1
|
Cylinder DM, van Zundert AA, Solt K, van Swinderen B. Time to Wake Up! The Ongoing Search for General Anesthetic Reversal Agents. Anesthesiology 2024; 140:610-627. [PMID: 38349760 PMCID: PMC10868874 DOI: 10.1097/aln.0000000000004846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
How general anesthetics work remains a topic of ongoing study. A parallel field of research has sought to identify methods to reverse general anesthesia. Reversal agents could shorten patients' recovery time and potentially reduce the risk of postoperative complications. An incomplete understanding of the mechanisms of general anesthesia has hampered the pursuit for reversal agents. Nevertheless, the search for reversal agents has furthered understanding of the mechanisms underlying general anesthesia. The study of potential reversal agents has highlighted the importance of rigorous criteria to assess recovery from general anesthesia in animal models, and has helped identify key arousal systems (e.g., cholinergic, dopaminergic, and orexinergic systems) relevant to emergence from general anesthesia. Furthermore, the effects of reversal agents have been found to be inconsistent across different general anesthetics, revealing differences in mechanisms among these drugs. The presynapse and glia probably also contribute to general anesthesia recovery alongside postsynaptic receptors. The next stage in the search for reversal agents will have to consider alternate mechanisms encompassing the tripartite synapse.
Collapse
Affiliation(s)
- Drew M. Cylinder
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - André A.J. van Zundert
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Department of Anaesthesia and Perioperative Medicine, Royal Brisbane and Women’s Hospital, The University of Queensland, Brisbane, QLD, Australia
| | - Ken Solt
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, U.S.A
- Department of Anaesthesia, Harvard Medical School, Boston, MA, U.S.A
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
2
|
Beitchman JA, Krishna G, Bromberg CE, Thomas TC. Effects of isoflurane and urethane anesthetics on glutamate neurotransmission in rat brain using in vivo amperometry. BMC Neurosci 2023; 24:52. [PMID: 37817064 PMCID: PMC10563344 DOI: 10.1186/s12868-023-00822-3] [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: 03/16/2023] [Accepted: 09/19/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Aspects of glutamate neurotransmission implicated in normal and pathological conditions are predominantly evaluated using in vivo recording paradigms in rats anesthetized with isoflurane or urethane. Urethane and isoflurane anesthesia influence glutamate neurotransmission through different mechanisms; however, real-time outcome measures of potassium chloride (KCl)-evoked glutamate overflow and glutamate clearance kinetics have not been compared within and between regions of the brain. In order to maintain rigor and reproducibility within the literature between the two most common methods of anesthetized in vivo recording of glutamate, we compared glutamate signaling as a function of anesthesia and brain region in the rat strain most used in neuroscience. METHODS In the following experiments, in vivo amperometric recordings of KCl-evoked glutamate overflow and glutamate clearance kinetics (uptake rate and T80) in the cortex, hippocampus, and thalamus were performed using glutamate-selective microelectrode arrays (MEAs) in young adult male, Sprague-Dawley rats anesthetized with either isoflurane or urethane. RESULTS Potassium chloride (KCl)-evoked glutamate overflow was similar under urethane and isoflurane anesthesia in all brain regions studied. Analysis of glutamate clearance determined that the uptake rate was significantly faster (53.2%, p < 0.05) within the thalamus under urethane compared to isoflurane, but no differences were measured in the cortex or hippocampus. Under urethane, glutamate clearance parameters were region-dependent, with significantly faster glutamate clearance in the thalamus compared to the cortex but not the hippocampus (p < 0.05). No region-dependent differences were measured for glutamate overflow using isoflurane. CONCLUSIONS These data support that amperometric recordings of KCl-evoked glutamate under isoflurane and urethane anesthesia result in similar and comparable data. However, certain parameters of glutamate clearance can vary based on choice of anesthesia and brain region. In these circumstances, special considerations are needed when comparing previous literature and planning future experiments.
Collapse
Affiliation(s)
- Joshua A Beitchman
- Department of Child Health, University of Arizona College of Medicine - Phoenix, 425 N. 5th St. | 322 ABC-1 Building, Phoenix, AZ, 85004-2127, USA
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix Children's Hospital, Phoenix, AZ, USA
- College of Graduate Studies, Midwestern University, Glendale, AZ, USA
| | - Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine - Phoenix, 425 N. 5th St. | 322 ABC-1 Building, Phoenix, AZ, 85004-2127, USA
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Caitlin E Bromberg
- Department of Child Health, University of Arizona College of Medicine - Phoenix, 425 N. 5th St. | 322 ABC-1 Building, Phoenix, AZ, 85004-2127, USA
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine - Phoenix, 425 N. 5th St. | 322 ABC-1 Building, Phoenix, AZ, 85004-2127, USA.
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix Children's Hospital, Phoenix, AZ, USA.
- Phoenix VA Healthcare System, Phoenix, AZ, USA.
| |
Collapse
|
3
|
Qiu J, Yang Y, Liu J, Zhao W, Li Q, Zhu T, Liang P, Zhou C. The volatile anesthetic isoflurane differentially inhibits voltage-gated sodium channel currents between pyramidal and parvalbumin neurons in the prefrontal cortex. Front Neural Circuits 2023; 17:1185095. [PMID: 37396397 PMCID: PMC10311640 DOI: 10.3389/fncir.2023.1185095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/25/2023] [Indexed: 07/04/2023] Open
Abstract
Background How volatile anesthetics work remains poorly understood. Modulations of synaptic neurotransmission are the direct cellular mechanisms of volatile anesthetics in the central nervous system. Volatile anesthetics such as isoflurane may reduce neuronal interaction by differentially inhibiting neurotransmission between GABAergic and glutamatergic synapses. Presynaptic voltage-dependent sodium channels (Nav), which are strictly coupled with synaptic vesicle exocytosis, are inhibited by volatile anesthetics and may contribute to the selectivity of isoflurane between GABAergic and glutamatergic synapses. However, it is still unknown how isoflurane at clinical concentrations differentially modulates Nav currents between excitatory and inhibitory neurons at the tissue level. Methods In this study, an electrophysiological recording was applied in cortex slices to investigate the effects of isoflurane on Nav between parvalbumin (PV+) and pyramidal neurons in PV-cre-tdTomato and/or vglut2-cre-tdTomato mice. Results Isoflurane at clinically relevant concentrations produced a hyperpolarizing shift in the voltage-dependent inactivation and slowed the recovery time from the fast inactivation in both cellular subtypes. Since the voltage of half-maximal inactivation was significantly depolarized in PV+ neurons compared to that of pyramidal neurons, isoflurane inhibited the peak Nav currents in pyramidal neurons more potently than those of PV+ neurons (35.95 ± 13.32% vs. 19.24 ± 16.04%, P = 0.036 by the Mann-Whitney test). Conclusions Isoflurane differentially inhibits Nav currents between pyramidal and PV+ neurons in the prefrontal cortex, which may contribute to the preferential suppression of glutamate release over GABA release, resulting in the net depression of excitatory-inhibitory circuits in the prefrontal cortex.
Collapse
Affiliation(s)
- Jingxuan Qiu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Yaoxin Yang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Jin Liu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Wenling Zhao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qian Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Peng Liang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Beitchman JA, Krishna G, Bromberg CE, Thomas TC. Effects of isoflurane and urethane anesthetics on glutamate neurotransmission in rat brain using in vivo amperometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.16.528856. [PMID: 36824899 PMCID: PMC9949081 DOI: 10.1101/2023.02.16.528856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Aspects of glutamate neurotransmission implicated in normal and pathological conditions are often evaluated using in vivo recording paradigms in rats anesthetized with isoflurane or urethane. Urethane and isoflurane anesthesia influence glutamate neurotransmission through different mechanisms; however real-time outcome measures of potassium chloride (KCl)-evoked glutamate overflow and glutamate clearance kinetics have not been compared within and between regions of the brain. In the following experiments, in vivo amperometric recordings of KCl-evoked glutamate overflow and glutamate clearance kinetics (uptake rate and T80) in the cortex, hippocampus and thalamus were performed using glutamate-selective microelectrode arrays (MEAs) in young adult male, Sprague-Dawley rats anesthetized with isoflurane or urethane. Potassium chloride (KCl)-evoked glutamate overflow was similar under urethane and isoflurane anesthesia in all brain regions studied. Analysis of glutamate clearance determined that the uptake rate was significantly faster (53.2%, p<0.05) within the thalamus under urethane compared to isoflurane, but no differences were measured in the cortex or hippocampus. Under urethane, glutamate clearance parameters were region dependent, with significantly faster glutamate clearance in the thalamus compared to the cortex but not the hippocampus (p<0.05). No region dependent differences were measured for glutamate overflow using isoflurane. These data support that amperometric recordings of glutamate under isoflurane and urethane anesthesia result in mostly similar and comparable data. However, certain parameters of glutamate uptake vary based on choice of anesthesia and brain region. Special considerations must be given to these areas when considering comparison to previous literature and when planning future experiments.
Collapse
Affiliation(s)
- Joshua A. Beitchman
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, USA
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
- College of Graduate Studies, Midwestern University, Glendale, AZ, USA
| | - Gokul Krishna
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, USA
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Caitlin E. Bromberg
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, USA
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Theresa Currier Thomas
- BARROW Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, USA
- Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
- Phoenix VA Healthcare System, Phoenix, AZ, USA
| |
Collapse
|
5
|
Hypoxia-triggered O-GlcNAcylation in the brain drives the glutamate-glutamine cycle and reduces sensitivity to sevoflurane in mice. Br J Anaesth 2022; 129:703-715. [PMID: 36031420 DOI: 10.1016/j.bja.2022.06.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 06/11/2022] [Accepted: 06/29/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Hypersensitivity to general anaesthetics predicts adverse postoperative outcomes in patients. Hypoxia exerts extensive pathophysiological effects on the brain; however, whether hypoxia influences sevoflurane sensitivity and its underlying mechanisms remain poorly understood. METHODS Mice were acclimated to hypoxia (oxygen 10% for 8 h day-1) for 28 days and anaesthetised with sevoflurane; the effective concentrations for 50% of the animals (EC50) showing loss of righting reflex (LORR) and loss of tail-pinch withdrawal response (LTWR) were determined. Positron emission tomography-computed tomography, O-glycoproteomics, seahorse analysis, carbon-13 tracing, site-specific mutagenesis, and electrophysiological techniques were performed to explore the underlying mechanisms. RESULTS Compared with the control group, the hypoxia-acclimated mice required higher concentrations of sevoflurane to present LORR and LTWR (EC50LORR: 1.61 [0.03]% vs 1.46 [0.04]%, P<0.01; EC50LTWR: 2.46 [0.14]% vs 2.22 [0.06]%, P<0.01). Hypoxia-induced reduction in sevoflurane sensitivity was correlated with elevation of protein O-linked N-acetylglucosamine (O-GlcNAc) modification in brain, especially in the thalamus, and could be abolished by 6-diazo-5-oxo-l-norleucine, a glutamine fructose-6-phosphate amidotransferase inhibitor, and mimicked by thiamet-G, a selective O-GlcNAcase inhibitor. Mechanistically, O-GlcNAcylation drives de novo synthesis of glutamine from glucose in astrocytes and promotes the glutamate-glutamine cycle, partially via glycolytic flux and activation of glutamine synthetase. CONCLUSIONS Intermittent hypoxia exposure decreased mouse sensitivity to sevoflurane anaesthesia through enhanced O-GlcNAc-dependent modulation of the glutamate-glutamine cycle in the brain.
Collapse
|
6
|
Carton L, Auger F, Laloux C, Durieux N, Kyheng M, Potey C, Bergeron S, Rolland B, Deguil J, Bordet R. Effects of acute ethanol and/or diazepam exposure on immediate and delayed hippocampal metabolite levels in rats anesthetized with isoflurane. Fundam Clin Pharmacol 2022; 36:687-698. [DOI: 10.1111/fcp.12764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 01/13/2022] [Accepted: 01/31/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Louise Carton
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, Degenerative and Vascular Cognitive Disorders, UMR‐S1172, Pharmacology Department Lille France
| | - Florent Auger
- Lille In vivo Imaging and Functional Exploration platform Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41‐UMS 2014‐PLBS Lille France
| | - Charlotte Laloux
- Lille In vivo Imaging and Functional Exploration platform Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41‐UMS 2014‐PLBS Lille France
| | - Nicolas Durieux
- Lille In vivo Imaging and Functional Exploration platform Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41‐UMS 2014‐PLBS Lille France
| | - Maéva Kyheng
- ULR 2694‐METRICS: Évaluation des Technologies de Santé et des Pratiques Médicales Univ. Lille, CHU Lille Lille France
- Département de Biostatistiques CHU Lille Lille France
| | - Camille Potey
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, Degenerative and Vascular Cognitive Disorders, UMR‐S1172, Pharmacology Department Lille France
| | - Sandrine Bergeron
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, Degenerative and Vascular Cognitive Disorders, UMR‐S1172, Pharmacology Department Lille France
| | - Benjamin Rolland
- Service Universitaire d'Addictologie de Lyon CH Le Vinatier, Hospices Civils de Lyon Bron France
- Inserm U1028, CNRS UMR5292 Université Claude Bernard Lyon 1 Bron France
| | - Julie Deguil
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, Degenerative and Vascular Cognitive Disorders, UMR‐S1172, Pharmacology Department Lille France
| | - Régis Bordet
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, Degenerative and Vascular Cognitive Disorders, UMR‐S1172, Pharmacology Department Lille France
| |
Collapse
|
7
|
Li Y, Li J, Shi Y, Zhou X, Feng W, Han L, Ma D, Jiang H, Yuan Y. Urinary Aromatic Amino Acid Metabolites Associated With Postoperative Emergence Agitation in Paediatric Patients After General Anaesthesia: Urine Metabolomics Study. Front Pharmacol 2022; 13:932776. [PMID: 35928271 PMCID: PMC9343964 DOI: 10.3389/fphar.2022.932776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/22/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Emergence agitation (EA) is very common in paediatric patients during recovery from general anaesthesia, but underlying mechanisms remain unknown. This prospective study was designed to profile preoperative urine metabolites and identify potential biomarkers that can predict the occurrence of EA. Methods: A total of 224 patients were screened for recruitment; of those, preoperative morning urine samples from 33 paediatric patients with EA and 33 non-EA gender- and age-matched patients after being given sevoflurane general anaesthesia were analysed by ultra-high-performance liquid chromatography (UHPLC) coupled with a Q Exactive Plus mass spectrometer. Univariate analysis and orthogonal projection to latent structures squares-discriminant analysis (OPLS-DA) were used to analyse these metabolites. The least absolute shrinkage and selection operator (LASSO) regression was used to identify predictive variables. The predictive model was evaluated through the receiver operating characteristic (ROC) analysis and then further assessed with 10-fold cross-validation. Results: Seventy-seven patients completed the study, of which 33 (42.9%) patients developed EA. EA and non-EA patients had many differences in preoperative urine metabolic profiling. Sixteen metabolites including nine aromatic amino acid metabolites, acylcarnitines, pyridoxamine, porphobilinogen, 7-methylxanthine, and 5′-methylthioadenosine were found associated with an increased risk of EA, and they all exhibited higher levels in the EA group than in the non-EA group. The main metabolic pathways involved in these metabolic changes included phenylalanine, tyrosine and tryptophan metabolisms. Among these potential biomarkers, L-tyrosine had the best predictive value with an odds ratio (OR) (95% CI) of 5.27 (2.20–12.63) and the AUC value of 0.81 (0.70–0.91) and was robust with internal 10-fold cross-validation. Conclusion: Urinary aromatic amino acid metabolites are closely associated with EA in paediatric patients, and further validation with larger cohorts and mechanistic studies is needed. Clinical Trial Registration:clinicaltrials.gov, identifier NCT04807998
Collapse
Affiliation(s)
- Yueyue Li
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jingjie Li
- Department of Anaesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhuan Shi
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xuhui Zhou
- Department of Anaesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanqing Feng
- Department of Anaesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Han
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Hong Jiang
- Department of Anaesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Hong Jiang, ; Yongfang Yuan,
| | - Yongfang Yuan
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- *Correspondence: Hong Jiang, ; Yongfang Yuan,
| |
Collapse
|
8
|
Williams RA, Johnson KW, Lee FS, Hemmings HC, Platholi J. A Common Human Brain-Derived Neurotrophic Factor Polymorphism Leads to Prolonged Depression of Excitatory Synaptic Transmission by Isoflurane in Hippocampal Cultures. Front Mol Neurosci 2022; 15:927149. [PMID: 35813074 PMCID: PMC9260310 DOI: 10.3389/fnmol.2022.927149] [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: 04/23/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Multiple presynaptic and postsynaptic targets have been identified for the reversible neurophysiological effects of general anesthetics on synaptic transmission and neuronal excitability. However, the synaptic mechanisms involved in persistent depression of synaptic transmission resulting in more prolonged neurological dysfunction following anesthesia are less clear. Here, we show that brain-derived neurotrophic factor (BDNF), a growth factor implicated in synaptic plasticity and dysfunction, enhances glutamate synaptic vesicle exocytosis, and that attenuation of vesicular BDNF release by isoflurane contributes to transient depression of excitatory synaptic transmission in mice. This reduction in synaptic vesicle exocytosis by isoflurane was acutely irreversible in neurons that release less endogenous BDNF due to a polymorphism (BDNF Val66Met; rs6265) compared to neurons from wild-type mice. These effects were prevented by exogenous application of BDNF. Our findings identify a role for a common human BDNF single nucleotide polymorphism in persistent changes of synaptic function following isoflurane exposure. These short-term persistent alterations in excitatory synaptic transmission indicate a role for human genetic variation in anesthetic effects on synaptic plasticity and neurocognitive function.
Collapse
Affiliation(s)
- Riley A. Williams
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States
| | - Kenneth W. Johnson
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Francis S. Lee
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States,Department of Psychiatry, Sackler Institute for Developmental Psychobiology, Weill Cornell Medicine, New York, NY, United States,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Hugh C. Hemmings
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States,Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Jimcy Platholi
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States,*Correspondence: Jimcy Platholi,
| |
Collapse
|
9
|
Ma Z, Zheng JC, Li T, Xie Z, Kang L. Janus Kinase Mediates Faster Recovery From Sevoflurane Anesthesia Than Isoflurane Anesthesia in the Migratory Locusts. Front Physiol 2022; 13:806746. [PMID: 35431999 PMCID: PMC9006988 DOI: 10.3389/fphys.2022.806746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/24/2022] [Indexed: 12/03/2022] Open
Abstract
Inhalation anesthetics isoflurane and sevoflurane have been widely used in clinical practice for anesthesia. However, the molecular mechanisms underlying the faster recovery from sevoflurane anesthesia than isoflurane anesthesia remain largely undetermined. Herein, we use RNA-seq, RNA interference, quantitative real-time PCR and western blotting to explore the mechanisms of recovery from isoflurane and sevoflurane anesthesia in the migratory locusts. Although the migratory locusts show similar anesthetic responses to these two chemicals in corresponding half-maximal effective concentrations (EC50s), the recovery from sevoflurane anesthesia is significantly faster than that for isoflurane anesthesia after 30 min of anesthetic exposure. Transcriptome analysis shows that those transcripts involved in cytoskeletal components, Janus kinase (JAK) pathway and cuticle protein are differentially expressed in locust brains in response to isoflurane and sevoflurane. RNAi knockdown confirms that Actin, Myosin-like protein 84B (Mlp84B), JAK and cuticle protein NCP56 do not affect anesthetic response of the locusts to these two chemical anesthetics. Moreover, actin, Mlp84B and NCP56 do not affect differential recovery from isoflurane and sevoflurane anesthesia, whereas RNAi knockdown of JAK and its partner STAT5B does not affect anesthetic recovery from isoflurane but elongates recovery duration from sevoflurane anesthesia. Thus, JAK may mediate faster recovery from sevoflurane anesthesia than from isoflurane anesthesia in the migratory locust. This finding provides novel insights into the molecular mechanism underlying faster recovery from sevoflurane anesthesia than isoflurane anesthesia.
Collapse
Affiliation(s)
- Zongyuan Ma
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Jialin C. Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tianzuo Li
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- *Correspondence: Zhongcong Xie,
| | - Le Kang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
- Le Kang,
| |
Collapse
|
10
|
Speigel IA, Hemmings HC. Selective inhibition of gamma aminobutyric acid release from mouse hippocampal interneurone subtypes by the volatile anaesthetic isoflurane. Br J Anaesth 2021; 127:587-599. [PMID: 34384592 DOI: 10.1016/j.bja.2021.06.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The cellular and molecular mechanisms by which general anaesthesia occurs is poorly understood. Hippocampal interneurone subpopulations, which are critical regulators of cognitive function, have diverse neurophysiological and synaptic properties, but their responses to anaesthetics are unclear. METHODS We used live-cell imaging of fluorescent biosensors expressed in mouse hippocampal neurones to delineate interneurone subtype-specific effects of isoflurane on synaptic vesicle exocytosis. The role of voltage-gated sodium channel (Nav) subtype expression in determining isoflurane sensitivity was probed by overexpression or knockdown of specific Nav subtypes in identified interneurones. RESULTS Clinically relevant concentrations of isoflurane differentially inhibited synaptic vesicle exocytosis: to 83.1% (11.7%) of control in parvalbumin-expressing interneurones, and to 58.6% (13.3%) and 64.5% (8.5%) of control in somatostatin-expressing interneurones and glutamatergic neurones, respectively. The relative expression of Nav1.1 (associated with lower sensitivity) and Nav1.6 (associated with higher sensitivity) determined the sensitivity of exocytosis to isoflurane. CONCLUSIONS Isoflurane inhibits synaptic vesicle exocytosis from hippocampal glutamatergic neurones and GABAergic interneurones in a cell-type-specific manner depending on their expression of voltage-gated sodium channel subtypes.
Collapse
Affiliation(s)
- Iris A Speigel
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA.
| | - Hugh C Hemmings
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
11
|
Platholi J, Hemmings HC. Effects of general anesthetics on synaptic transmission and plasticity. Curr Neuropharmacol 2021; 20:27-54. [PMID: 34344292 PMCID: PMC9199550 DOI: 10.2174/1570159x19666210803105232] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 11/22/2022] Open
Abstract
General anesthetics depress excitatory and/or enhance inhibitory synaptic transmission principally by modulating the function of glutamatergic or GABAergic synapses, respectively, with relative anesthetic agent-specific mechanisms. Synaptic signaling proteins, including ligand- and voltage-gated ion channels, are targeted by general anesthetics to modulate various synaptic mechanisms, including presynaptic neurotransmitter release, postsynaptic receptor signaling, and dendritic spine dynamics to produce their characteristic acute neurophysiological effects. As synaptic structure and plasticity mediate higher-order functions such as learning and memory, long-term synaptic dysfunction following anesthesia may lead to undesirable neurocognitive consequences depending on the specific anesthetic agent and the vulnerability of the population. Here we review the cellular and molecular mechanisms of transient and persistent general anesthetic alterations of synaptic transmission and plasticity.
Collapse
Affiliation(s)
- Jimcy Platholi
- Cornell University Joan and Sanford I Weill Medical College Ringgold standard institution - Anesthesiology New York, New York. United States
| | - Hugh C Hemmings
- Cornell University Joan and Sanford I Weill Medical College Ringgold standard institution - Anesthesiology New York, New York. United States
| |
Collapse
|
12
|
Wang CM, Chen WC, Zhang Y, Lin S, He HF. Update on the Mechanism and Treatment of Sevoflurane-Induced Postoperative Cognitive Dysfunction. Front Aging Neurosci 2021; 13:702231. [PMID: 34305576 PMCID: PMC8296910 DOI: 10.3389/fnagi.2021.702231] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Sevoflurane is one of the most widely used anesthetics for the induction and maintenance of general anesthesia in surgical patients. Sevoflurane treatment may increase the incidence of postoperative cognitive dysfunction (POCD), and patients with POCD exhibit lower cognitive abilities than before the operation. POCD affects the lives of patients and places an additional burden on patients and their families. Understanding the mechanism of sevoflurane-induced POCD may improve prevention and treatment of POCD. In this paper, we review the diagnosis of POCD, introduce animal models of POCD in clinical research, analyze the possible mechanisms of sevoflurane-induced POCD, and summarize advances in treatment for this condition.
Collapse
Affiliation(s)
- Cong-Mei Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Wei-Can Chen
- Department of Anesthesiology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Yan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Department of Anesthesiology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China.,Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, Australia.,Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - He-Fan He
- Department of Anesthesiology, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| |
Collapse
|
13
|
Chávez-Pichardo ME, Reyes-Bravo DY, Mendoza-Trejo MS, Marín-López AG, Giordano M, Hernández-Chan N, Domínguez-Marchan K, Ortega-Rosales LC, Rodríguez VM. Brain alterations in GABA, glutamate and glutamine markers after chronic atrazine exposure in the male albino rat. Arch Toxicol 2020; 94:3217-3230. [PMID: 32561961 DOI: 10.1007/s00204-020-02806-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/15/2020] [Indexed: 11/28/2022]
Abstract
Atrazine (ATR; 2-chloro-4-ethylamino-6-isopropylamino-s-triazine) is an herbicide widely used to kill annual grasses and broadleaf weeds in crops such as corn, sorghum, and sugarcane. Studies in rodents have shown that chronic ATR exposure is associated with alterations in the nigrostriatal dopaminergic pathway such as hyperactivity, decreased striatal dopamine levels, and diminished numbers of tyrosine hydroxylase positive cells in substantia nigra pars compacta. However, the effects of ATR on neurotransmitters such as GABA and glutamate have been scarcely studied. To evaluate the impact of ATR on motor and anxiety tasks, tissue levels of GABA, glutamate, glutamine, and extracellular and potassium-evoked release of glutamate in the striatum, we daily exposed Sprague-Dawley male rats to 1 or 10 mg ATR/kg of body weight for 12-14 months. As previously reported, chronic ATR exposure causes hyperactivity in the group exposed to 10 mg ATR/kg and increased anxiety in both groups exposed to ATR. GABA, glutamate, and glutamine levels were differentially altered in brain regions related to nigrostriatal and mesolimbic systems, the amygdala, and the prefrontal cortex. The groups exposed to 10 mg ATR/kg showed increased extracellular levels and release of glutamate in the striatum. These neurochemical alterations could underlie the behavioral changes observed in rats. These results indicate that chronic exposure to the herbicide ATR disrupts the neurochemistry of several brain structures and could be a risk factor for the development of neurodegenerative diseases.
Collapse
Affiliation(s)
- M E Chávez-Pichardo
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Querétaro, 76230, México
| | - D Y Reyes-Bravo
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Querétaro, 76230, México
| | - M S Mendoza-Trejo
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Querétaro, 76230, México
| | - A G Marín-López
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Querétaro, 76230, México
| | - M Giordano
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Querétaro, 76230, México
| | - N Hernández-Chan
- Facultad de Ingeniería, Universidad Autónoma de Querétaro, Querétaro, México
| | - K Domínguez-Marchan
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Querétaro, 76230, México
| | - L C Ortega-Rosales
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Querétaro, 76230, México
| | - V M Rodríguez
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Querétaro, Querétaro, 76230, México.
| |
Collapse
|
14
|
Schildt A, de Vries EFJ, Willemsen ATM, Giacobbo BL, Moraga-Amaro R, Sijbesma JWA, van Waarde A, Sossi V, Dierckx RAJO, Doorduin J. Effect of Dopamine D 2 Receptor Antagonists on [ 18F]-FEOBV Binding. Mol Pharm 2020; 17:865-872. [PMID: 32011892 PMCID: PMC7054895 DOI: 10.1021/acs.molpharmaceut.9b01129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The
interaction of dopaminergic and cholinergic neurotransmission
in, e.g., Parkinson’s disease has been well established. Here,
D2 receptor antagonists were used to assess changes in
[18F]-FEOBV binding to the vesicular acetylcholine transporter
(VAChT) in rodents using positron emission tomography (PET). After
pretreatment with either 10 mg/kg haloperidol, 1 mg/kg raclopride,
or vehicle, 90 min dynamic PET scans were performed with arterial
blood sampling. The net influx rate (Ki) was obtained from Patlak graphical analysis, using a metabolite-corrected
plasma input function and dynamic PET data. [18F]-FEOBV
concentration in whole-blood or plasma and the metabolite-corrected
plasma input function were not significantly changed by the pretreatments
(adjusted p > 0.07, Cohen’s d 0.28–1.89) while the area-under-the-curve (AUC) of the parent
fraction of [18F]-FEOBV was significantly higher after
haloperidol treatment (adjusted p = 0.022, Cohen’s d = 2.51) than in controls. Compared to controls, the AUC
of [18F]-FEOBV, normalized for injected dose and body weight,
was nonsignificantly increased in the striatum after haloperidol (adjusted p = 0.4, Cohen’s d = 1.77) and raclopride
(adjusted p = 0.052, Cohen’s d = 1.49) treatment, respectively. No changes in the AUC of [18F]-FEOBV were found in the cerebellum (Cohen’s d 0.63–0.74). Raclopride treatment nonsignificantly
increased Ki in the striatum 1.3-fold
compared to control rats (adjusted p = 0.1, Cohen’s d = 1.1) while it reduced Ki in the cerebellum by 28% (adjusted p = 0.0004,
Cohen’s d = 2.2) compared to control rats.
Pretreatment with haloperidol led to a nonsignificant reduction in Ki in the striatum (10%, adjusted p = 1, Cohen’s d = 0.44) and a 40–50%
lower Ki than controls in all other brain
regions (adjusted p < 0.0005, Cohen’s d = 3.3–4.7). The changes in Ki induced by the selective D2 receptor antagonist
raclopride can in part be quantified using [18F]-FEOBV
PET imaging. Haloperidol, a nonselective D2/σ receptor
antagonist, either paradoxically decreased cholinergic activity or
blocked off-target [18F]-FEOBV binding to σ receptors.
Hence, further studies evaluating the binding of [18F]-FEOBV
to σ receptors using selective σ receptor ligands are
necessary.
Collapse
Affiliation(s)
- Anna Schildt
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, The Netherlands.,Department of Physics and Astronomy, University of British Columbia, 143-2211 Wesbrook Mall, Vancouver, BC V6T 2B5, Canada
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, The Netherlands
| | - Antoon T M Willemsen
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, The Netherlands
| | - Bruno Lima Giacobbo
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, The Netherlands
| | - Rodrigo Moraga-Amaro
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, The Netherlands
| | - Jürgen W A Sijbesma
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, The Netherlands
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, The Netherlands
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, 143-2211 Wesbrook Mall, Vancouver, BC V6T 2B5, Canada
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, The Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, The Netherlands
| |
Collapse
|
15
|
Syntaxin1A Neomorphic Mutations Promote Rapid Recovery from Isoflurane Anesthesia in Drosophila melanogaster. Anesthesiology 2020; 131:555-568. [PMID: 31356232 DOI: 10.1097/aln.0000000000002850] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Mutations in the presynaptic protein syntaxin1A modulate general anesthetic effects in vitro and in vivo. Coexpression of a truncated syntaxin1A protein confers resistance to volatile and intravenous anesthetics, suggesting a target mechanism distinct from postsynaptic inhibitory receptor processes. Hypothesizing that recovery from anesthesia may involve a presynaptic component, the authors tested whether syntaxin1A mutations facilitated recovery from isoflurane anesthesia in Drosophila melanogaster. METHODS A truncated syntaxin1A construct was expressed in Drosophila neurons. The authors compared effects on isoflurane induction versus recovery in syntaxin1A mutant animals by probing behavioral responses to mechanical stimuli. The authors also measured synaptic responses from the larval neuromuscular junction using sharp intracellular recordings, and performed Western blots to determine whether the truncated syntaxin1A is associated with presynaptic core complexes. RESULTS Drosophila expressing a truncated syntaxin1A (syx, n = 40) were resistant to isoflurane induction for a behavioral responsiveness endpoint (ED50 0.30 ± 0.01% isoflurane, P < 0.001) compared with control (0.240 ± 0.002% isoflurane, n = 40). Recovery from isoflurane anesthesia was also faster, with syx-expressing flies showing greater levels of responsiveness earlier in recovery (reaction proportion 0.66 ± 0.48, P < 0.001, n = 68) than controls (0.22 ± 0.42, n = 68 and 0.33 ± 0.48, n = 66). Measuring excitatory junction potentials of larvae coexpressing the truncated syntaxin1A protein showed a greater recovery of synaptic function, compared with controls (17.39 ± 3.19 mV and 10.29 ± 4.88 mV, P = 0.014, n = 8 for both). The resistance-promoting truncated syntaxin1A was not associated with presynaptic core complexes, in the presence or absence of isoflurane anesthesia. CONCLUSIONS The same neomorphic syntaxin1A mutation that confers isoflurane resistance in cell culture and nematodes also produces isoflurane resistance in Drosophila. Resistance in Drosophila is, however, most evident at the level of recovery from anesthesia, suggesting that the syntaxin1A target affects anesthesia maintenance and recovery processes rather than induction. The absence of truncated syntaxin1A from the presynaptic complex suggests that the resistance-promoting effect of this molecule occurs before core complex formation.
Collapse
|
16
|
Isoflurane Modulates Hippocampal Cornu Ammonis Pyramidal Neuron Excitability by Inhibition of Both Transient and Persistent Sodium Currents in Mice. Anesthesiology 2020; 131:94-104. [PMID: 31166240 DOI: 10.1097/aln.0000000000002753] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Volatile anesthetics inhibit presynaptic voltage-gated sodium channels to reduce neurotransmitter release, but their effects on excitatory neuron excitability by sodium current inhibition are unclear. The authors hypothesized that inhibition of transient and persistent neuronal sodium currents by the volatile anesthetic isoflurane contributes to reduced hippocampal pyramidal neuron excitability. METHODS Whole-cell patch-clamp recordings of sodium currents of hippocampal cornu ammonis pyramidal neurons were performed in acute mouse brain slices. The actions of isoflurane on both transient and persistent sodium currents were analyzed at clinically relevant concentrations of isoflurane. RESULTS The median inhibitory concentration of isoflurane for inhibition of transient sodium currents was 1.0 ± 0.3 mM (~3.7 minimum alveolar concentration [MAC]) from a physiologic holding potential of -70 mV. Currents from a hyperpolarized holding potential of -120 mV were minimally inhibited (median inhibitory concentration = 3.6 ± 0.7 mM, ~13.3 MAC). Isoflurane (0.55 mM; ~2 MAC) shifted the voltage-dependence of steady-state inactivation by -6.5 ± 1.0 mV (n = 11, P < 0.0001), but did not affect the voltage-dependence of activation. Isoflurane increased the time constant for sodium channel recovery from 7.5 ± 0.6 to 12.7 ± 1.3 ms (n = 13, P < 0.001). Isoflurane also reduced persistent sodium current density (median inhibitory concentration = 0.4 ± 0.1 mM, ~1.5 MAC) and resurgent currents. Isoflurane (0.55 mM; ~2 MAC) reduced action potential amplitude, and hyperpolarized resting membrane potential from -54.6 ± 2.3 to -58.7 ± 2.1 mV (n = 16, P = 0.001). CONCLUSIONS Isoflurane at clinically relevant concentrations inhibits both transient and persistent sodium currents in hippocampal cornu ammonis pyramidal neurons. These mechanisms may contribute to reductions in both hippocampal neuron excitability and synaptic neurotransmission.
Collapse
|
17
|
Hao X, Ou M, Zhang D, Zhao W, Yang Y, Liu J, Yang H, Zhu T, Li Y, Zhou C. The Effects of General Anesthetics on Synaptic Transmission. Curr Neuropharmacol 2020; 18:936-965. [PMID: 32106800 PMCID: PMC7709148 DOI: 10.2174/1570159x18666200227125854] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/20/2020] [Accepted: 02/26/2020] [Indexed: 02/08/2023] Open
Abstract
General anesthetics are a class of drugs that target the central nervous system and are widely used for various medical procedures. General anesthetics produce many behavioral changes required for clinical intervention, including amnesia, hypnosis, analgesia, and immobility; while they may also induce side effects like respiration and cardiovascular depressions. Understanding the mechanism of general anesthesia is essential for the development of selective general anesthetics which can preserve wanted pharmacological actions and exclude the side effects and underlying neural toxicities. However, the exact mechanism of how general anesthetics work is still elusive. Various molecular targets have been identified as specific targets for general anesthetics. Among these molecular targets, ion channels are the most principal category, including ligand-gated ionotropic receptors like γ-aminobutyric acid, glutamate and acetylcholine receptors, voltage-gated ion channels like voltage-gated sodium channel, calcium channel and potassium channels, and some second massager coupled channels. For neural functions of the central nervous system, synaptic transmission is the main procedure for which information is transmitted between neurons through brain regions, and intact synaptic function is fundamentally important for almost all the nervous functions, including consciousness, memory, and cognition. Therefore, it is important to understand the effects of general anesthetics on synaptic transmission via modulations of specific ion channels and relevant molecular targets, which can lead to the development of safer general anesthetics with selective actions. The present review will summarize the effects of various general anesthetics on synaptic transmissions and plasticity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yu Li
- Address correspondence to these authors at the Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China; E-mail: and Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, P.R. China; E-mail:
| | - Cheng Zhou
- Address correspondence to these authors at the Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China; E-mail: and Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, P.R. China; E-mail:
| |
Collapse
|
18
|
El-Refai NA, Shehata JH, Lotfy A, Elbadawy AM, Abdel Rahman RA, Gamaleldin TM, Dobal NM, Mohamed AA, Farrag TA, Shafik YM, Kamal AF. Role of preoperative pregabalin in reducing inhalational anesthetic requirements in abdominal hysterectomy: randomized controlled trial. Minerva Anestesiol 2020; 86:56-63. [DOI: 10.23736/s0375-9393.19.13734-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
Ou M, Zhao W, Liu J, Liang P, Huang H, Yu H, Zhu T, Zhou C. The General Anesthetic Isoflurane Bilaterally Modulates Neuronal Excitability. iScience 2019; 23:100760. [PMID: 31926429 PMCID: PMC6956953 DOI: 10.1016/j.isci.2019.100760] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/16/2019] [Accepted: 12/06/2019] [Indexed: 02/05/2023] Open
Abstract
Volatile anesthetics induce hyperactivity during induction while producing anesthesia at higher concentrations. They also bidirectionally modulate many neuronal functions. However, the neuronal mechanism is unclear. The effects of isoflurane on sodium channel currents were analyzed in acute mouse brain slices, including sodium leak (NALCN) currents and voltage-gated sodium channels (Nav) currents. Isoflurane at sub-anesthetic concentrations increased the spontaneous firing rate of CA3 pyramidal neurons, whereas anesthetic concentrations of isoflurane decreased the firing rate. Isoflurane at sub-anesthetic concentrations enhanced NALCN conductance but minimally inhibited Nav currents. Isoflurane at anesthetic concentrations depressed Nav currents and action potential amplitudes. Isoflurane at sub-anesthetic concentrations depolarized resting membrane potential (RMP) of neurons, whereas hyperpolarized the RMP at anesthetic concentrations. Isoflurane at low concentrations induced hyperactivity in vivo, which was diminished in NALCN knockdown mice. In conclusion, enhancement of NALCN by isoflurane contributes to its bidirectional modulation of neuronal excitability and the hyperactivity during induction. Volatile anesthetic isoflurane exerts bidirectional modulation of neuronal excitability Isoflurane enhances NALCN conductance at sub-anesthetic concentration NALCN knockdown diminishes behavioral hyperactivity during isoflurane induction
Collapse
Affiliation(s)
- Mengchan Ou
- Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China; Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Wenling Zhao
- Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China; Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Jin Liu
- Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China; Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Peng Liang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Han Huang
- Department of Anesthesiology, West China Second Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Hai Yu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Cheng Zhou
- Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China; Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China.
| |
Collapse
|
20
|
Murphy C, Krause B, Banks M. Selective effects of isoflurane on cortico-cortical feedback afferent responses in murine non-primary neocortex. Br J Anaesth 2019; 123:488-496. [PMID: 31383363 DOI: 10.1016/j.bja.2019.06.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/28/2019] [Accepted: 06/22/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND General anaesthetics affect loss of consciousness by disrupting information-passing and integration within thalamo-cortical (TC) networks. Feedback cortical connections that carry internally generated signals such as expectation and attention appear more sensitive to anaesthesia than feedforward signals. However, direct evidence for this effect in non-primary cortex is lacking. In addition, direct comparisons between TC core and matrix, and between cortico-cortical (CC) feedforward and feedback responses have not been reported. METHODS We investigated the disruption of synaptic responses by isoflurane of four distinct afferent pathways to non-primary neocortex. We independently activated TC core and matrix and reciprocal CC (feedforward and feedback) pathways using optogenetic techniques, and compared the relative sensitivity of synaptic responses to isoflurane. RESULTS Under control conditions, activation of axon terminals of all pathways evoked postsynaptic currents (recorded extracellularly) and postsynaptic potentials in pyramidal neurones. CC feedback responses were substantially more sensitive to isoflurane (0 to 0.53 mM) compared with TC core, TC matrix, or CC feedforward pathways. CONCLUSION Differential sensitivity of CC feedback synaptic responses to isoflurane in a clinically relevant range suggests a role for disruption of these afferents in the hypnotic effects of anaesthetic agents.
Collapse
Affiliation(s)
- Caitlin Murphy
- Physiology Graduate Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Bryan Krause
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Matthew Banks
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
21
|
Isoflurane Inhibits Dopaminergic Synaptic Vesicle Exocytosis Coupled to Ca V2.1 and Ca V2.2 in Rat Midbrain Neurons. eNeuro 2019; 6:eN-NWR-0278-18. [PMID: 30680310 PMCID: PMC6345200 DOI: 10.1523/eneuro.0278-18.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 11/21/2022] Open
Abstract
Volatile anesthetics affect neuronal signaling by poorly understood mechanisms. Activation of central dopaminergic pathways has been implicated in emergence from general anesthesia. The volatile anesthetic isoflurane differentially inhibits glutamatergic and GABAergic synaptic vesicle (SV) exocytosis by reducing presynaptic Ca2+ influx without affecting the Ca2+-exocytosis relationship, but its effects on dopaminergic exocytosis are unclear. We tested the hypothesis that isoflurane inhibits exocytosis in dopaminergic neurons. We used electrical stimulation or depolarization by elevated extracellular KCl to evoke exocytosis measured by quantitative live-cell fluorescence imaging in cultured rat ventral tegmental area neurons. Using trains of electrically evoked action potentials (APs), isoflurane inhibited exocytosis in dopaminergic neurons to a greater extent (30 ± 4% inhibition; p < 0.0001) than in non-dopaminergic neurons (15 ± 5% inhibition; p = 0.014). Isoflurane also inhibited exocytosis evoked by elevated KCl in dopaminergic neurons (35 ± 6% inhibition; p = 0.0007), but not in non-dopaminergic neurons (2 ± 4% inhibition). Pharmacological isolation of presynaptic Ca2+ channel subtypes showed that isoflurane inhibited KCl-evoked exocytosis mediated exclusively by either CaV2.1 (P/Q-type Ca2+ channels; 30 ± 5% inhibition; p = 0.0002) or by CaV2.2 (N-type Ca2+ channels; 35 ± 11% inhibition; p = 0.015). Additionally, isoflurane inhibited single AP-evoked Ca2+ influx by 41 ± 3% and single AP-evoked exocytosis by 34 ± 6%. Comparable reductions in exocytosis and Ca2+ influx were produced by lowering extracellular [Ca2+]. Thus, isoflurane inhibits exocytosis from dopaminergic neurons by a mechanism distinct from that in non-dopaminergic neurons involving reduced Ca2+ entry through CaV2.1 and/or CaV2.2.
Collapse
|
22
|
Hentschke H, Raz A, Krause BM, Murphy CA, Banks MI. Disruption of cortical network activity by the general anaesthetic isoflurane. Br J Anaesth 2019; 119:685-696. [PMID: 29121295 DOI: 10.1093/bja/aex199] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2017] [Indexed: 02/03/2023] Open
Abstract
Background Actions of general anaesthetics on activity in the cortico-thalamic network likely contribute to loss of consciousness and disconnection from the environment. Previously, we showed that the general anaesthetic isoflurane preferentially suppresses cortically evoked synaptic responses compared with thalamically evoked synaptic responses, but how this differential sensitivity translates into changes in network activity is unclear. Methods We investigated isoflurane disruption of spontaneous and stimulus-induced cortical network activity using multichannel recordings in murine auditory thalamo-cortical brain slices. Results Under control conditions, afferent stimulation elicited short latency, presumably monosynaptically driven, spiking responses, as well as long latency network bursts that propagated horizontally through the cortex. Isoflurane (0.05-0.6 mM) suppressed spiking activity overall, but had a far greater effect on network bursts than on early spiking responses. At isoflurane concentrations >0.3 mM, network bursts were almost entirely blocked, even with increased stimulation intensity and in response to paired (thalamo-cortical + cortical layer 1) stimulation, while early spiking responses were <50% blocked. Isoflurane increased the threshold for eliciting bursts, decreased their propagation speed and prevented layer 1 afferents from facilitating burst induction by thalamo-cortical afferents. Conclusions Disruption of horizontal activity spread and of layer 1 facilitation of thalamo-cortical responses likely contribute to the mechanism by which suppression of cortical feedback connections disrupts sensory awareness under anaesthesia.
Collapse
Affiliation(s)
- H Hentschke
- Department of Anesthesiology, Experimental Anesthesiology Section, University Hospital of Tübingen, Tübingen, Germany
| | - A Raz
- Department of Anesthesiology, University of Wisconsin, Madison, WI, USA.,Department of Anesthesiology, Rambam Health Care Campus, Haifa, Israel
| | - B M Krause
- Department of Anesthesiology, University of Wisconsin, Madison, WI, USA
| | - C A Murphy
- Department of Anesthesiology, University of Wisconsin, Madison, WI, USA.,Physiology Graduate Training Program, University of Wisconsin, Madison, WI, USA
| | - M I Banks
- Department of Anesthesiology, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
23
|
Banks MI, Moran NS, Krause BM, Grady SM, Uhlrich DJ, Manning KA. Altered stimulus representation in rat auditory cortex is not causal for loss of consciousness under general anaesthesia. Br J Anaesth 2018; 121:605-615. [PMID: 30115259 DOI: 10.1016/j.bja.2018.05.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/13/2018] [Accepted: 05/21/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Current concepts suggest that impaired representation of information in cortical networks contributes to loss of consciousness under anaesthesia. We tested this idea in rat auditory cortex using information theory analysis of multiunit responses recorded under three anaesthetic agents with different molecular targets: isoflurane, propofol, and dexmedetomidine. We reasoned that if changes in the representation of sensory stimuli are causal for loss of consciousness, they should occur regardless of the specific anaesthetic agent. METHODS Spiking responses were recorded with chronically implanted microwire arrays in response to acoustic stimuli incorporating varied temporal and spectral dynamics. Experiments consisted of four drug conditions: awake (pre-drug), sedation (i.e. intact righting reflex), loss of consciousness (a dose just sufficient to cause loss of righting reflex), and recovery. Measures of firing rate, spike timing, and mutual information were analysed as a function of drug condition. RESULTS All three drugs decreased spontaneous and evoked spiking activity and modulated spike timing. However, changes in mutual information were inconsistent with altered stimulus representation being causal for loss of consciousness. First, direction of change in mutual information was agent-specific, increasing under dexmedetomidine and decreasing under isoflurane and propofol. Second, mutual information did not decrease at the transition between sedation and LOC for any agent. Changes in mutual information under anaesthesia correlated strongly with changes in precision and reliability of spike timing, consistent with the importance of temporal stimulus features in driving auditory cortical activity. CONCLUSIONS The primary sensory cortex is not the locus for changes in representation of information causal for loss of consciousness under anaesthesia.
Collapse
Affiliation(s)
- M I Banks
- Department of Anesthesiology, University of Wisconsin, Madison, WI, USA.
| | - N S Moran
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - B M Krause
- Department of Anesthesiology, University of Wisconsin, Madison, WI, USA
| | - S M Grady
- Department of Anesthesiology, University of Wisconsin, Madison, WI, USA
| | - D J Uhlrich
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| | - K A Manning
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
24
|
Whitmore C, Cook AR, Mann T, Price ME, Emery E, Roughley N, Flint D, Stubbs S, Armstrong SJ, Rice H, Tattersall JEH. The efficacy of HI-6 DMS in a sustained infusion against percutaneous VX poisoning in the guinea-pig. Toxicol Lett 2017; 293:207-215. [PMID: 29129798 DOI: 10.1016/j.toxlet.2017.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/02/2017] [Accepted: 11/06/2017] [Indexed: 11/29/2022]
Abstract
Post-exposure nerve agent treatment usually includes administration of an oxime, which acts to restore function of the enzyme acetylcholinesterase (AChE). For immediate treatment of military personnel, this is usually administered with an autoinjector device, or devices containing the oxime such as pralidoxime, atropine and diazepam. In addition to the autoinjector, it is likely that personnel exposed to nerve agents, particularly by the percutaneous route, will require further treatment at medical facilities. As such, there is a need to understand the relationship between dose rate, plasma concentration, reactivation of AChE activity and efficacy, to provide supporting evidence for oxime infusions in nerve agent poisoning. Here, it has been demonstrated that intravenous infusion of HI-6, in combination with atropine, is efficacious against a percutaneous VX challenge in the conscious male Dunkin-Hartley guinea-pig. Inclusion of HI-6, in addition to atropine in the treatment, improved survival when compared to atropine alone. Additionally, erythrocyte AChE activity following poisoning was found to be dose dependent, with an increased dose rate of HI-6 (0.48mg/kg/min) resulting in increased AChE activity. As far as we are aware, this is the first study to correlate the pharmacokinetic profile of HI-6 with both its pharmacodynamic action of reactivating nerve agent inhibited AChE and with its efficacy against a persistent nerve agent exposure challenge in the same conscious animal.
Collapse
Affiliation(s)
- C Whitmore
- CBR (Chemical, Biological, Radiological), Dstl Porton Down, Salisbury, Wiltshire, SP4 0JQ, United Kingdom.
| | - A R Cook
- CBR (Chemical, Biological, Radiological), Dstl Porton Down, Salisbury, Wiltshire, SP4 0JQ, United Kingdom
| | - T Mann
- CBR (Chemical, Biological, Radiological), Dstl Porton Down, Salisbury, Wiltshire, SP4 0JQ, United Kingdom
| | - M E Price
- CBR (Chemical, Biological, Radiological), Dstl Porton Down, Salisbury, Wiltshire, SP4 0JQ, United Kingdom
| | - E Emery
- CBR (Chemical, Biological, Radiological), Dstl Porton Down, Salisbury, Wiltshire, SP4 0JQ, United Kingdom
| | - N Roughley
- CBR (Chemical, Biological, Radiological), Dstl Porton Down, Salisbury, Wiltshire, SP4 0JQ, United Kingdom
| | - D Flint
- CBR (Chemical, Biological, Radiological), Dstl Porton Down, Salisbury, Wiltshire, SP4 0JQ, United Kingdom
| | - S Stubbs
- CBR (Chemical, Biological, Radiological), Dstl Porton Down, Salisbury, Wiltshire, SP4 0JQ, United Kingdom
| | - S J Armstrong
- CBR (Chemical, Biological, Radiological), Dstl Porton Down, Salisbury, Wiltshire, SP4 0JQ, United Kingdom
| | - H Rice
- CBR (Chemical, Biological, Radiological), Dstl Porton Down, Salisbury, Wiltshire, SP4 0JQ, United Kingdom
| | - J E H Tattersall
- CBR (Chemical, Biological, Radiological), Dstl Porton Down, Salisbury, Wiltshire, SP4 0JQ, United Kingdom
| |
Collapse
|
25
|
Fong R, Khokhar S, Chowdhury AN, Xie KG, Wong JHY, Fox AP, Xie Z. Caffeine accelerates recovery from general anesthesia via multiple pathways. J Neurophysiol 2017; 118:1591-1597. [PMID: 28659466 PMCID: PMC5596131 DOI: 10.1152/jn.00393.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/22/2017] [Accepted: 06/22/2017] [Indexed: 12/19/2022] Open
Abstract
Various studies have explored different ways to speed emergence from anesthesia. Previously, we have shown that three drugs that elevate intracellular cAMP (forskolin, theophylline, and caffeine) accelerate emergence from anesthesia in rats. However, our earlier studies left two main questions unanswered. First, were cAMP-elevating drugs effective at all anesthetic concentrations? Second, given that caffeine was the most effective of the drugs tested, why was caffeine more effective than forskolin since both drugs elevate cAMP? In our current study, emergence time from anesthesia was measured in adult rats exposed to 3% isoflurane for 60 min. Caffeine dramatically accelerated emergence from anesthesia, even at the high level of anesthetic employed. Caffeine has multiple actions including blockade of adenosine receptors. We show that the selective A2a adenosine receptor antagonist preladenant or the intracellular cAMP ([cAMP]i)-elevating drug forskolin, accelerated recovery from anesthesia. When preladenant and forskolin were tested together, the effect on anesthesia recovery time was additive indicating that these drugs operate via different pathways. Furthermore, the combination of preladenant and forskolin was about as effective as caffeine suggesting that both A2A receptor blockade and [cAMP]i elevation play a role in caffeine's ability to accelerate emergence from anesthesia. Because anesthesia in rodents is thought to be similar to that in humans, these results suggest that caffeine might allow for rapid and uniform emergence from general anesthesia in humans at all anesthetic concentrations and that both the elevation of [cAMP]i and adenosine receptor blockade play a role in this response.NEW & NOTEWORTHY Currently, there is no method to accelerate emergence from anesthesia. Patients "wake" when they clear the anesthetic from their systems. Previously, we have shown that caffeine can accelerate emergence from anesthesia. In this study, we show that caffeine is effective even at high levels of anesthetic. We also show that caffeine operates by both elevating intracellular cAMP levels and by blocking adenosine receptors. This complicated pharmacology makes caffeine especially effective in accelerating emergence from anesthesia.
Collapse
Affiliation(s)
- Robert Fong
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois
| | - Suhail Khokhar
- College of Medicine, University of Illinois, School of Life Sciences, Chicago, Illinois
| | - Atif N Chowdhury
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kelvin G Xie
- School of Engineering and Applied Science, Washington University, St Louis, Missouri
| | | | - Aaron P Fox
- Department of Neurobiology, Pharmacology and Physiology, The University of Chicago, Chicago, Illinois
| | - Zheng Xie
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois;
| |
Collapse
|
26
|
Yin YQ, Wang LF, Chen C, Gao T, Zhao ZF, Li CH. In vivo field recordings effectively monitor the mouse cortex and hippocampus under isoflurane anesthesia. Neural Regen Res 2016; 11:1951-1955. [PMID: 28197191 PMCID: PMC5270433 DOI: 10.4103/1673-5374.197136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2016] [Indexed: 02/05/2023] Open
Abstract
Isoflurane is a widely used inhaled anesthetic in the clinical setting. However, the mechanism underlying its effect on consciousness is under discussion. Therefore, we investigated the effect of isoflurane on the hippocampus and cortex using an in vivo field recording approach. Our results showed that 1.3%, 0.8%, and 0.4% isoflurane exerted an inhibitory influence on the mouse hippocampus and cortex. Further, high frequency bands in the cortex and hippocampus showed greater suppression with increasing isoflurane concentration. Our findings suggest that in vivo field recordings can monitor the effect of isoflurane anesthesia on the mouse cortex and hippocampus.
Collapse
Affiliation(s)
- Yi-qing Yin
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Li-fang Wang
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Chao Chen
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Teng Gao
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Zi-fang Zhao
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Cheng-hui Li
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
27
|
Sensitivity to isoflurane anesthesia increases in autism spectrum disorder Shank3 +/∆c mutant mouse model. Neurotoxicol Teratol 2016; 60:69-74. [PMID: 27856360 DOI: 10.1016/j.ntt.2016.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 10/14/2016] [Accepted: 11/09/2016] [Indexed: 11/20/2022]
Abstract
Autism is a heterogeneous developmental disorder characterized by impaired social interaction, impaired communication skills, and restricted and repetitive behavior. The abnormal behaviors of these patients can make their anesthetic and perioperative management difficult. Evidence in the literature suggests that some patients with autism or specific autism spectrum disorders (ASD) exhibit altered responses to pain and to anesthesia or sedation. A genetic mouse model of one particular ASD, Phelan McDermid Syndrome, has been developed that has a Shank3 haplotype truncation (Shank3+/Δc). These mice exhibit important characteristics of autism that mimic human autistic behavior. Our study demonstrates that a Shank3+/ΔC mutation in mice is associated with a reduction in both the MAC and RREC50 of isoflurane and down regulation of NR1 in vestibular nuclei and PSD95 in spinal cord. Decreased expression of NR1 and PSD95 in the central nervous system of Shank3+/ΔC mice could help reduce the MAC and RREC50 of isoflurane, which would warrant confirmation in a clinical study. If Shank3 mutations are found to affect anesthetic sensitivity in patients with ASD, better communication and stricter monitoring of anesthetic depth may be necessary.
Collapse
|
28
|
Covarrubias M, Barber AF, Carnevale V, Treptow W, Eckenhoff RG. Mechanistic Insights into the Modulation of Voltage-Gated Ion Channels by Inhalational Anesthetics. Biophys J 2016; 109:2003-11. [PMID: 26588560 DOI: 10.1016/j.bpj.2015.09.032] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/31/2015] [Accepted: 09/30/2015] [Indexed: 12/20/2022] Open
Abstract
General anesthesia is a relatively safe medical procedure, which for nearly 170 years has allowed life saving surgical interventions in animals and people. However, the molecular mechanism of general anesthesia continues to be a matter of importance and debate. A favored hypothesis proposes that general anesthesia results from direct multisite interactions with multiple and diverse ion channels in the brain. Neurotransmitter-gated ion channels and two-pore K+ channels are key players in the mechanism of anesthesia; however, new studies have also implicated voltage-gated ion channels. Recent biophysical and structural studies of Na+ and K+ channels strongly suggest that halogenated inhalational general anesthetics interact with gates and pore regions of these ion channels to modulate function. Here, we review these studies and provide a perspective to stimulate further advances.
Collapse
Affiliation(s)
- Manuel Covarrubias
- Department of Neuroscience and Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania.
| | - Annika F Barber
- Department of Neuroscience, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Werner Treptow
- Laboratorio de Biologia Teorica e Computacional, Universidade de Brasilia, Brazil
| | - Roderic G Eckenhoff
- Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Hara M, Zhou ZY, Hemmings HC. α2-Adrenergic Receptor and Isoflurane Modulation of Presynaptic Ca2+ Influx and Exocytosis in Hippocampal Neurons. Anesthesiology 2016; 125:535-46. [PMID: 27337223 PMCID: PMC4988866 DOI: 10.1097/aln.0000000000001213] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Evidence indicates that the anesthetic-sparing effects of α2-adrenergic receptor (AR) agonists involve α2A-AR heteroreceptors on nonadrenergic neurons. Since volatile anesthetics inhibit neurotransmitter release by reducing synaptic vesicle (SV) exocytosis, the authors hypothesized that α2-AR agonists inhibit nonadrenergic SV exocytosis and thereby potentiate presynaptic inhibition of exocytosis by isoflurane. METHODS Quantitative imaging of fluorescent biosensors of action potential-evoked SV exocytosis (synaptophysin-pHluorin) and Ca influx (GCaMP6) were used to characterize presynaptic actions of the clinically used α2-AR agonists dexmedetomidine and clonidine, and their interaction with isoflurane, in cultured rat hippocampal neurons. RESULTS Dexmedetomidine (0.1 μM, n = 10) or clonidine (0.5 μM, n = 8) inhibited action potential-evoked exocytosis (54 ± 5% and 59 ± 8% of control, respectively; P < 0.001). Effects on exocytosis were blocked by the subtype-nonselective α2-AR antagonist atipamezole or the α2A-AR-selective antagonist BRL 44408 but not by the α2C-AR-selective antagonist JP 1302. Dexmedetomidine inhibited exocytosis and presynaptic Ca influx without affecting Ca coupling to exocytosis, consistent with an effect upstream of Ca-exocytosis coupling. Exocytosis coupled to both N-type and P/Q-type Ca channels was inhibited by dexmedetomidine or clonidine. Dexmedetomidine potentiated inhibition of exocytosis by 0.7 mM isoflurane (to 42 ± 5%, compared to 63 ± 8% for isoflurane alone; P < 0.05). CONCLUSIONS Hippocampal SV exocytosis is inhibited by α2A-AR activation in proportion to reduced Ca entry. These effects are additive with those of isoflurane, consistent with a role for α2A-AR presynaptic heteroreceptor inhibition of nonadrenergic synaptic transmission in the anesthetic-sparing effects of α2A-AR agonists.
Collapse
Affiliation(s)
- Masato Hara
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065, USA
- Department of Anesthesiology, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Zhen-Yu Zhou
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Hugh C. Hemmings
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065, USA
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
30
|
Pancuronium enhances isoflurane anesthesia in rats via inhibition of cerebral nicotinic acetylcholine receptors. J Anesth 2016; 30:671-6. [PMID: 27146658 DOI: 10.1007/s00540-016-2178-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/21/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE This study was conducted to elucidate the mechanism of enhancement of volatile anesthetics by neuromuscular blocking agents in rats and to consider the relevance of this enhancement to clinical anesthesia. METHODS Male Sprague-Dawley rats were used. After confirming a movement in response to tail clamping under 1.1 % isoflurane anesthesia, response was determined when the tail clamp was applied at several points after microinjection of pancuronium into the lateral ventricle. Arousal responses to microinjection of nicotine into the lateral ventricle were assessed with or without pretreatment with intraventricular pancuronium. The intravenous 50 % effective dose (ED50) and 95 % effective dose (ED95) for neuromuscular blockade with pancuronium administered in a cumulative fashion at 1.1 % isoflurane were calculated. RESULTS Intraventricular pancuronium dose-dependently reduced the response to tail clamping, and the dose required to show immobilization of 50 % of rats (intraventricular ED50) was 1.62 µg/kg. Pretreatment with pancuronium at 6 µg/kg significantly reduced the effect of awakening by nicotine under isoflurane anesthesia (P = 0.044). The intravenous ED50 and ED95 for neuromuscular blockade were 63 µg/kg (90 % confidence interval [CI] 52-75 µg/kg) and 133 µg/kg (90 % CI 109-158 µg/kg), respectively. The ratio of intraventricular ED50 to intravenous ED50 was 0.026. CONCLUSION Pancuronium microinjection into the lateral ventricle dose-dependently enhances the depth of isoflurane anesthesia, which might be caused by inhibition of neuronal nicotinic acetylcholine receptor transmission in the cerebrum. Intravenous injection of pancuronium at high doses might increase the cerebrospinal concentration to a level at which an effect can be observed.
Collapse
|
31
|
Nestor CC, Qiu J, Padilla SL, Zhang C, Bosch MA, Fan W, Aicher SA, Palmiter RD, Rønnekleiv OK, Kelly MJ. Optogenetic Stimulation of Arcuate Nucleus Kiss1 Neurons Reveals a Steroid-Dependent Glutamatergic Input to POMC and AgRP Neurons in Male Mice. Mol Endocrinol 2016; 30:630-44. [PMID: 27093227 DOI: 10.1210/me.2016-1026] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Kisspeptin (Kiss1) neurons are essential for reproduction, but their role in the control of energy balance and other homeostatic functions remains unclear. Proopiomelanocortin (POMC) and agouti-related peptide (AgRP) neurons, located in the arcuate nucleus (ARC) of the hypothalamus, integrate numerous excitatory and inhibitory inputs to ultimately regulate energy homeostasis. Given that POMC and AgRP neurons are contacted by Kiss1 neurons in the ARC (Kiss1(ARC)) and they express androgen receptors, Kiss1(ARC) neurons may mediate the orexigenic action of testosterone via POMC and/or AgRP neurons. Quantitative PCR analysis of pooled Kiss1(ARC) neurons revealed that mRNA levels for Kiss1 and vesicular glutamate transporter 2 were higher in castrated male mice compared with gonad-intact males. Single-cell RT-PCR analysis of yellow fluorescent protein (YFP) ARC neurons harvested from males injected with AAV1-EF1α-DIO-ChR2:YFP revealed that 100% and 88% expressed mRNAs for Kiss1 and vesicular glutamate transporter 2, respectively. Whole-cell, voltage-clamp recordings from nonfluorescent postsynaptic ARC neurons showed that low frequency photo-stimulation (0.5 Hz) of Kiss1-ChR2:YFP neurons elicited a fast glutamatergic inward current in POMC and AgRP neurons. Paired-pulse, photo-stimulation revealed paired-pulse depression, which is indicative of greater glutamate release, in the castrated male mice compared with gonad-intact male mice. Group I and group II metabotropic glutamate receptor agonists depolarized and hyperpolarized POMC and AgRP neurons, respectively, which was mimicked by high frequency photo-stimulation (20 Hz) of Kiss1(ARC) neurons. Therefore, POMC and AgRP neurons receive direct steroid- and frequency-dependent glutamatergic synaptic input from Kiss1(ARC) neurons in male mice, which may be a critical pathway for Kiss1 neurons to help coordinate energy homeostasis and reproduction.
Collapse
Affiliation(s)
- Casey C Nestor
- Department of Physiology and Pharmacology (C.CN., J.Q., C.Z., M.A.B., S.A.A., O.K.R., M.J.K.) and Anesthesiology and Perioperative Medicine and Knight Cardiovascular Institute (W.F.), Oregon Health & Science University, Portland, Oregon 97239; Division of Neuroscience (O.K.R., M.J.K.), Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006; and Howard Hughes Medical Institute (S.L.P., R.D.P.), University of Washington, Seattle, Washington 98195
| | - Jian Qiu
- Department of Physiology and Pharmacology (C.CN., J.Q., C.Z., M.A.B., S.A.A., O.K.R., M.J.K.) and Anesthesiology and Perioperative Medicine and Knight Cardiovascular Institute (W.F.), Oregon Health & Science University, Portland, Oregon 97239; Division of Neuroscience (O.K.R., M.J.K.), Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006; and Howard Hughes Medical Institute (S.L.P., R.D.P.), University of Washington, Seattle, Washington 98195
| | - Stephanie L Padilla
- Department of Physiology and Pharmacology (C.CN., J.Q., C.Z., M.A.B., S.A.A., O.K.R., M.J.K.) and Anesthesiology and Perioperative Medicine and Knight Cardiovascular Institute (W.F.), Oregon Health & Science University, Portland, Oregon 97239; Division of Neuroscience (O.K.R., M.J.K.), Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006; and Howard Hughes Medical Institute (S.L.P., R.D.P.), University of Washington, Seattle, Washington 98195
| | - Chunguang Zhang
- Department of Physiology and Pharmacology (C.CN., J.Q., C.Z., M.A.B., S.A.A., O.K.R., M.J.K.) and Anesthesiology and Perioperative Medicine and Knight Cardiovascular Institute (W.F.), Oregon Health & Science University, Portland, Oregon 97239; Division of Neuroscience (O.K.R., M.J.K.), Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006; and Howard Hughes Medical Institute (S.L.P., R.D.P.), University of Washington, Seattle, Washington 98195
| | - Martha A Bosch
- Department of Physiology and Pharmacology (C.CN., J.Q., C.Z., M.A.B., S.A.A., O.K.R., M.J.K.) and Anesthesiology and Perioperative Medicine and Knight Cardiovascular Institute (W.F.), Oregon Health & Science University, Portland, Oregon 97239; Division of Neuroscience (O.K.R., M.J.K.), Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006; and Howard Hughes Medical Institute (S.L.P., R.D.P.), University of Washington, Seattle, Washington 98195
| | - Wei Fan
- Department of Physiology and Pharmacology (C.CN., J.Q., C.Z., M.A.B., S.A.A., O.K.R., M.J.K.) and Anesthesiology and Perioperative Medicine and Knight Cardiovascular Institute (W.F.), Oregon Health & Science University, Portland, Oregon 97239; Division of Neuroscience (O.K.R., M.J.K.), Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006; and Howard Hughes Medical Institute (S.L.P., R.D.P.), University of Washington, Seattle, Washington 98195
| | - Sue A Aicher
- Department of Physiology and Pharmacology (C.CN., J.Q., C.Z., M.A.B., S.A.A., O.K.R., M.J.K.) and Anesthesiology and Perioperative Medicine and Knight Cardiovascular Institute (W.F.), Oregon Health & Science University, Portland, Oregon 97239; Division of Neuroscience (O.K.R., M.J.K.), Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006; and Howard Hughes Medical Institute (S.L.P., R.D.P.), University of Washington, Seattle, Washington 98195
| | - Richard D Palmiter
- Department of Physiology and Pharmacology (C.CN., J.Q., C.Z., M.A.B., S.A.A., O.K.R., M.J.K.) and Anesthesiology and Perioperative Medicine and Knight Cardiovascular Institute (W.F.), Oregon Health & Science University, Portland, Oregon 97239; Division of Neuroscience (O.K.R., M.J.K.), Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006; and Howard Hughes Medical Institute (S.L.P., R.D.P.), University of Washington, Seattle, Washington 98195
| | - Oline K Rønnekleiv
- Department of Physiology and Pharmacology (C.CN., J.Q., C.Z., M.A.B., S.A.A., O.K.R., M.J.K.) and Anesthesiology and Perioperative Medicine and Knight Cardiovascular Institute (W.F.), Oregon Health & Science University, Portland, Oregon 97239; Division of Neuroscience (O.K.R., M.J.K.), Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006; and Howard Hughes Medical Institute (S.L.P., R.D.P.), University of Washington, Seattle, Washington 98195
| | - Martin J Kelly
- Department of Physiology and Pharmacology (C.CN., J.Q., C.Z., M.A.B., S.A.A., O.K.R., M.J.K.) and Anesthesiology and Perioperative Medicine and Knight Cardiovascular Institute (W.F.), Oregon Health & Science University, Portland, Oregon 97239; Division of Neuroscience (O.K.R., M.J.K.), Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon 97006; and Howard Hughes Medical Institute (S.L.P., R.D.P.), University of Washington, Seattle, Washington 98195
| |
Collapse
|
32
|
Isoflurane inhibits synaptic vesicle exocytosis through reduced Ca2+ influx, not Ca2+-exocytosis coupling. Proc Natl Acad Sci U S A 2015; 112:11959-64. [PMID: 26351670 DOI: 10.1073/pnas.1500525112] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Identifying presynaptic mechanisms of general anesthetics is critical to understanding their effects on synaptic transmission. We show that the volatile anesthetic isoflurane inhibits synaptic vesicle (SV) exocytosis at nerve terminals in dissociated rat hippocampal neurons through inhibition of presynaptic Ca(2+) influx without significantly altering the Ca(2+) sensitivity of SV exocytosis. A clinically relevant concentration of isoflurane (0.7 mM) inhibited changes in [Ca(2+)]i driven by single action potentials (APs) by 25 ± 3%, which in turn led to 62 ± 3% inhibition of single AP-triggered exocytosis at 4 mM extracellular Ca(2+) ([Ca(2+)]e). Lowering external Ca(2+) to match the isoflurane-induced reduction in Ca(2+) entry led to an equivalent reduction in exocytosis. These data thus indicate that anesthetic inhibition of neurotransmitter release from small SVs occurs primarily through reduced axon terminal Ca(2+) entry without significant direct effects on Ca(2+)-exocytosis coupling or on the SV fusion machinery. Isoflurane inhibition of exocytosis and Ca(2+) influx was greater in glutamatergic compared with GABAergic nerve terminals, consistent with selective inhibition of excitatory synaptic transmission. Such alteration in the balance of excitatory to inhibitory transmission could mediate reduced neuronal interactions and network-selective effects observed in the anesthetized central nervous system.
Collapse
|
33
|
Sahlholm K, Sijbesma JWA, Maas B, Kwizera C, Marcellino D, Ramakrishnan NK, Dierckx RAJO, Elsinga PH, van Waarde A. Pridopidine selectively occupies sigma-1 rather than dopamine D2 receptors at behaviorally active doses. Psychopharmacology (Berl) 2015; 232:3443-53. [PMID: 26159455 PMCID: PMC4537502 DOI: 10.1007/s00213-015-3997-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/15/2015] [Indexed: 02/07/2023]
Abstract
RATIONALE Dopamine stabilizers have stimulatory actions under low dopamine tone and inhibitory actions under high dopamine tone without eliciting catalepsy. These compounds are dopamine D2 receptor (D2R) antagonists or weak partial agonists and may have pro-mnemonic and neuroprotective effects. The mechanism underlying their stimulatory and neuroprotective actions is unknown but could involve sigma-1R binding. OBJECTIVES The present study examined sigma-1R and D2R occupancy by the dopamine stabilizer pridopidine (ACR16) at behaviorally relevant doses in living rats. METHODS Rats were administered 3 or 15 mg/kg pridopidine, or saline, before injection of the radiotracer (11)C-SA4503 (sigma-1R) or (11)C-raclopride (D2R). Some animals received 60 mg/kg pridopidine and were only scanned with (11)C-raclopride. Cerebral (11)C-SA4503 binding was quantified using metabolite-corrected plasma input data and distribution volume (V T) calculated by Logan graphical analysis. (11)C-raclopride binding was quantified using striatum-to-cerebellum ratios and binding potentials calculated with a simplified reference tissue model. RESULTS Cunningham-Lassen plots indicated sigma-1R occupancies of 57 ± 2 and 85 ± 2% after pretreatment of animals with 3 and 15 mg/kg pridopidine. A significant (44-66%) reduction of (11)C-raclopride binding was only observed at 60 mg/kg pridopidine. CONCLUSIONS At doses shown to elicit neurochemical and behavioral effects, pridopidine occupied a large fraction of sigma-1Rs and a negligible fraction of D2Rs. Significant D2R occupancy was only observed at a dose 20-fold higher than was required for sigma-1R occupancy. The characteristics of dopamine stabilizers may result from the combination of high sigma-1R and low D2R affinity.
Collapse
Affiliation(s)
- Kristoffer Sahlholm
- />Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- />Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jurgen W. A. Sijbesma
- />Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bram Maas
- />Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Chantal Kwizera
- />Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Daniel Marcellino
- />Department of Physiology and Institute of Biomedical Technology (ITB), Center for Biomedical Research of the Canary Islands (CIBICAN), University of La Laguna School of Medicine, Tenerife, Spain
| | - Nisha K. Ramakrishnan
- />Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rudi A. J. O. Dierckx
- />Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Philip H. Elsinga
- />Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Aren van Waarde
- />Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
34
|
Castro Fonseca MD, Da Silva JH, Ferraz VP, Gomez RS, Guatimosim C. Comparative presynaptic effects of the volatile anesthetics sevoflurane and isoflurane at the mouse neuromuscular junction. Muscle Nerve 2015; 52:876-84. [DOI: 10.1002/mus.24589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Matheus De Castro Fonseca
- Departamento de Morfologia, Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais; Av. Antônio Carlos, 6627 Belo Horizonte MG 31270-901 Brasil
| | - Janice Henriques Da Silva
- Departamento de Morfologia, Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais; Av. Antônio Carlos, 6627 Belo Horizonte MG 31270-901 Brasil
| | - Vany Perpetua Ferraz
- Departamento de Química, Instituto de Ciências Exatas; Universidade Federal de Minas Gerais; MG Brasil
| | - Renato Santiago Gomez
- Departamento de Cirurgia, Faculdade de Medicina; Universidade Federal de Minas Gerais; Belo Horizonte MG Brasil
| | - Cristina Guatimosim
- Departamento de Morfologia, Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais; Av. Antônio Carlos, 6627 Belo Horizonte MG 31270-901 Brasil
| |
Collapse
|
35
|
Carcenac C, Favier M, Vachez Y, Lacombe E, Carnicella S, Savasta M, Boulet S. Subthalamic deep brain stimulation differently alters striatal dopaminergic receptor levels in rats. Mov Disord 2015; 30:1739-49. [DOI: 10.1002/mds.26146] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 12/02/2014] [Accepted: 12/05/2014] [Indexed: 01/11/2023] Open
Affiliation(s)
- Carole Carcenac
- Institut National de la Santé et de la Recherche Médicale, Grenoble Institut des Neurosciences; Dynamique et Physiopathologie des Ganglions de la Base Grenoble France
- Grenoble University; Grenoble France
| | - Mathieu Favier
- Institut National de la Santé et de la Recherche Médicale, Grenoble Institut des Neurosciences; Dynamique et Physiopathologie des Ganglions de la Base Grenoble France
- Grenoble University; Grenoble France
| | - Yvan Vachez
- Institut National de la Santé et de la Recherche Médicale, Grenoble Institut des Neurosciences; Dynamique et Physiopathologie des Ganglions de la Base Grenoble France
- Grenoble University; Grenoble France
| | - Emilie Lacombe
- Institut National de la Santé et de la Recherche Médicale, Grenoble Institut des Neurosciences; Dynamique et Physiopathologie des Ganglions de la Base Grenoble France
- Grenoble University; Grenoble France
| | - Sébastien Carnicella
- Institut National de la Santé et de la Recherche Médicale, Grenoble Institut des Neurosciences; Dynamique et Physiopathologie des Ganglions de la Base Grenoble France
- Grenoble University; Grenoble France
| | - Marc Savasta
- Institut National de la Santé et de la Recherche Médicale, Grenoble Institut des Neurosciences; Dynamique et Physiopathologie des Ganglions de la Base Grenoble France
- Grenoble University; Grenoble France
- Centre Hospitalier Universitaire de Grenoble; Grenoble France
| | - Sabrina Boulet
- Institut National de la Santé et de la Recherche Médicale, Grenoble Institut des Neurosciences; Dynamique et Physiopathologie des Ganglions de la Base Grenoble France
- Grenoble University; Grenoble France
| |
Collapse
|
36
|
Kufahl PR, Peartree NA, Heintzelman KL, Chung M, Neisewander JL. Region-specific effects of isoflurane anesthesia on Fos immunoreactivity in response to intravenous cocaine challenge in rats with a history of repeated cocaine administration. Brain Res 2015; 1594:256-66. [PMID: 25451087 PMCID: PMC4805112 DOI: 10.1016/j.brainres.2014.10.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 10/14/2014] [Accepted: 10/15/2014] [Indexed: 12/29/2022]
Abstract
We have previously shown that acute intravenous (i.v.) administration of cocaine increases Fos immunoreactivity in rats under isoflurane anesthesia. Given that Fos expression is a marker of neural activation, the results suggested that isoflurane is appropriate for imaging cocaine effects under anesthesia. However, most imaging research in this area utilizes subjects with a history of repeated cocaine exposure and this drug history may interact with anesthetic use differently from acute cocaine exposure. Thus, this study further examined Fos expression under isoflurane in rats with a history of repeated i.v. cocaine administration. Rats received daily injections of either saline or cocaine (2mg/kg, i.v.) across 7 consecutive days, followed by 5 days of no drug exposure. On the test day, rats were either nonanesthetized or anesthetized under isoflurane and were given an acute challenge of cocaine (2mg/kg, i.v.). Additional saline-exposed controls received a saline challenge. Ninety min after the drug challenge, the rats were perfused under isoflurane anesthesia and their brains were processed for Fos protein immunohistochemistry. We found that challenge injections of cocaine following a regimen of repeated cocaine exposure resulted in Fos expression in the prefrontal cortex and striatum roughly equivalent to that found in rats who had received the cocaine challenge after a history of vehicle injections. Additionally, isoflurane anesthesia resulted in a heterogeneous attenuation of cocaine-induced Fos expression, with the most robust effect in the orbital cortex but no effect in the nucleus accumbens core (NAcC). These results indicate that cocaine-induced Fos is preserved in the NAcC under isoflurane, suggesting that isoflurane can be used in imaging studies involving cocaine effects in this region.
Collapse
Affiliation(s)
- Peter R Kufahl
- Department of Psychology, Arizona State University, Tempe, AZ 85287-1104, United States.
| | - Natalie A Peartree
- Department of Psychology, Arizona State University, Tempe, AZ 85287-1104, United States
| | - Krista L Heintzelman
- Department of Psychology, Arizona State University, Tempe, AZ 85287-1104, United States
| | - Maggie Chung
- Department of Psychology, Arizona State University, Tempe, AZ 85287-1104, United States
| | - Janet L Neisewander
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, United States
| |
Collapse
|
37
|
Wiese AJ, Brosnan RJ, Barter LS. Effects of acetylcholinesterase inhibition on quality of recovery from isoflurane-induced anesthesia in horses. Am J Vet Res 2014; 75:223-30. [PMID: 24564307 DOI: 10.2460/ajvr.75.3.223] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To compare effects of 2 acetylcholinesterase inhibitors on recovery quality of horses anesthetized with isoflurane. ANIMALS 6 horses in phase 1, 7 horses in phase 2A, and 14 horses in phase 2B. PROCEDURES The study comprised 3 phases (2 randomized, blinded crossover phases in horses undergoing orthopedic procedures and 1 prospective dose-determining phase). In phase 1, horses were anesthetized with isoflurane and received neostigmine or saline (0.9% NaCl) solution prior to anesthetic recovery. Phase 2A was a physostigmine dose-determining phase. In phase 2B, horses were anesthetized with isoflurane and received neostigmine or physostigmine prior to recovery. Objective recovery events were recorded and subjective visual analogue scale scores of recovery quality were assigned from video recordings. RESULTS Recovery measures in phase 1 were not different between horses receiving neostigmine or saline solution. In phase 2A, 0.04 mg of physostigmine/kg was the highest cumulative dose that did not cause clinically relevant adverse behavioral or gastrointestinal effects. Horses receiving physostigmine had higher mean ± SD visual analogue scale recovery scores (70.8 ± 13.3 mm) than did horses receiving neostigmine (62.4 ± 12.8 mm) in phase 2B, with fewer attempts until sternal and standing recovery. Incidence of colic behavior did not differ among groups. CONCLUSIONS AND CLINICAL RELEVANCE Inhibition with physostigmine improved anesthetic recovery quality in horses anesthetized with isoflurane, compared with recovery quality for horses receiving neostigmine. Inhibition of central muscarinic receptors by inhalation anesthetics may underlie emergence delirium in horses recovering from anesthesia.
Collapse
Affiliation(s)
- Ashley J Wiese
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616
| | | | | |
Collapse
|
38
|
NMDA receptor antagonists ketamine and Ro25-6981 inhibit evoked release of glutamate in vivo in the subiculum. Transl Psychiatry 2014; 4:e395. [PMID: 24893066 PMCID: PMC4080320 DOI: 10.1038/tp.2014.39] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 04/22/2014] [Indexed: 01/20/2023] Open
Abstract
Preclinical and clinical data have identified ketamine, a non-selective NMDAR (N-methyl-D-aspartate receptor) antagonist, as a promising medication for patients who do not respond to treatment with monoamine-based antidepressants. Moreover, unlike the current monoamine-based antidepressants, ketamine has a long-lasting effect already after a single dose. The mechanisms of ketamine action remain to be fully understood. Using a recently developed microelectrode array (MEA), which allows sub-second measurements of fluctuating glutamate concentrations, we studied here the effects of in vivo local application of the ketamine and of the N2B subunit-specific antagonist Ro25-6981 upon evoked glutamate release. Both ligands inhibit glutamate release in subregions of the hippocampus and prefrontal cortex. Likewise, acute systemic ketamine treatment, at an antidepressant dose, caused a reduction in evoked glutamate release in the subiculum. We suggest that the effects of ketamine and Ro25-6981 in the subiculum could involve blockade of presynaptic NMDA receptors containing N2B subunits.
Collapse
|
39
|
Modulation of a voltage-gated Na+ channel by sevoflurane involves multiple sites and distinct mechanisms. Proc Natl Acad Sci U S A 2014; 111:6726-31. [PMID: 24753583 DOI: 10.1073/pnas.1405768111] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Halogenated inhaled general anesthetic agents modulate voltage-gated ion channels, but the underlying molecular mechanisms are not understood. Many general anesthetic agents regulate voltage-gated Na(+) (NaV) channels, including the commonly used drug sevoflurane. Here, we investigated the putative binding sites and molecular mechanisms of sevoflurane action on the bacterial NaV channel NaChBac by using a combination of molecular dynamics simulation, electrophysiology, and kinetic analysis. Structural modeling revealed multiple sevoflurane interaction sites possibly associated with NaChBac modulation. Electrophysiologically, sevoflurane favors activation and inactivation at low concentrations (0.2 mM), and additionally accelerates current decay at high concentrations (2 mM). Explaining these observations, kinetic modeling suggests concurrent destabilization of closed states and low-affinity open channel block. We propose that the multiple effects of sevoflurane on NaChBac result from simultaneous interactions at multiple sites with distinct affinities. This multiple-site, multiple-mode hypothesis offers a framework to study the structural basis of general anesthetic action.
Collapse
|
40
|
Bardgett ME, Chen QH, Guo Q, Calderon AS, Andrade MA, Toney GM. Coping with dehydration: sympathetic activation and regulation of glutamatergic transmission in the hypothalamic PVN. Am J Physiol Regul Integr Comp Physiol 2014; 306:R804-13. [PMID: 24671240 DOI: 10.1152/ajpregu.00074.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Autonomic and endocrine profiles of chronic hypertension and heart failure resemble those of acute dehydration. Importantly, all of these conditions are associated with exaggerated sympathetic nerve activity (SNA) driven by glutamatergic activation of the hypothalamic paraventricular nucleus (PVN). Here, studies sought to gain insight into mechanisms of disease by determining the role of PVN ionotropic glutamate receptors in supporting SNA and mean arterial pressure (MAP) during dehydration and by elucidating mechanisms regulating receptor activity. Blockade of PVN N-methyl-D-aspartate (NMDA) receptors reduced (P < 0.01) renal SNA and MAP in urethane-chloralose-anesthetized dehydrated (DH) (48 h water deprivation) rats, but had no effect in euhydrated (EH) controls. Blockade of PVN α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors had no effect in either group. NMDA in PVN caused dose-dependent increases of renal SNA and MAP in both groups, but the maximum agonist evoked response (Emax) of the renal SNA response was greater (P < 0.05) in DH rats. The latter was not explained by increased PVN expression of NMDA receptor NR1 subunit protein, increased PVN neuronal excitability, or decreased brain water content. Interestingly, PVN injection of the pan-specific excitatory amino acid transporter (EAAT) inhibitor DL-threo-β-benzyloxyaspartic acid produced smaller sympathoexcitatory and pressor responses in DH rats, which was associated with reduced glial expression of EAAT2 in PVN. Like chronic hypertension and heart failure, dehydration increases excitatory NMDA receptor tone in PVN. Reduced glial-mediated glutamate uptake was identified as a key contributing factor. Defective glutamate uptake in PVN could therefore be an important, but as yet unexplored, mechanism driving sympathetic hyperactivity in chronic cardiovascular diseases.
Collapse
Affiliation(s)
| | - Qing-Hui Chen
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, Michigan
| | | | | | | | - Glenn M Toney
- Department of Physiology and Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas; and
| |
Collapse
|
41
|
Wang Q, Fong R, Mason P, Fox AP, Xie Z. Caffeine accelerates recovery from general anesthesia. J Neurophysiol 2014; 111:1331-40. [PMID: 24375022 PMCID: PMC3949308 DOI: 10.1152/jn.00792.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/26/2013] [Indexed: 11/22/2022] Open
Abstract
General anesthetics inhibit neurotransmitter release from both neurons and secretory cells. If inhibition of neurotransmitter release is part of an anesthetic mechanism of action, then drugs that facilitate neurotransmitter release may aid in reversing general anesthesia. Drugs that elevate intracellular cAMP levels are known to facilitate neurotransmitter release. Three cAMP elevating drugs (forskolin, theophylline, and caffeine) were tested; all three drugs reversed the inhibition of neurotransmitter release produced by isoflurane in PC12 cells in vitro. The drugs were tested in isoflurane-anesthetized rats. Animals were injected with either saline or saline containing drug. All three drugs dramatically accelerated recovery from isoflurane anesthesia, but caffeine was most effective. None of the drugs, at the concentrations tested, had significant effects on breathing rates, O2 saturation, heart rate, or blood pressure in anesthetized animals. Caffeine alone was tested on propofol-anesthetized rats where it dramatically accelerated recovery from anesthesia. The ability of caffeine to accelerate recovery from anesthesia for different chemical classes of anesthetics, isoflurane and propofol, opens the possibility that it will do so for all commonly used general anesthetics, although additional studies will be required to determine whether this is in fact the case. Because anesthesia in rodents is thought to be similar to that in humans, these results suggest that caffeine might allow for rapid and uniform emergence from general anesthesia in human patients.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Neurobiology, Pharmacology and Physiology, University of Chicago, Chicago, Illinois
| | | | | | | | | |
Collapse
|
42
|
van Swinderen B, Kottler B. Explaining general anesthesia: a two-step hypothesis linking sleep circuits and the synaptic release machinery. Bioessays 2014; 36:372-81. [PMID: 24449137 DOI: 10.1002/bies.201300154] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Several general anesthetics produce their sedative effect by activating endogenous sleep pathways. We propose that general anesthesia is a two-step process targeting sleep circuits at low doses, and synaptic release mechanisms across the entire brain at the higher doses required for surgery. Our hypothesis synthesizes data from a variety of model systems, some which require sleep (e.g. rodents and adult flies) and others that probably do not sleep (e.g. adult nematodes and cultured cell lines). Non-sleeping systems can be made insensitive (or hypersensitive) to some anesthetics by modifying a single pre-synaptic protein, syntaxin1A. This suggests that the synaptic release machinery, centered on the highly conserved SNARE complex, is an important target of general anesthetics in all animals. A careful consideration of SNARE architecture uncovers a potential mechanism for general anesthesia, which may be the primary target in animals that do not sleep, but a secondary target in animals that sleep.
Collapse
Affiliation(s)
- Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | | |
Collapse
|
43
|
Hu N, Guo D, Wang H, Xie K, Wang C, Li Y, Wang C, Wang C, Yu Y, Wang G. Involvement of the blood-brain barrier opening in cognitive decline in aged rats following orthopedic surgery and high concentration of sevoflurane inhalation. Brain Res 2014; 1551:13-24. [PMID: 24440777 DOI: 10.1016/j.brainres.2014.01.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/09/2014] [Accepted: 01/14/2014] [Indexed: 11/17/2022]
Abstract
The underlying causes of postoperative cognitive decline (POCD) in old patients remained unelucidated, and there are little descriptions on mechanisms associated with the blood-brain barrier (BBB) disruption during POCD. We therefore tested the effects of orthopedic surgery with different concentrations of sevoflurane for 2 h on the behavior test and the BBB permeability in aged rats. 18-month rats were divided into control group and surgical group with propofol anesthesia (0.7 mgkg(-1) min(-1)) and 1.0 MAC, 1.3 MAC, and 1.5 MAC sevoflurane inhalation for 2 h. We assessed their cognitive function via Y-maze and fear conditioning test on day 1, 3, and 7 after experiments. Animals were then assigned to control group, propofol (2 h, 0.7 mgkg(-1) min(-1)) group, surgery plus propofol group and surgery plus 1.5 MAC sevoflurane inhalation for 2h. Their hippocampal BBB permeability was detected with Evans blue quantification. Alterations of tight junctions in hippocampus were measured with occludin and claudin-5 western blot. Then we assessed matrix metalloproteinase-2,9 (MMP-2,9) via western blot and immunohistochemistry staining at day 1, 3, 7, and 14 after experiments. Surgery impaired cognitive function and increased Evans blue leakage into the hippocampus in aged rats while 2 h of 1.5 MAC sevoflurane inhalation potentiated these effects. Surgery induced occludin protein expression decreases and MMP-2,9 proteins increase and these influences can be enhanced by high concentration of sevoflurane inhalation. In conclusion, 1.5 MAC sevoflurane for 2 h exacerbated cognitive impairment induced by orthopedic surgery in aged rats and the breach in BBB may be involved in this process.
Collapse
Affiliation(s)
- Nan Hu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, China.
| | - Dongyong Guo
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, China.
| | - Haiyun Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, China.
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, China.
| | - Chao Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, China.
| | - Yize Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, China.
| | - Chunyan Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, China.
| | - Chenxu Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, China.
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, China.
| | - Guolin Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, China.
| |
Collapse
|
44
|
Ferris MJ, Calipari ES, Yorgason JT, Jones SR. Examining the complex regulation and drug-induced plasticity of dopamine release and uptake using voltammetry in brain slices. ACS Chem Neurosci 2013; 4:693-703. [PMID: 23581570 DOI: 10.1021/cn400026v] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Fast scan cyclic voltammetry in brain slices (slice voltammetry) has been used over the last several decades to increase substantially our understanding of the complex local regulation of dopamine release and uptake in the striatum. This technique is routinely used for the study of changes that occur in the dopamine system associated with various disease states and pharmacological treatments, and to study mechanisms of local circuitry regulation of dopamine terminal function. In the context of this Review, we compare the relative advantages of voltammetry using striatal slice preparations versus in vivo preparations, and highlight recent advances in our understanding of dopamine release and uptake in the striatum specifically from studies that use slice voltammetry in drug-naïve animals and animals with a history of psychostimulant self-administration.
Collapse
Affiliation(s)
- Mark J. Ferris
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Erin S. Calipari
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Jordan T. Yorgason
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Sara R. Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| |
Collapse
|