physiological situation. This is particularly true regarding measurements of cardiac function in mice. To address these concerns, we used a permanently implanted Doppler ultrasonic flow probe on the ascending aorta and coronary artery occluder for repeated measurements of ascending aortic blood flow (cardiac output) in conscious mice, at rest and during exercise, before and during coronary artery occlusion/reperfusion and infarction. The conscious mouse model permits detailed monitoring of within-animal changes in cardiac function during myocardial ischemia, reperfusion, and infarction in an intact, complex model free of the confounding influences of anesthetics, surgical trauma, and restraint stress. Results from this study suggest that previous protocols may have overestimated resting baseline values and underestimated cardiac output reserve. Using these procedures in currently available spontaneous or engineered mouse mutants has the potential to be of major importance for advancing the concepts and methods that drive cardiovascular research.
Collapse
Affiliation(s)
- Heidi L Lujan
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | |
Collapse
|
15
|
Lujan HL, Janbaih H, Feng HZ, Jin JP, DiCarlo SE. Myocardial ischemia, reperfusion, and infarction in chronically instrumented, intact, conscious, and unrestrained mice. Am J Physiol Regul Integr Comp Physiol 2012; 302:R1384-400. [PMID: 22538514 DOI: 10.1152/ajpregu.00095.2012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the United States alone, the National Heart, Lung, and Blood Institute (NHLBI) has invested several hundred million dollars in pursuit of myocardial infarct-sparing therapies. However, due largely to methodological limitations, this investment has not produced any notable clinical application or cardioprotective therapy. Among the major methodological limitations is the reliance on animal models that do not mimic the clinical situation. In this context, the limited use of conscious animal models is of major concern. In fact, whenever possible, studies of cardiovascular physiology and pathophysiology should be conducted in conscious, complex models to avoid the complications associated with the use of anesthesia and surgical trauma. The mouse has significant advantages over other experimental models for the investigation of infarct-sparing therapies. The mouse is inexpensive, has a high throughput, and presents the ability of one to create genetically modified models. However, successful infarct-sparing therapies in anesthetized mice or isolated mouse hearts may not be successful in more complex models, including conscious mice. Accordingly, a conscious mouse model of myocardial ischemia and reperfusion has the potential to be of major importance for advancing the concepts and methods that drive the development of infarct-sparing therapies. Therefore, we describe, for the first time, the use of an intact, conscious, and unrestrained mouse model of myocardial ischemia-reperfusion and infarction. The conscious mouse model permits occlusion and reperfusion of the left anterior descending coronary artery in an intact, complex model free of the confounding influences of anesthetics and surgical trauma. This methodology may be adopted for advancing the concepts and ideas that drive cardiovascular research.
Collapse
Affiliation(s)
- Heidi L Lujan
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | | | |
Collapse
|
16
|
Ye Z, Huang YM, Wang E, Zuo ZY, Guo QL. Sevoflurane-induced delayed neuroprotection involves mitoKATP channel opening and PKC ε activation. Mol Biol Rep 2012; 39:5049-57. [DOI: 10.1007/s11033-011-1290-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 11/30/2011] [Indexed: 11/29/2022]
|
17
|
Sloan RC, Rosenbaum M, O'Rourke D, Oppelt K, Frasier CR, Waston CA, Allan AG, Brown DA. High doses of ketamine-xylazine anesthesia reduce cardiac ischemia-reperfusion injury in guinea pigs. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2011; 50:349-354. [PMID: 21640030 PMCID: PMC3103285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 09/24/2010] [Accepted: 11/15/2010] [Indexed: 05/30/2023]
Abstract
Choosing an appropriate anesthetic protocol that will have minimal effect on experimental design can be difficult. Guinea pigs have highly variable responses to a variety of injectable anesthetics, including ketamine-xylazine (KX). Because of this variability, supplemental doses often are required to obtain an adequate plane of anesthesia. Our group studies the isolated guinea pig heart, and we must anesthetize guinea pigs prior to harvesting this organ. In this study, we sought to determine whether a higher dose of KX protected isolated guinea pig hearts against myocardial ischemia-reperfusion injury. Male Hartley guinea pigs (Crl:HA; 275 to 300 g; n = 14) were anesthetized with either of 2 doses of KX (K: 85 mg/kg, X: 15 mg/kg; or K: 200 mg/kg, X: 60 mg/kg). After thoracotomy, hearts underwent 20 min of ischemia followed by 2 h of reperfusion. The high dose of KX significantly reduced myocardial infarct size as compared with the low dose (36% ± 3% and 51% ± 6%, respectively). Furthermore, the high dose of KX improved hemodynamic function over that associated with the low dose as measured by increases in both left ventricular developed pressure (49 ± 4 and 30 ± 8 mm Hg, respectively) and maximal rate of left ventricular relaxation (-876 ± 70 and -576 ± 120 mm Hg/s, respectively). However, the high dose of KX did not alter the maximal rate of left ventricular contraction or coronary flow. These results suggest that supplementation of KX to ensure an adequate anesthetic plane may introduce unwanted variability in ischemia-reperfusion studies.
Collapse
Affiliation(s)
- Ruben C Sloan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
- Departments of Exercise and Sport Science East Carolina University, Greenville, North Carolina
| | - Matthew Rosenbaum
- Comparative Medicine, East Carolina University, Greenville, North Carolina
| | - Dorcas O'Rourke
- Comparative Medicine, East Carolina University, Greenville, North Carolina
| | - Karen Oppelt
- Comparative Medicine, East Carolina University, Greenville, North Carolina
| | - Chad R Frasier
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Corinne A Waston
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Amanda G Allan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - David A Brown
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
- Departments of Exercise and Sport Science East Carolina University, Greenville, North Carolina
| |
Collapse
|
18
|
Schifilliti D, Grasso G, Conti A, Fodale V. Anaesthetic-related neuroprotection: intravenous or inhalational agents? CNS Drugs 2010; 24:893-907. [PMID: 20932063 DOI: 10.2165/11584760-000000000-00000] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
In designing the anaesthetic plan for patients undergoing surgery, the choice of anaesthetic agent may often appear irrelevant and the best results obtained by the use of a technique or a drug with which the anaesthesia care provider is familiar. Nevertheless, in those surgical procedures (cardiopulmonary bypass, carotid surgery and cerebral aneurysm surgery) and clinical situations (subarachnoid haemorrhage, stroke, brain trauma and post-cardiac arrest resuscitation) where protecting the CNS is a priority, the choice of anaesthetic drug assumes a fundamental role. Treating patients with a neuroprotective agent may be a consideration in improving overall neurological outcome. Therefore, a clear understanding of the relative degree of protection provided by various agents becomes essential in deciding on the most appropriate anaesthetic treatment geared to these objectives. This article surveys the current literature on the effects of the most commonly used anaesthetic drugs (volatile and gaseous inhalation, and intravenous agents) with regard to their role in neuroprotection. A systematic search was performed in the MEDLINE, Cumulative Index to Nursing and Allied Health Literature (CINHAL®) and Cochrane Library databases using the following keywords: 'brain' (with the limits 'newborn' or 'infant' or 'child' or 'neonate' or 'neonatal' or 'animals') AND 'neurodegeneration' or 'apoptosis' or 'toxicity' or 'neuroprotection' in combination with individual drug names ('halothane', 'isoflurane', 'desflurane', 'sevoflurane', 'nitrous oxide', 'xenon', 'barbiturates', 'thiopental', 'propofol', 'ketamine'). Over 600 abstracts for articles published from January 1980 to April 2010, including studies in animals, humans and in vitro, were examined, but just over 100 of them were considered and reviewed for quality. Taken as a whole, the available data appear to indicate that anaesthetic drugs such as barbiturates, propofol, xenon and most volatile anaesthetics (halothane, isoflurane, desflurane, sevoflurane) show neuroprotective effects that protect cerebral tissue from adverse events--such as apoptosis, degeneration, inflammation and energy failure--caused by chronic neurodegenerative diseases, ischaemia, stroke or nervous system trauma. Nevertheless, in several studies, the administration of gaseous, volatile and intravenous anaesthetics (especially isoflurane and ketamine) was also associated with dose-dependent and exposure time-dependent neurodegenerative effects in the developing animal brain. At present, available experimental data do not support the selection of any one anaesthetic agent over the others. Furthermore, the relative benefit of one anaesthetic versus another, with regard to neuroprotective potential, is unlikely to form a rational basis for choice. Each drug has some undesirable adverse effects that, together with the patient's medical and surgical history, appear to be decisive in choosing the most suitable anaesthetic agent for a specific situation. Moreover, it is important to highlight that many of the studies in the literature have been conducted in animals or in vitro; hence, results and conclusions of most of them may not be directly applied to the clinical setting. For these reasons, and given the serious implications for public health, we believe that further investigation--geared mainly to clarifying the complex interactions between anaesthetic drug actions and specific mechanisms involved in brain injury, within a setting as close as possible to the clinical situation--is imperative.
Collapse
Affiliation(s)
- Daniela Schifilliti
- Department of Neuroscience Psychiatric and Anesthesiological Sciences, University of Messina, Messina, Italy
| | | | | | | |
Collapse
|
19
|
β-Amyloid-related peptides potentiate K+-evoked glutamate release from adult rat hippocampal slices. Neurobiol Aging 2010; 31:1164-72. [DOI: 10.1016/j.neurobiolaging.2008.08.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Revised: 07/17/2008] [Accepted: 08/14/2008] [Indexed: 11/21/2022]
|
20
|
Sevoflurane preconditioning reverses impairment of hippocampal long-term potentiation induced by myocardial ischaemia-reperfusion injury. Eur J Anaesthesiol 2010; 26:961-8. [PMID: 19687741 DOI: 10.1097/eja.0b013e328330e968] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND OBJECTIVE We sought to test whether a transient myocardial ischaemia can induce impairment of hippocampal long-term potentiation (LTP) and whether sevoflurane preconditioning can provide robust protective effects on this neurological impairment. METHODS Wistar rats were subjected to a transient coronary artery occlusion for 30 min. Sevoflurane preconditioning was performed by exposure to 1.0 minimum alveolar concentration of sevoflurane for 1 h and washout for 30 min before myocardial ischaemia. Hippocampal LTP was evaluated during a 7-day observation period. The expressions of haem oxygenase-1 mRNA, tumour necrosis factor-alpha mRNA and interleukin-1beta mRNA in the hippocampus were analysed by quantitative reverse transcription-PCR. RESULTS LTP was significantly inhibited 1 and 3 days after the transient myocardial ischaemia in the control group when compared with the animals subjected to a sham operation without coronary occlusion, and the LTP recovered to a normal magnitude 7 days later. Sevoflurane preconditioning remarkably reversed the transient inhibition of LTP observed at 1 and 3 days after myocardial ischaemia. Compared with the sham animals, the expressions of haem oxygenase-1 mRNA, tumour necrosis factor-alpha mRNA and interleukin-1beta mRNA in the hippocampus of the control rats were significantly increased during the early stage after myocardial ischaemia (1-3 days), and the increases of these cytokines were attenuated by sevoflurane pretreatment. CONCLUSION Sevoflurane preconditioning induced neuroprotection against impairment of hippocampal LTP resulting from myocardial ischaemia and reperfusion.
Collapse
|
21
|
Li L, Zuo Z. Isoflurane preconditioning improves short-term and long-term neurological outcome after focal brain ischemia in adult rats. Neuroscience 2009; 164:497-506. [PMID: 19679170 DOI: 10.1016/j.neuroscience.2009.08.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Revised: 08/04/2009] [Accepted: 08/06/2009] [Indexed: 01/07/2023]
Abstract
Isoflurane preconditioning improved short-term neurological outcome after focal brain ischemia in adult rats. It is not known whether desflurane induces a delayed phase of preconditioning in the brain and whether isoflurane preconditioning-induced neuroprotection is long-lasting. Two months-old Sprague-Dawley male rats were exposed to or were not exposed to isoflurane or desflurane for 30 min and then subjected to a 90 min middle cerebral arterial occlusion (MCAO) at 24 h after the anesthetic exposure. Neurological outcome was evaluated at 24 h or 4 weeks after the MCAO. The density of the terminal deoxynucleotidyl transferase biotinylated UTP nick end labeling (TUNEL) positive cells in the penumbral cerebral cortex were assessed 4 weeks after the MCAO. Also, rats were pretreated with isoflurane or desflurane for 30 min. Their cerebral cortices were harvested for quantifying B-cell lymphoma-2 (Bcl-2) expression 24 h later. Here, we showed that pretreatment with 1.1% or 2.2% isoflurane, but not with 6% or 12% desflurane, increased Bcl-2 expression in the cerebral cortex, improved neurological functions and reduced infarct volumes evaluated at 24 h after the MCAO. Isoflurane preconditioning also improved neurological functions and reduced brain infarct volumes in rats evaluated 4 weeks after the MCAO. Isoflurane preconditioning also decreased the density of TUNEL-positive cells in the penumbral cerebral cortex. We conclude that isoflurane preconditioning improves short-term and long-term neurological outcome and reduces delayed cell death after transient focal brain ischemia in adult rats. Bcl-2 may be involved in the isoflurane preconditioning effect. Desflurane pretreatment did not induce a delayed phase of neuroprotection.
Collapse
Affiliation(s)
- L Li
- Department of Anesthesiology, University of Virginia, 1 Hospital Drive, Charlottesville, VA 22908, USA
| | | |
Collapse
|
22
|
Kim JA, Li L, Zuo Z. Isoflurane induces a postconditioning effect on bovine pulmonary arterial endothelial cells exposed to oxygen-glucose deprivation. Eur J Pharmacol 2009; 615:144-9. [PMID: 19464284 DOI: 10.1016/j.ejphar.2009.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2008] [Revised: 04/30/2009] [Accepted: 05/14/2009] [Indexed: 01/08/2023]
Abstract
Application of volatile anesthetics during the onset of reperfusion reduced ischemia-induced cardiac and brain injury (anesthetic postconditioning). This study was designed to evaluate whether volatile anesthetics induced a postconditioning effect in endothelial cells. Bovine pulmonary arterial endothelial cell (BPAEC) cultures were exposed to oxygen-glucose deprivation, a condition to simulate ischemia in vitro, for 3 h. The volatile anesthetics isoflurane and desflurane were applied during the early phase of simulated reperfusion. Cell injury was quantified by lactate dehydrogenase (LDH) release and flow cytometrical measurement after annexin V and propidium iodide staining. Oxygen-glucose deprivation and the subsequent simulated reperfusion increased LDH release and annexin V-positive staining cells, a characteristic of cell apoptosis. Posttreatment with isoflurane, but not desflurane, reduced this cell injury. This protection was apparent even when 2% isoflurane was applied at 60 min after the onset of reperfusion. The isoflurane postconditioning effect was abolished by glybenclamide, a general ATP sensitive K(+) (K(ATP)) channel blocker, 5-hydroxydecanoate, a mitochondrial K(ATP) channel blocker, and chelerythrine, a protein kinase C inhibitor. Diazoxide, a mitochondrial K(ATP) channel activator, applied at the onset of reperfusion also decreased oxygen-glucose deprivation-induced endothelial cell injury. This diazoxide-induced protection was abolished by chelerythrine and 5-hydroxydecanoate. We conclude that isoflurane induced a postconditioning effect in BPAEC. The effective time window of isoflurane postconditioning was from 0 to 60 min after the onset of reperfusion. This isoflurane postconditioning effect may be mediated by mitochondrial K(ATP) channels and PKC. PKC may be downstream of mitochondrial K(ATP) channels for this isoflurane effect.
Collapse
Affiliation(s)
- Jie Ae Kim
- Department of Anesthesiology, University of Virginia, Charlottesville 22908-0710, USA
| | | | | |
Collapse
|
23
|
Matchett GA, Allard MW, Martin RD, Zhang JH. Neuroprotective effect of volatile anesthetic agents: molecular mechanisms. Neurol Res 2009; 31:128-34. [PMID: 19298752 DOI: 10.1179/174313209x393546] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
INTRODUCTION Intra-operative cerebral ischemia can be catastrophic, and volatile anesthetic agents have been recognized for their potential neuroprotective properties since the 1960s. In this review, we examine the neuroprotective effects of five volatile anesthetic agents in current or recent clinical use: isoflurane, sevoflurane, desflurane, halothane and enflurane. METHODS A review of publications in the National Library of Medicine and National Institutes of Health database from 1970 to 2007 was conducted. RESULTS Volatile anesthetic agents have been shown to be neuroprotective in multiple animal works of ischemic brain injury. Short-term neuroprotection (<1 week post-ischemia) in experimental cerebral ischemia has been reported in multiple works, although long-term neuroprotection (> or = 1 week post-ischemia) remains controversial. Comparison works have not demonstrated superiority of one specific volatile agent over another in experimental models of brain injury. Relatively few human works have examined the protective effects of volatile anesthetic agents and conclusive evidence of a neuroprotective effect has yet to emerge from human works. CONCLUSION Proposed mechanisms related to the neuroprotective effect of volatile anesthetic agents include activation of ATP-dependent potassium channels, up-regulation of nitric oxide synthase, reduction of excitotoxic stressors and cerebral metabolic rate, augmentation of peri-ischemic cerebral blood flow and up-regulation of antiapoptotic factors including MAP kinases.
Collapse
Affiliation(s)
- Gerald A Matchett
- Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | | | | | | |
Collapse
|
24
|
Lee IH, Culley DJ, Baxter MG, Xie Z, Tanzi RE, Crosby G. Spatial memory is intact in aged rats after propofol anesthesia. Anesth Analg 2008; 107:1211-5. [PMID: 18806029 DOI: 10.1213/ane.0b013e31817ee879] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND We have previously demonstrated that aged rats have persistent impairment of spatial memory after sedation with nitrous oxide or general anesthesia with isoflurane-nitrous oxide. Propofol has different receptor mechanisms of action and a favorable short-term recovery profile, and it has been proposed that propofol is devoid of enduring effects on cognitive performance. No studies have investigated this question in aged subjects, however, so we designed an experiment to examine the long-term effects of propofol anesthesia on spatial working memory. METHODS Eighteen-mo-old rats were randomized to 2 h of 100% oxygen-propofol anesthesia (n=11) or to a control group that breathed 100% oxygen (n=10). Propofol was administered by continuous infusion via a tail vein catheter. Rats breathed spontaneously and rectal temperature was maintained. Mean arterial blood pressure was measured noninvasively and a venous blood gas was obtained just before discontinuation of propofol. After a 2-day recovery, spatial working memory was assessed for 14 days using a 12-arm radial maze. The number of total errors, number of correct choices to first error, and time to complete the maze was recorded and analyzed using a repeated measure analysis of variance (ANOVA), with P<0.05 being considered statistically significant. RESULTS The average propofol infusion rate was 0.6+/-0.1 mg.kg (-1).min(-1), a rate corresponding to a 50% effective concentration dose in adult rats. Mean arterial blood pressure during anesthesia was 100+/-2 mm Hg and venous blood gases remained in the normal range. There was no difference between the control and previously anesthetized rats on any measure of radial arm maze performance, indicating propofol anesthesia produces no lasting impairment in spatial working memory in aged rats. CONCLUSIONS In aged rats, propofol anesthesia is devoid of the persistent memory effects observed with other general anesthetics in this model. Thus, while it appears that the state of general anesthesia is neither necessary nor sufficient for development of postanesthetic memory impairment, the choice of anesthetics may play a role in late cognitive outcome in the aged.
Collapse
Affiliation(s)
- In Ho Lee
- Department of Anesthesiology, Korea University Guro Hospital, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
25
|
Xu^ X, Kim JA, Zuo Z. Isoflurane preconditioning reduces mouse microglial activation and injury induced by lipopolysaccharide and interferon-gamma. Neuroscience 2008; 154:1002-8. [PMID: 18495358 PMCID: PMC2492758 DOI: 10.1016/j.neuroscience.2008.04.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 03/14/2008] [Accepted: 04/02/2008] [Indexed: 11/30/2022]
Abstract
Activation and injury of microglial cells are involved in a broad range of brain diseases including stroke, brain infection and neurodegenerative diseases. However, there is very little information regarding how to reduce microglial reaction and preserve these cells to provide neuroprotection. Here, we showed that the incubation of C8-B4 mouse microglial cells with lipopolysaccharide (LPS) plus interferon-gamma (IFNgamma) for 24 h decreased the viability of these cells. Pretreatment of these cells with 1%, 2% or 3% isoflurane, a commonly used volatile anesthetic, for 1 h at 30 min before the exposure to LPS plus IFNgamma attenuated the reduction of cell viability (preconditioning effect). LPS plus IFNgamma also activated these microglial cells to express inducible nitric oxide synthase (iNOS) and to induce accumulation of nitrite, a stable oxidation product of nitric oxide, in the incubation medium. Isoflurane preconditioning attenuated these LPS plus IFNgamma effects on the iNOS expression and nitrite accumulation. Aminoguanidine, an iNOS inhibitor, attenuated the LPS plus IFNgamma-induced glutamate release and decrease of microglial viability. Isoflurane preconditioning also reduced LPS plus IFNgamma-induced glutamate release. Exogenous glutamate decreased microglial viability. Finally, the isoflurane preconditioning-induced protection was abolished by chelerythrine, a protein kinase C inhibitor. These results suggest that LPS plus IFNgamma activates the iNOS-nitric oxide-glutamate pathway to induce microglial injury and that this activation is attenuated by isoflurane preconditioning. Protein kinase C may be involved in the isoflurane preconditioning effects.
Collapse
Affiliation(s)
- Xuebing Xu^
- Department of Anesthesiology, University of Virginia, Charlottesville, U.S.A
- Department of Anesthesiology, the First People's Hospital of Guangzhou, Guangzhou, China
| | - Jie Ae Kim
- Department of Anesthesiology, University of Virginia, Charlottesville, U.S.A
- Department of Anesthesiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Korea
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, U.S.A
| |
Collapse
|
26
|
Jung HH, Lee JJ, Washington JM, Zuo Z. Inability of volatile anesthetics to inhibit oxygen-glucose deprivation-induced glutamate release via glutamate transporters and anion channels in rat corticostriatal slices. Brain Res 2008; 1227:234-9. [PMID: 18619419 DOI: 10.1016/j.brainres.2008.06.063] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Revised: 06/16/2008] [Accepted: 06/17/2008] [Indexed: 10/21/2022]
Abstract
Ischemia-induced extracellular glutamate accumulation and the subsequent excitotoxicity contribute significantly to ischemic brain injury. Volatile anesthetics have been shown to reduce ischemic brain injury. Here, we showed that oxygen-glucose deprivation (OGD, to simulate ischemia in vitro) increased extracellular glutamate accumulation in the corticostriatal slices of adult rats. This increased accumulation was reduced by dihydrokinate, a glutamate transporter type 2 inhibitor, and 4,4'-dinitrostilbene-2,2'-disulfonic acid, a blocker for volume-activated anion channels. The volatile anesthetics isoflurane, sevoflurane and desflurane at clinically relevant concentrations did not affect the OGD-induced extracellular glutamate accumulation from brain slices of adult rats. Isoflurane also did not change the OGD-induced extracellular glutamate accumulation from brain slices of newborn/young rats. These results suggest that the OGD-induced glutamate accumulation involves reversed transport of glutamate via glutamate transporters and volume-activated anion channels. Volatile anesthetics may not inhibit this extracellular glutamate accumulation.
Collapse
Affiliation(s)
- Hae-Hyuk Jung
- Department of Anesthesiology, University of Virginia Health System, 1 Hospital Drive, PO Box 800710, Charlottesville, Virginia 22908-0710, USA
| | | | | | | |
Collapse
|
27
|
Postconditioning with isoflurane reduced ischemia-induced brain injury in rats. Anesthesiology 2008; 108:1055-62. [PMID: 18497606 DOI: 10.1097/aln.0b013e3181730257] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Preexposure of brain to isoflurane, a commonly used anesthetic, induces ischemic tolerance. This phenomenon is called isoflurane preconditioning. However, it is not known whether isoflurane application after ischemia provides neuroprotection. METHODS Corticostriatal slices (400 microm) freshly prepared from adult male Sprague-Dawley rats were subjected to a 15-min oxygen-glucose deprivation (OGD; to simulate ischemia in vitro). Isoflurane was applied after OGD. Brain slices were harvested 2 h after OGD for measuring 2,3,5-triphenyltetrazolium chloride (TTC) conversion to quantify cell injury. Adult male Sprague-Dawley rats were also subjected to middle cerebral arterial occlusion for 90 min and then treated with or without 2% isoflurane for 60 min started at the onset of reperfusion. The infarct volumes, neurologic deficit scores, and performance on rotarod were evaluated at 24 h after the onset of reperfusion. RESULTS Isoflurane applied immediately after the 15-min OGD for 30 min dose-dependently reversed the OGD-induced decrease of TTC conversion. The TTC conversion was 34 +/- 16% and 58 +/- 28% of the control, respectively, for OGD alone and OGD plus 2% isoflurane (P < 0.05, n = 12). Application of 2% isoflurane for 30 min started at 10 min after the OGD also reduced the OGD-decreased TTC conversion. The presence of 0.3 microm glibenclamide, a general adenosine 5'-triphosphate-sensitive potassium channel blocker, or 500 microm 5-hydroxydecanoic acid, a mitochondrial adenosine 5'-triphosphate-sensitive potassium channel blocker, during the application of 2% isoflurane abolished the isoflurane preservation of TTC conversion. Application of isoflurane during reperfusion also improved neurologic outcome after brain ischemia. CONCLUSIONS The results suggest that isoflurane administrated after OGD or brain ischemia provides neuroprotection. Mitochondrial adenosine 5'-triphosphate-sensitive potassium channels may be involved in this protection.
Collapse
|
28
|
Gressens P, Dingley J, Plaisant F, Porter H, Schwendimann L, Verney C, Tooley J, Thoresen M. Analysis of neuronal, glial, endothelial, axonal and apoptotic markers following moderate therapeutic hypothermia and anesthesia in the developing piglet brain. Brain Pathol 2007; 18:10-20. [PMID: 17924981 DOI: 10.1111/j.1750-3639.2007.00095.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Hypothermia (HT) by whole body (WBC) or selective head cooling (SHC) reduces hypoxic-ischemic (HI) brain injury; however, whether prolonged hypothermia and/or anesthesia disrupts immature brain development, eg, increases apoptosis, is unknown. Anesthesia increases apoptosis in immature animals. We investigated whether neuroprotective hypothermia and anesthesia disrupts normal brain development. Thirty-eight pigs <24 h old were randomized between five groups and were killed after 72 h: eighteen received a global hypoxic-ischemic insult under anesthesia, eight subsequently cooled by SHC with WBC to T(rectal) 34.5 degrees C for 24 h, followed by 48 h normothermia (NT) at T(rectal) 39.0 degrees C, while 10 remained normothermic. Sixteen underwent anesthetized sham hypoxic-ischemic, six then following normothermia and 10 following hypothermia protocols. There were four normothermic controls. The hypothermia groups demonstrated significant brain hypothermia. In the hypoxic-ischemic groups this conferred approximately 60% neuroprotection reducing histological injury scores in all brain areas. Immunohistochemical/histochemical analyses of neuronal, glial, endothelial, axonal, transcriptional apoptotic markers in areas devoid of histological lesions revealed no hypothermia/normothermia group and differences whether exposed to hypoxic-ischemic or not. Neither 36-h anesthesia nor 24-h hypothermia produced adverse effects at 4-day survival on a panel of brain maturation/neural death markers in newborn pigs. Longer survival studies are necessary to verify the safety of hypothermia in the developing brain.
Collapse
|