1
|
Magruder T, Isenhart M, Striepe MV, Mannisto A, Jannie KM, Smith J, McCarson KE, Christian DT, Duric V. Ketamine - An Imperfect Wonder Drug? Biochem Pharmacol 2024; 229:116516. [PMID: 39218043 DOI: 10.1016/j.bcp.2024.116516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Ketamine is a potent sedative and dissociative anesthetic agent that has been used clinically for over 50 years since it was first developed in the 1960 s as an alternative to phencyclidine (PCP). When compared to PCP, ketamine exhibited a much lower incidence of severe side effects, including hallucinations, leading to its increased popularity in clinical practice. Ketamine was initially used as an anesthetic agent, especially in emergency medicine and in surgical procedures where rapid induction and recovery was necessary. However, over the last few decades, ketamine was found to have additional clinically useful properties making it effective in the treatment of a variety of other conditions. Presently, ketamine has a wide range of clinical uses beyond anesthesia including management of acute and chronic pain, as well as treatment of psychiatric disorders such as major depression. In addition to various clinical uses, ketamine is also recognized as a common drug of abuse sought for its hallucinogenic and sedative effects. This review focuses on exploring the different clinical and non-clinical uses of ketamine and its overall impact on patient care.
Collapse
Affiliation(s)
- Tanner Magruder
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, USA
| | - Marielle Isenhart
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, USA
| | - Maximillian V Striepe
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, USA
| | - Andrew Mannisto
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, USA
| | - Karry M Jannie
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, USA
| | - Jolene Smith
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, USA
| | - Kenneth E McCarson
- Department of Pharmacology, Toxicology and Experimental Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Daniel T Christian
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, USA
| | - Vanja Duric
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, USA.
| |
Collapse
|
2
|
Ko JC, Murillo C, Weil AB, Kreuzer M, Moore GE. Electroencephalographic and Cardiovascular Assessments of Isoflurane-Anesthetized Dogs. Vet Sci 2024; 11:514. [PMID: 39453106 PMCID: PMC11512366 DOI: 10.3390/vetsci11100514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
This study investigated the use of frontal electroencephalography (EEG) to monitor varying levels of isoflurane anesthesia in dogs. The patient state index (PSI), burst suppression ratio (SR), and waveforms, were continuously recorded while mean arterial blood pressure (MBP), heart rate, responses to electric stimuli, and subjective anesthetic "depth" were assessed every 3 min. At deep anesthesia (2.5× MAC - 3.2%), the PSI (6.5 ± 10.8) and MBP (45.6 ± 16.4 mmHg) were the lowest, and SR was the highest (78.3 ± 24.0%). At 1× MAC (1.3%), the PSI and MBP increased significantly to 47.8 ± 12.6 and 99.8 ± 13.2, respectively, and SR decreased to 0.5 ± 2.5%. The EEG was predominantly isoelectric at 2×-2.5× MAC, indicating unconsciousness and unresponsiveness. As anesthesia lightened, waveforms transitioned to flatter and faster activity patterns with a response to noxious stimuli, suggesting regained consciousness. The PSI and MBP exhibited a stronger correlation (ρ = 0.8098, p = 0.001) than the relationship of PSI with heart rate (ρ = -0.2089, p = 0.249). Five of the six dogs experienced rough recovery, possibly due to high SR and low MBP. These findings suggest that EEG monitoring in dogs can be a valuable tool for the real-time tracking of brain states and can be used to guide the management of isoflurane anesthesia.
Collapse
Affiliation(s)
- Jeff C. Ko
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (C.M.); (A.B.W.)
| | - Carla Murillo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (C.M.); (A.B.W.)
| | - Ann B. Weil
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (C.M.); (A.B.W.)
| | - Matthias Kreuzer
- Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, 80333 München, Germany;
| | - George E. Moore
- Department of Veterinary Administration, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA;
| |
Collapse
|
3
|
Pardo-Valencia J, Moreno-Gomez M, Mercado N, Pro B, Ammann C, Humanes-Valera D, Foffani G. Local wakefulness-like activity of layer 5 cortex under general anaesthesia. J Physiol 2024; 602:5289-5307. [PMID: 39316039 DOI: 10.1113/jp286417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 08/08/2024] [Indexed: 09/25/2024] Open
Abstract
Consciousness, defined as being aware of and responsive to one's surroundings, is characteristic of normal waking life and typically is lost during sleep and general anaesthesia. The traditional view of consciousness as a global brain state has evolved toward a more sophisticated interplay between global and local states, with the presence of local sleep in the awake brain and local wakefulness in the sleeping brain. However, this interplay is not clear for general anaesthesia, where loss of consciousness was recently suggested to be associated with a global state of brain-wide synchrony that selectively involves layer 5 cortical pyramidal neurons across sensory, motor and associative areas. According to this global view, local wakefulness of layer 5 cortex should be incompatible with deep anaesthesia, a hypothesis that deserves to be scrutinised with causal manipulations. Here, we show that unilateral chemogenetic activation of layer 5 pyramidal neurons in the sensorimotor cortex of isoflurane-anaesthetised mice induces a local state transition from slow-wave activity to tonic firing in the transfected hemisphere. This wakefulness-like activity dramatically disrupts layer 5 interhemispheric synchrony with mirror-image locations in the contralateral hemisphere, but does not reduce the level of unconsciousness under deep anaesthesia, nor in the transitions to/from anaesthesia. Global layer 5 synchrony may thus be a sufficient condition for anaesthesia-induced unconsciousness, but is not a necessary one, at least under isoflurane anaesthesia. Local wakefulness-like activity of layer 5 cortex can be induced and maintained under deep anaesthesia, encouraging further investigation into the local vs. global aspects of anaesthesia-induced unconsciousness. KEY POINTS: The neural correlates of consciousness have evolved from global brain states to a nuanced interplay between global and local states, evident in terms of local sleep in awake brains and local wakefulness in sleeping brains. The concept of local wakefulness remains unclear for general anaesthesia, where the loss of consciousness has been recently suggested to involve brain-wide synchrony of layer 5 cortical neurons. We found that local wakefulness-like activity of layer 5 cortical can be chemogenetically induced in anaesthetised mice without affecting the depth of anaesthesia or the transitions to and from unconsciousness. Global layer 5 synchrony may thus be a sufficient but not necessary feature for the unconsciousness induced by general anaesthesia. Local wakefulness-like activity of layer 5 neurons is compatible with general anaesthesia, thus promoting further investigation into the local vs. global aspects of anaesthesia-induced unconsciousness.
Collapse
Affiliation(s)
- Jesús Pardo-Valencia
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Escuela Técnica Superior de Ingenieros de Telecomunicación, Universidad Politécnica de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
| | - Miryam Moreno-Gomez
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
- PhD Program in Neuroscience, Universidad Autonoma de Madrid-Cajal Institute, Madrid, Spain
| | - Noelia Mercado
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
| | - Beatriz Pro
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
| | - Claudia Ammann
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain
| | - Desire Humanes-Valera
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
| | - Guglielmo Foffani
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
- Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Gotoh M, Dezawa S, Takashima I, Yamamoto S. Effects of focal cortical cooling on somatosensory evoked potentials in rats. Brain Res 2024; 1840:148995. [PMID: 38735427 DOI: 10.1016/j.brainres.2024.148995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/26/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024]
Abstract
Although the focal brain cooling technique is widely used to examine brain function, the effects of cortical temperature at various levels on sensory information processing and neural mechanisms remain underexplored. To elucidate the mechanisms of temperature modulation in somatosensory processing, this study aimed to examine how P1 and N1 deflections of somatosensory evoked potentials (SEPs) depend on cortical temperature and how excitatory and inhibitory inputs contribute to this temperature dependency. SEPs were generated through electrical stimulation of the contralateral forepaw in anesthetized rats. The SEPs were recorded while cortical temperatures were altered between 17-38 °C either without any antagonists, with a gamma-aminobutyric acid type A (GABAA) receptor antagonist (gabazine), with an aminomethylphosphonic acid (AMPA) receptor antagonist (NBQX), or with an N-Methyl-D-aspartic acid (NMDA) receptor antagonist ([R]-CPP). The effects of different gabazine concentrations (0, 1, and 10 µM) were examined in the 35-38 °C range. The P1/N1 amplitudes and their peak-to-peak differences plotted against cortical temperature showed an inverted U relationship with a maximum at approximately 27.5 °C when no antagonists were administered. The negative correlation between these amplitudes and temperatures of ≥ 27.5 °C plateaued after gabazine administration, which occurred progressively as the gabazine concentration increased. In contrast, the correlation remained negative after the administration of NBQX and (R)-CPP. These results suggest that GABAergic inhibitory inputs contribute to the negative correlation between SEP amplitude and cortical temperature around the physiological cortical temperature.
Collapse
Affiliation(s)
- Mizuho Gotoh
- Department of Information Technology and Human Factors, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba Japan; Department of Rehabilitation for Brain Functions, Research Institute of National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Shinnosuke Dezawa
- Department of Information Technology and Human Factors, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba Japan; Faculty of Medical and Health Sciences, Tsukuba International University, Tsuchiura, Japan
| | - Ichiro Takashima
- Department of Information Technology and Human Factors, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba Japan; Department of Information, Artificial Intelligence and Data Science, Daiichi Institute of Technology, Tokyo, Japan
| | - Shinya Yamamoto
- Department of Information Technology and Human Factors, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba Japan.
| |
Collapse
|
5
|
Li M, Zhang R, Wu S, Cheng L, Fu H, Qu L. Isoflurane anesthesia decreases excitability of inhibitory neurons in the basolateral amygdala leading to anxiety‑like behavior in aged mice. Exp Ther Med 2024; 28:399. [PMID: 39171147 PMCID: PMC11336806 DOI: 10.3892/etm.2024.12688] [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: 03/13/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024] Open
Abstract
Anxiety after surgery can be a major factor leading to postoperative cognitive dysfunction, particularly in elderly patients. The role of inhibitory neurons in the basolateral amygdala (BLA) in anxiety-like behaviors in aged mice following isoflurane anesthesia remains unclear. Therefore, the present study aimed to investigate the role of inhibitory neurons in isoflurane-treated mice. A total of 30 C57BL/6 mice (age, 13 months) were allocated into the control and isoflurane anesthesia groups (15 mice/group) and were then subjected to several neurological assessments. Behavioral testing using an elevated plus maze test showed that aged mice in the isoflurane anesthesia group displayed significant anxiety-like behavior, since they spent more time in the closed arm, exhibited more wall climbing behavior and covered more distance. In addition, whole-cell patch-clamp recording revealed that the excitability of the BLA excitatory neurons was notably increased following mice anesthesia with isoflurane, while that of inhibitory neurons was markedly reduced. Following mice treatment with diazepam, the excitability of the BLA inhibitory neurons was notably increased compared with that of the excitatory neurons, which was significantly attenuated. Overall, the results of the current study indicated that anxiety-like behavior could occur in aged mice after isoflurane anesthesia, which could be caused by a reduced excitability of the inhibitory neurons in the BLA area. This process could enhance excitatory neuronal activity in aged mice, thus ultimately promoting the onset of anxiety-like behaviors.
Collapse
Affiliation(s)
- Mengyuan Li
- Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Ruijiao Zhang
- Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Shiyin Wu
- Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Liqin Cheng
- Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Huan Fu
- Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Liangchao Qu
- Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330000, P.R. China
- Department of Anesthesia and Surgery, People's Hospital of Ganjiang New District, Nanchang, Jiangxi 341099, P.R. China
| |
Collapse
|
6
|
Brenna CTA, Goldstein BI, Zarate CA, Orser BA. Repurposing General Anesthetic Drugs to Treat Depression: A New Frontier for Anesthesiologists in Neuropsychiatric Care. Anesthesiology 2024; 141:222-237. [PMID: 38856663 DOI: 10.1097/aln.0000000000005037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
During the last 100 years, the role of anesthesiologists in psychiatry has focused primarily on facilitating electroconvulsive therapy and mitigating postoperative delirium and other perioperative neurocognitive disorders. The discovery of the rapid and sustained antidepressant properties of ketamine, and early results suggesting that other general anesthetic drugs (including nitrous oxide, propofol, and isoflurane) have antidepressant properties, has positioned anesthesiologists at a new frontier in the treatment of neuropsychiatric disorders. Moreover, shared interest in understanding the biologic underpinnings of anesthetic drugs as psychotropic agents is eroding traditional academic boundaries between anesthesiology and psychiatry. This article presents a brief overview of anesthetic drugs as novel antidepressants and identifies promising future candidates for the treatment of depression. The authors issue a call to action and outline strategies to foster collaborations between anesthesiologists and psychiatrists as they work toward the common goals of repurposing anesthetic drugs as antidepressants and addressing mood disorders in surgical patients.
Collapse
Affiliation(s)
- Connor T A Brenna
- Department of Anesthesiology & Pain Medicine and Department of Physiology, University of Toronto, Toronto, Canada; Perioperative Brain Health Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Benjamin I Goldstein
- Centre for Addiction and Mental Health, Toronto, Canada; Department of Psychiatry and Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Beverley A Orser
- Department of Anesthesiology & Pain Medicine and Department of Physiology, University of Toronto, Toronto, Canada; Perioperative Brain Health Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| |
Collapse
|
7
|
Yu Q, Wang Y, Gu L, Shao W, Gu J, Liu L, Lian X, Xu Q, Zhang Y, Yang Y, Zhang Z, Wu Y, Ma H, Shen Y, Ye W, Wu Y, Yang H, Chen L, Nagayasu K, Zhang H. Dorsal raphe nucleus to basolateral amygdala 5-HTergic neural circuit modulates restoration of consciousness during sevoflurane anesthesia. Biomed Pharmacother 2024; 176:116937. [PMID: 38870632 DOI: 10.1016/j.biopha.2024.116937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/06/2024] [Accepted: 06/09/2024] [Indexed: 06/15/2024] Open
Abstract
The advent of general anesthesia (GA) has significant implications for clinical practice. However, the exact mechanisms underlying GA-induced transitions in consciousness remain elusive. Given some similarities between GA and sleep, the sleep-arousal neural nuclei and circuits involved in sleep-arousal, including the 5-HTergic system, could be implicated in GA. Herein, we utilized pharmacology, optogenetics, chemogenetics, fiber photometry, and retrograde tracing to demonstrate that both endogenous and exogenous activation of the 5-HTergic neural circuit between the dorsal raphe nucleus (DR) and basolateral amygdala (BLA) promotes arousal and facilitates recovery of consciousness from sevoflurane anesthesia. Notably, the 5-HT1A receptor within this pathway holds a pivotal role. Our findings will be conducive to substantially expanding our comprehension of the neural circuit mechanisms underlying sevoflurane anesthesia and provide a potential target for modulating consciousness, ultimately leading to a reduction in anesthetic dose requirements and side effects.
Collapse
Affiliation(s)
- Qian Yu
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - YuLing Wang
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - LeYuan Gu
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - WeiHui Shao
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - JiaXuan Gu
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Lu Liu
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - XiTing Lian
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Qing Xu
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - YuanLi Zhang
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Yue Yang
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - ZhuoYue Zhang
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - YaXuan Wu
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - HaiXiang Ma
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Yue Shen
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China
| | - Wen Ye
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China
| | - YanHui Wu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China
| | - HuiFang Yang
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China
| | - LiHai Chen
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 606-8501, Japan
| | - HongHai Zhang
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou 310006, China; Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
8
|
Gao X, Li Z, Chai J, Li S, Pan X, Liu J, Li L, Qin S, Kang Y, Zhu Y. Electroencephalographic insights into the pathophysiological mechanisms of emergence delirium in children and corresponding clinical treatment strategies. Front Pharmacol 2024; 15:1349105. [PMID: 38962301 PMCID: PMC11219819 DOI: 10.3389/fphar.2024.1349105] [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: 12/04/2023] [Accepted: 02/26/2024] [Indexed: 07/05/2024] Open
Abstract
Emergence delirium is a common postoperative complication in patients undergoing general anesthesia, especially in children. In severe cases, it can cause unnecessary self-harm, affect postoperative recovery, lead to parental dissatisfaction, and increase medical costs. With the widespread use of inhalation anesthetic drugs (such as sevoflurane and desflurane), the incidence of emergence delirium in children is gradually increasing; however, its pathogenesis in children is complex and unclear. Several studies have shown that age, pain, and anesthetic drugs are strongly associated with the occurrence of emergence delirium. Alterations in central neurophysiology are essential intermediate processes in the development of emergence delirium. Compared to adults, the pediatric nervous system is not fully developed; therefore, the pediatric electroencephalogram may vary slightly by age. Moreover, pain and anesthetic drugs can cause changes in the excitability of the central nervous system, resulting in electroencephalographic changes. In this paper, we review the pathogenesis of and prevention strategies for emergence delirium in children from the perspective of brain electrophysiology-especially for commonly used pharmacological treatments-to provide the basis for understanding the development of emergence delirium as well as its prevention and treatment, and to suggest future research direction.
Collapse
Affiliation(s)
- Xin Gao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhichao Li
- Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jun Chai
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Si Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuanyuan Pan
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jie Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Linxing Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shangyuan Qin
- Department of Anesthesiology, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Yihan Kang
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Youzhuang Zhu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Orser BA. Discovering the Intriguing Properties of Extrasynaptic γ-Aminobutyric Acid Type A Receptors. Anesthesiology 2024; 140:1192-1200. [PMID: 38624275 DOI: 10.1097/aln.0000000000004949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Tonic inhibition in mouse hippocampal CA1 pyramidal neurons is mediated by α5 subunit-containing γ-aminobutyric acid type A receptors. By Caraiscos VB, Elliott EM, You-Ten KE, Cheng VY, Belelli D, Newell JG, Jackson MF, Lambert JJ, Rosahl TW, Wafford KA, MacDonald JF, Orser BA. Proc Natl Acad Sci U S A 2004; 101:3662-7. Reprinted with permission. In this Classic Paper Revisited, the author recounts the scientific journey leading to a report published in the Proceedings of the National Academy of Sciences (PNAS) and shares several personal stories from her formative years and "research truths" that she has learned along the way. Briefly, the principal inhibitory neurotransmitter in the brain, γ-aminobutyric acid (GABA), was conventionally thought to regulate cognitive processes by activating synaptic GABA type A (GABAA) receptors and generating transient inhibitory synaptic currents. However, the author's laboratory team discovered a novel nonsynaptic form of tonic inhibition in hippocampal pyramidal neurons, mediated by extrasynaptic GABAA receptors that are pharmacologically distinct from synaptic GABAA receptors. This tonic current is highly sensitive to most general anesthetics, including sevoflurane and propofol, and likely contributes to the memory-blocking properties of these drugs. Before the publication in PNAS, the subunit composition of GABAA receptors that generate the tonic current was unknown. The team's research showed that GABAA receptors containing the α5 subunit (α5GABAARs) generated the tonic inhibitory current in hippocampal neurons. α5GABAARs are highly sensitive to GABA, desensitize slowly, and are thus well suited for detecting low, persistent, ambient concentrations of GABA in the extracellular space. Interest in α5GABAARs has surged since the PNAS report, driven by their pivotal roles in cognitive processes and their potential as therapeutic targets for treating various neurologic disorders.
Collapse
Affiliation(s)
- Beverley A Orser
- Department of Anesthesiology and Pain Medicine, and Department of Physiology, University of Toronto, Toronto, Canada
| |
Collapse
|
10
|
Obert DP, Killing D, Happe T, Tamas P, Altunkaya A, Dragovic SZ, Kreuzer M, Schneider G, Fenzl T. Substance specific EEG patterns in mice undergoing slow anesthesia induction. BMC Anesthesiol 2024; 24:167. [PMID: 38702608 PMCID: PMC11067159 DOI: 10.1186/s12871-024-02552-3] [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: 03/06/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024] Open
Abstract
The exact mechanisms and the neural circuits involved in anesthesia induced unconsciousness are still not fully understood. To elucidate them valid animal models are necessary. Since the most commonly used species in neuroscience are mice, we established a murine model for commonly used anesthetics/sedatives and evaluated the epidural electroencephalographic (EEG) patterns during slow anesthesia induction and emergence. Forty-four mice underwent surgery in which we inserted a central venous catheter and implanted nine intracranial electrodes above the prefrontal, motor, sensory, and visual cortex. After at least one week of recovery, mice were anesthetized either by inhalational sevoflurane or intravenous propofol, ketamine, or dexmedetomidine. We evaluated the loss and return of righting reflex (LORR/RORR) and recorded the electrocorticogram. For spectral analysis we focused on the prefrontal and visual cortex. In addition to analyzing the power spectral density at specific time points we evaluated the changes in the spectral power distribution longitudinally. The median time to LORR after start anesthesia ranged from 1080 [1st quartile: 960; 3rd quartile: 1080]s under sevoflurane anesthesia to 1541 [1455; 1890]s with ketamine. Around LORR sevoflurane as well as propofol induced a decrease in the theta/alpha band and an increase in the beta/gamma band. Dexmedetomidine infusion resulted in a shift towards lower frequencies with an increase in the delta range. Ketamine induced stronger activity in the higher frequencies. Our results showed substance-specific changes in EEG patterns during slow anesthesia induction. These patterns were partially identical to previous observations in humans, but also included significant differences, especially in the low frequencies. Our study emphasizes strengths and limitations of murine models in neuroscience and provides an important basis for future studies investigating complex neurophysiological mechanisms.
Collapse
Affiliation(s)
- David P Obert
- School of Medicine and Health, Department of Anesthesiology and Intensive Care, Technical University of Munich, 81675, Munich, Germany
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts's General Hospital, Boston, MA, 02114, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - David Killing
- School of Medicine and Health, Department of Anesthesiology and Intensive Care, Technical University of Munich, 81675, Munich, Germany
| | - Tom Happe
- School of Medicine and Health, Department of Anesthesiology and Intensive Care, Technical University of Munich, 81675, Munich, Germany
| | - Philipp Tamas
- School of Medicine and Health, Department of Anesthesiology and Intensive Care, Technical University of Munich, 81675, Munich, Germany
| | - Alp Altunkaya
- School of Medicine and Health, Department of Anesthesiology and Intensive Care, Technical University of Munich, 81675, Munich, Germany
| | - Srdjan Z Dragovic
- School of Medicine and Health, Department of Anesthesiology and Intensive Care, Technical University of Munich, 81675, Munich, Germany
| | - Matthias Kreuzer
- School of Medicine and Health, Department of Anesthesiology and Intensive Care, Technical University of Munich, 81675, Munich, Germany
| | - Gerhard Schneider
- School of Medicine and Health, Department of Anesthesiology and Intensive Care, Technical University of Munich, 81675, Munich, Germany
| | - Thomas Fenzl
- School of Medicine and Health, Department of Anesthesiology and Intensive Care, Technical University of Munich, 81675, Munich, Germany.
| |
Collapse
|
11
|
Abdallah RM, Hasan HE, Hammad A. Predictive modeling of skin permeability for molecules: Investigating FDA-approved drug permeability with various AI algorithms. PLOS DIGITAL HEALTH 2024; 3:e0000483. [PMID: 38568888 PMCID: PMC10990209 DOI: 10.1371/journal.pdig.0000483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 03/05/2024] [Indexed: 04/05/2024]
Abstract
The transdermal route of drug administration has gained popularity for its convenience and bypassing the first-pass metabolism. Accurate skin permeability prediction is crucial for successful transdermal drug delivery (TDD). In this study, we address this critical need to enhance TDD. A dataset comprising 441 records for 140 molecules with diverse LogKp values was characterized. The descriptor calculation yielded 145 relevant descriptors. Machine learning models, including MLR, RF, XGBoost, CatBoost, LGBM, and ANN, were employed for regression analysis. Notably, LGBM, XGBoost, and gradient boosting models outperformed others, demonstrating superior predictive accuracy. Key descriptors influencing skin permeability, such as hydrophobicity, hydrogen bond donors, hydrogen bond acceptors, and topological polar surface area, were identified and visualized. Cluster analysis applied to the FDA-approved drug dataset (2326 compounds) revealed four distinct clusters with significant differences in molecular characteristics. Predicted LogKp values for these clusters offered insights into the permeability variations among FDA-approved drugs. Furthermore, an investigation into skin permeability patterns across 83 classes of FDA-approved drugs based on the ATC code showcased significant differences, providing valuable information for drug development strategies. The study underscores the importance of accurate skin permeability prediction for TDD, emphasizing the superior performance of nonlinear machine learning models. The identified key descriptors and clusters contribute to a nuanced understanding of permeability characteristics among FDA-approved drugs. These findings offer actionable insights for drug design, formulation, and prioritization of molecules with optimum properties, potentially reducing reliance on costly experimental testing. Future research directions include offering promising applications in pharmaceutical research and formulation within the burgeoning field of computer-aided drug design.
Collapse
Affiliation(s)
- Rami M. Abdallah
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Zarqa University, Zarqa, Jordan
| | - Hisham E. Hasan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Zarqa University, Zarqa, Jordan
| | - Ahmad Hammad
- Department of Artificial Intelligence, Faculty of Information Technology, Middle East University, Amman, Jordan
| |
Collapse
|
12
|
Wan J, Ma L, Jiao X, Dong W, Lin J, Qiu Y, Wu W, Liu Q, Chen C, Huang H, Li S, Zheng H, Wu Y. Impaired synaptic plasticity and decreased excitability of hippocampal glutamatergic neurons mediated by BDNF downregulation contribute to cognitive dysfunction in mice induced by repeated neonatal exposure to ketamine. CNS Neurosci Ther 2024; 30:e14604. [PMID: 38332635 PMCID: PMC10853651 DOI: 10.1111/cns.14604] [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: 11/05/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 02/10/2024] Open
Abstract
AIM Repeated exposure to ketamine during the neonatal period in mice leads to cognitive impairments in adulthood. These impairments are likely caused by synaptic plasticity and excitability damage. We investigated the precise role of brain-derived neurotrophic factor (BDNF) in the cognitive impairments induced by repeated ketamine exposure during the neonatal period. METHODS We evaluated the cognitive function of mice using the Morris water maze test and novel object recognition test. Western blotting and immunofluorescence were used to detect the protein levels of BDNF. Western blotting, Golgi-Cox staining, transmission electron microscopy, and long-term potentiation (LTP) recordings were used to assess synaptic plasticity in the hippocampus. The excitability of neurons was evaluated using c-Fos. In the intervention experiment, pAdeno-CaMKIIα-BDNF-mNeuronGreen was injected into the hippocampal CA1 region of mice to increase the level of BDNF. The excitability of neurons was enhanced using a chemogenetic approach. RESULTS Our findings suggest that cognitive impairments in mice repeatedly exposed to ketamine during the neonatal period are associated with downregulated BDNF protein level, synaptic plasticity damage, and decreased excitability of glutamatergic neurons in the hippocampal CA1 region. Furthermore, the specific upregulation of BDNF in glutamatergic neurons of the hippocampal CA1 region and the enhancement of excitability can improve impaired synaptic plasticity and cognitive function in mice. CONCLUSION BDNF downregulation mediates synaptic plasticity and excitability damage, leading to cognitive impairments in adulthood following repeated ketamine exposure during the neonatal period.
Collapse
Affiliation(s)
- Jie Wan
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Linhui Ma
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xinhao Jiao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Wei Dong
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Jiatao Lin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Yongkang Qiu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Weifeng Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Qiang Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Chen Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - He Huang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| | - Shuai Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yuqing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhouChina
| |
Collapse
|
13
|
Tauber JM, Brincat SL, Stephen EP, Donoghue JA, Kozachkov L, Brown EN, Miller EK. Propofol-mediated Unconsciousness Disrupts Progression of Sensory Signals through the Cortical Hierarchy. J Cogn Neurosci 2024; 36:394-413. [PMID: 37902596 PMCID: PMC11161138 DOI: 10.1162/jocn_a_02081] [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: 10/31/2023]
Abstract
A critical component of anesthesia is the loss of sensory perception. Propofol is the most widely used drug for general anesthesia, but the neural mechanisms of how and when it disrupts sensory processing are not fully understood. We analyzed local field potential and spiking recorded from Utah arrays in auditory cortex, associative cortex, and cognitive cortex of nonhuman primates before and during propofol-mediated unconsciousness. Sensory stimuli elicited robust and decodable stimulus responses and triggered periods of stimulus-related synchronization between brain areas in the local field potential of Awake animals. By contrast, propofol-mediated unconsciousness eliminated stimulus-related synchrony and drastically weakened stimulus responses and information in all brain areas except for auditory cortex, where responses and information persisted. However, we found stimuli occurring during spiking Up states triggered weaker spiking responses than in Awake animals in auditory cortex, and little or no spiking responses in higher order areas. These results suggest that propofol's effect on sensory processing is not just because of asynchronous Down states. Rather, both Down states and Up states reflect disrupted dynamics.
Collapse
Affiliation(s)
- John M Tauber
- Massachusetts Institute of Technology, Cambridge, MA
| | | | | | | | - Leo Kozachkov
- Massachusetts Institute of Technology, Cambridge, MA
| | - Emery N Brown
- Massachusetts Institute of Technology, Cambridge, MA
- Massachusetts General Hospital, Boston
- Harvard University, Cambridge, MA
| | - Earl K Miller
- Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
14
|
Okada R, Ikegaya Y, Matsumoto N. Short-Term Preexposure to Novel Enriched Environment Augments Hippocampal Ripples in Urethane-Anesthetized Mice. Biol Pharm Bull 2024; 47:1021-1027. [PMID: 38797694 DOI: 10.1248/bpb.b24-00118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Learning and memory are affected by novel enriched environment, a condition where animals play and interact with a variety of toys and conspecifics. Exposure of animals to the novel enriched environments improves memory by altering neural plasticity during natural sleep, a process called memory consolidation. The hippocampus, a pivotal brain region for learning and memory, generates high-frequency oscillations called ripples during sleep, which is required for memory consolidation. Naturally occurring sleep shares characteristics in common with general anesthesia in terms of extracellular oscillations, guaranteeing anesthetized animals suitable to examine neural activity in a sleep-like state. However, it is poorly understood whether the preexposure of animals to the novel enriched environment modulates neural activity in the hippocampus under subsequent anesthesia. To ask this question, we allowed mice to freely explore the novel enriched environment or their standard environment, anesthetized them, and recorded local field potentials in the hippocampal CA1 area. We then compared the characteristics of hippocampal ripples between the two groups and found that the amplitude of ripples and the number of successive ripples were larger in the novel enriched environment group than in the standard environment group, suggesting that the afferent synaptic input from the CA3 area to the CA1 area was higher when the animals underwent the novel enriched environment. These results underscore the importance of prior experience that surpasses subsequent physical states from the neurophysiological point of view.
Collapse
Affiliation(s)
- Rio Okada
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
- Institute for AI and Beyond, The University of Tokyo
- Center for Information and Neural Networks, National Institute of Information and Communications Technology
| | - Nobuyoshi Matsumoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
- Institute for AI and Beyond, The University of Tokyo
| |
Collapse
|
15
|
Monteil A, Guérineau NC, Gil-Nagel A, Parra-Diaz P, Lory P, Senatore A. New insights into the physiology and pathophysiology of the atypical sodium leak channel NALCN. Physiol Rev 2024; 104:399-472. [PMID: 37615954 DOI: 10.1152/physrev.00014.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/13/2023] [Accepted: 08/15/2023] [Indexed: 08/25/2023] Open
Abstract
Cell excitability and its modulation by hormones and neurotransmitters involve the concerted action of a large repertoire of membrane proteins, especially ion channels. Unique complements of coexpressed ion channels are exquisitely balanced against each other in different excitable cell types, establishing distinct electrical properties that are tailored for diverse physiological contributions, and dysfunction of any component may induce a disease state. A crucial parameter controlling cell excitability is the resting membrane potential (RMP) set by extra- and intracellular concentrations of ions, mainly Na+, K+, and Cl-, and their passive permeation across the cell membrane through leak ion channels. Indeed, dysregulation of RMP causes significant effects on cellular excitability. This review describes the molecular and physiological properties of the Na+ leak channel NALCN, which associates with its accessory subunits UNC-79, UNC-80, and NLF-1/FAM155 to conduct depolarizing background Na+ currents in various excitable cell types, especially neurons. Studies of animal models clearly demonstrate that NALCN contributes to fundamental physiological processes in the nervous system including the control of respiratory rhythm, circadian rhythm, sleep, and locomotor behavior. Furthermore, dysfunction of NALCN and its subunits is associated with severe pathological states in humans. The critical involvement of NALCN in physiology is now well established, but its study has been hampered by the lack of specific drugs that can block or agonize NALCN currents in vitro and in vivo. Molecular tools and animal models are now available to accelerate our understanding of how NALCN contributes to key physiological functions and the development of novel therapies for NALCN channelopathies.
Collapse
Affiliation(s)
- Arnaud Monteil
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nathalie C Guérineau
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
| | - Antonio Gil-Nagel
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Paloma Parra-Diaz
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Philippe Lory
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
| | - Adriano Senatore
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| |
Collapse
|
16
|
Speigel I, Patel K, Osman V, Hemmings HC. Volatile anesthetics inhibit presynaptic cGMP signaling to depress presynaptic excitability in rat hippocampal neurons. Neuropharmacology 2023; 240:109705. [PMID: 37683886 PMCID: PMC10772825 DOI: 10.1016/j.neuropharm.2023.109705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 07/21/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023]
Abstract
Volatile anesthetics alter presynaptic function through effects on Ca2+ influx and neurotransmitter release. These actions are proposed to play important roles in their pleiotropic neurophysiological effects including immobility, unconsciousness and amnesia. Nitric oxide and cyclic guanosine monophosphate (NO/cGMP) signaling has been implicated in presynaptic mechanisms, and disruption of NO/cGMP signaling has been shown to alter sensitivity to volatile anesthetics in vivo. We investigated volatile anesthetic actions NO/cGMP signaling in relation to presynaptic function in cultured rat hippocampal neurons using pharmacological tools and genetically encoded biosensors and sequestering probes of cGMP levels. Using the fluorescent cGMP biosensor cGull, we found that electrical stimulation-evoked NMDA-type glutamate receptor-independent presynaptic cGMP transients were inhibited 33.2% by isoflurane (0.51 mM) and 26.4% by sevoflurane (0.57 mM) (p < 0.0001) compared to control stimulation without anesthetic. Stimulation-evoked cGMP transients were blocked by the nonselective inhibitor of nitric oxide synthase N-ω-nitro-l-arginine, but not by the selective neuronal nitric oxide synthase inhibitor N5-(1-imino-3-butenyl)-l-ornithine. Isoflurane and sevoflurane inhibition of stimulation-evoked increases in presynaptic Ca2+ concentration, measured with synaptophysin-GCaMP6f, and of synaptic vesicle exocytosis, measured with synaptophysin-pHlourin, was attenuated in neurons expressing the cGMP scavenger protein sponge (inhibition of exocytosis reduced by 54% for isoflurane and by 53% for sevoflurane). The anesthetic-induced reduction in presynaptic excitability was partially occluded by inhibition of HCN channels, a cGMP-modulated excitatory ion channel that can facilitate glutamate release. We propose that volatile anesthetics depress presynaptic cGMP signaling and downstream effectors like HCN channels that are essential to presynaptic function and excitability. These findings identify novel mechanisms by which volatile anesthetics depress synaptic transmission via second messenger signaling involving the NO/cGMP pathway in hippocampal neurons.
Collapse
Affiliation(s)
- Iris Speigel
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Kishan Patel
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Vanessa Osman
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Hugh C Hemmings
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, 10065, USA; Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
17
|
Osman V, Speigel I, Patel K, Hemmings HC. Isoflurane Alters Presynaptic Endoplasmic Reticulum Calcium Dynamics in Wild-Type and Malignant Hyperthermia-Susceptible Rodent Hippocampal Neurons. eNeuro 2023; 10:ENEURO.0114-23.2023. [PMID: 37591734 PMCID: PMC10467020 DOI: 10.1523/eneuro.0114-23.2023] [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: 04/05/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 08/19/2023] Open
Abstract
Volatile anesthetics reduce excitatory synaptic transmission by both presynaptic and postsynaptic mechanisms which include inhibition of depolarization-evoked increases in presynaptic Ca2+ concentration and blockade of postsynaptic excitatory glutamate receptors. The presynaptic sites of action leading to reduced electrically evoked increases in presynaptic Ca2+ concentration and Ca2+-dependent exocytosis are unknown. Endoplasmic reticulum (ER) of Ca2+ release via ryanodine receptor 1 (RyR1) and uptake by SERCA are essential for regulation intracellular Ca2+ and are potential targets for anesthetic action. Mutations in sarcoplasmic reticulum (SR) release channels mediate volatile anesthetic-induced malignant hyperthermia (MH), a potentially fatal pharmacogenetic condition characterized by unregulated Ca2+ release and muscle hypermetabolism. However, the impact of MH mutations on neuronal function are unknown. We used primary cultures of postnatal hippocampal neurons to analyze volatile anesthetic-induced changes in ER Ca2+ dynamics using a genetically encoded ER-targeted fluorescent Ca2+ sensor in both rat and mouse wild-type (WT) neurons and in mouse mutant neurons harboring the RYR1 T4826I MH-susceptibility mutation. The volatile anesthetic isoflurane reduced both baseline and electrical stimulation-evoked increases in ER Ca2+ concentration in neurons independent of its depression of presynaptic cytoplasmic Ca2+ concentrations. Isoflurane and sevoflurane, but not propofol, depressed depolarization-evoked increases in ER Ca2+ concentration significantly more in mouse RYR1 T4826I mutant neurons than in wild-type neurons. The RYR1 T4826I mutant neurons also showed markedly greater isoflurane-induced reductions in presynaptic cytosolic Ca2+ concentration and synaptic vesicle (SV) exocytosis. These findings implicate RyR1 as a molecular target for the effects of isoflurane on presynaptic Ca2+ handling.
Collapse
Affiliation(s)
- Vanessa Osman
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065
| | - Iris Speigel
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065
| | - Kishan Patel
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065
| | - Hugh C Hemmings
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
18
|
Zhang S, Wang Y, Zhang S, Huang C, Ding Q, Xia J, Wu D, Gao W. Emerging Anesthetic Nanomedicines: Current State and Challenges. Int J Nanomedicine 2023; 18:3913-3935. [PMID: 37489141 PMCID: PMC10363368 DOI: 10.2147/ijn.s417855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/10/2023] [Indexed: 07/26/2023] Open
Abstract
Anesthetics, which include both local and general varieties, are a unique class of drugs widely utilized in clinical surgery to alleviate pain and promote relaxation in patients. Although numerous anesthetics and their traditional formulations are available in the market, only a select few exhibit excellent anesthetic properties that meet clinical requirements. The main challenges are the potential toxic and adverse effects of anesthetics, as well as the presence of the blood-brain barrier (BBB), which makes it difficult for most general anesthetics to effectively penetrate to the brain. Loading anesthetics onto nanocarriers as anesthetic nanomedicines might address these challenges and improve anesthesia effectiveness, reduce toxic and adverse effects, while significantly enhance the efficiency of general anesthetics passing through the BBB. Consequently, anesthetic nanomedicines play a crucial role in the field of anesthesia. Despite their significance, research on anesthetic nanomedicines is still in its infancy, especially when compared to other types of nanomedicines in terms of depth and breadth. Although local anesthetic nanomedicines have received considerable attention and essentially meet clinical needs, there are few reported instances of nanomedicines for general anesthetics. Given the extensive usage of anesthetics and the many of them need for improved performance, emerging anesthetic nanomedicines face both unparalleled opportunities and considerable challenges in terms of theory and technology. Thus, a comprehensive summary with systematic analyses of anesthetic nanomedicines is urgently required. This review provides a comprehensive summary of the classification, properties, and research status of anesthetic nanomedicines, along with an exploration of their opportunities and challenges. In addition, future research directions and development prospects are discussed. It is hoped that researchers from diverse disciplines will collaborate to study anesthetic nanomedicines and develop them as a valuable anesthetic dosage form for clinical surgery.
Collapse
Affiliation(s)
- Shuo Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Yishu Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Shuai Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Chengqi Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Qiyang Ding
- Department of Anesthesiology & Center for Brain Science & Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Ji Xia
- Department of Anesthesiology & Center for Brain Science & Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Daocheng Wu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Wei Gao
- Department of Anesthesiology & Center for Brain Science & Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| |
Collapse
|
19
|
Spencer KA, Woods CB, Worstman HM, Johnson SC, Ramirez JM, Morgan PG, Sedensky MM. TREK-1 and TREK-2 Knockout Mice Are Not Resistant to Halothane or Isoflurane. Anesthesiology 2023; 139:63-76. [PMID: 37027798 PMCID: PMC10247454 DOI: 10.1097/aln.0000000000004577] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
BACKGROUND A variety of molecular targets for volatile anesthetics have been suggested, including the anesthetic-sensitive potassium leak channel, TREK-1. Knockout of TREK-1 is reported to render mice resistant to volatile anesthetics, making TREK-1 channels compelling targets for anesthetic action. Spinal cord slices from mice, either wild type or an anesthetic- hypersensitive mutant, Ndufs4, display an isoflurane-induced outward potassium leak that correlates with their minimum alveolar concentrations and is blocked by norfluoxetine. The hypothesis was that TREK-1 channels conveyed this current and contribute to the anesthetic hypersensitivity of Ndufs4. The results led to evaluation of a second TREK channel, TREK-2, in control of anesthetic sensitivity. METHODS The anesthetic sensitivities of mice carrying knockout alleles of Trek-1 and Trek-2, the double knockout Trek-1;Trek-2, and Ndufs4;Trek-1 were measured. Neurons from spinal cord slices from each mutant were patch clamped to characterize isoflurane-sensitive currents. Norfluoxetine was used to identify TREK-dependent currents. RESULTS The mean values for minimum alveolar concentrations (± SD) between wild type and two Trek-1 knockout alleles in mice (P values, Trek-1 compared to wild type) were compared. For wild type, minimum alveolar concentration of halothane was 1.30% (0.10), and minimum alveolar concentration of isoflurane was 1.40% (0.11); for Trek-1tm1Lex, minimum alveolar concentration of halothane was 1.27% (0.11; P = 0.387), and minimum alveolar concentration of isoflurane was 1.38% (0.09; P = 0.268); and for Trek-1tm1Lzd, minimum alveolar concentration of halothane was 1.27% (0.11; P = 0.482), and minimum alveolar concentration of isoflurane was 1.41% (0.12; P = 0.188). Neither allele was resistant for loss of righting reflex. The EC50 values of Ndufs4;Trek-1tm1Lex did not differ from Ndufs4 (for Ndufs4, EC50 of halothane, 0.65% [0.05]; EC50 of isoflurane, 0.63% [0.05]; and for Ndufs4;Trek-1tm1Lex, EC50 of halothane, 0.58% [0.07; P = 0.004]; and EC50 of isoflurane, 0.61% [0.06; P = 0.442]). Loss of TREK-2 did not alter anesthetic sensitivity in a wild-type or Trek-1 genetic background. Loss of TREK-1, TREK-2, or both did not alter the isoflurane-induced currents in wild-type cells but did cause them to be norfluoxetine insensitive. CONCLUSIONS Loss of TREK channels did not alter anesthetic sensitivity in mice, nor did it eliminate isoflurane-induced transmembrane currents. However, the isoflurane-induced currents are norfluoxetine-resistant in Trek mutants, indicating that other channels may function in this role when TREK channels are deleted. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Kira A Spencer
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle WA, 98105, USA
| | - Christian B Woods
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101, USA
| | - Hailey M Worstman
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101, USA
| | - Simon C Johnson
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101, USA
- Applied Sciences, Translational Biosciences, Northumbria University, Ellison A521A, UK (current)
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, 98105, USA
| | - Philip G Morgan
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle WA, 98105, USA
| | - Margaret M Sedensky
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle WA, 98105, USA
| |
Collapse
|
20
|
Tauber JM, Brincat SL, Stephen EP, Donaghue JA, Kozachkov L, Brown EN, Miller EK. Propofol Mediated Unconsciousness Disrupts Progression of Sensory Signals through the Cortical Hierarchy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.25.546463. [PMID: 37425684 PMCID: PMC10327085 DOI: 10.1101/2023.06.25.546463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
A critical component of anesthesia is the loss sensory perception. Propofol is the most widely used drug for general anesthesia, but the neural mechanisms of how and when it disrupts sensory processing are not fully understood. We analyzed local field potential (LFP) and spiking recorded from Utah arrays in auditory cortex, associative cortex, and cognitive cortex of non-human primates before and during propofol mediated unconsciousness. Sensory stimuli elicited robust and decodable stimulus responses and triggered periods of stimulus-induced coherence between brain areas in the LFP of awake animals. By contrast, propofol mediated unconsciousness eliminated stimulus-induced coherence and drastically weakened stimulus responses and information in all brain areas except for auditory cortex, where responses and information persisted. However, we found stimuli occurring during spiking Up states triggered weaker spiking responses than in awake animals in auditory cortex, and little or no spiking responses in higher order areas. These results suggest that propofol's effect on sensory processing is not just due to asynchronous down states. Rather, both Down states and Up states reflect disrupted dynamics.
Collapse
Affiliation(s)
- John M. Tauber
- The Picower Institute for Learning & Memory, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Scott L. Brincat
- The Picower Institute for Learning & Memory, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Emily P. Stephen
- Department of Mathematics and Statistics, Boston University, Boston, MA 02215, USA
| | - Jacob A. Donaghue
- The Picower Institute for Learning & Memory, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Leo Kozachkov
- Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Emery N. Brown
- The Picower Institute for Learning & Memory, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Institute for Data, Systems, and Society, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Anesthesia, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Earl K. Miller
- The Picower Institute for Learning & Memory, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| |
Collapse
|
21
|
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
|
22
|
Lawn T, Martins D, O'Daly O, Williams S, Howard M, Dipasquale O. The effects of propofol anaesthesia on molecular-enriched networks during resting-state and naturalistic listening. Neuroimage 2023; 271:120018. [PMID: 36935083 PMCID: PMC10410200 DOI: 10.1016/j.neuroimage.2023.120018] [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: 10/31/2022] [Accepted: 03/09/2023] [Indexed: 03/19/2023] Open
Abstract
Placing a patient in a state of anaesthesia is crucial for modern surgical practice. However, the mechanisms by which anaesthetic drugs, such as propofol, impart their effects on consciousness remain poorly understood. Propofol potentiates GABAergic transmission, which purportedly has direct actions on cortex as well as indirect actions via ascending neuromodulatory systems. Functional imaging studies to date have been limited in their ability to unravel how these effects on neurotransmission impact the system-level dynamics of the brain. Here, we leveraged advances in multi-modal imaging, Receptor-Enriched Analysis of functional Connectivity by Targets (REACT), to investigate how different levels of propofol-induced sedation alter neurotransmission-related functional connectivity (FC), both at rest and when individuals are exposed to naturalistic auditory stimulation. Propofol increased GABA-A- and noradrenaline transporter-enriched FC within occipital and somatosensory regions respectively. Additionally, during auditory stimulation, the network related to the dopamine transporter showed reduced FC within bilateral regions of temporal and mid/posterior cingulate cortices, with the right temporal cluster showing an interaction between auditory stimulation and level of consciousness. In bringing together these micro- and macro-scale systems, we provide support for both direct GABAergic and indirect noradrenergic and dopaminergic-related network changes under propofol sedation. Further, we delineate a cognition-related reconfiguration of the dopaminergic network, highlighting the utility of REACT to explore the molecular substrates of consciousness and cognition.
Collapse
Affiliation(s)
- Timothy Lawn
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's college London, London, UK.
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's college London, London, UK
| | - Owen O'Daly
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's college London, London, UK
| | - Steve Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's college London, London, UK
| | - Matthew Howard
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's college London, London, UK
| | - Ottavia Dipasquale
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's college London, London, UK
| |
Collapse
|
23
|
Hu JJ, Liu Y, Yao H, Cao B, Liao H, Yang R, Chen P, Song XJ. Emergence of consciousness from anesthesia through ubiquitin degradation of KCC2 in the ventral posteromedial nucleus of the thalamus. Nat Neurosci 2023; 26:751-764. [PMID: 36973513 DOI: 10.1038/s41593-023-01290-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/23/2023] [Indexed: 03/29/2023]
Abstract
The emergence of consciousness from anesthesia, once assumed to be a passive process, is now considered as an active and controllable process. In the present study, we show in mice that, when the brain is forced into a minimum responsive state by diverse anesthetics, a rapid downregulation of K+/Cl- cotransporter 2 (KCC2) in the ventral posteromedial nucleus (VPM) serves as a common mechanism by which the brain regains consciousness. Ubiquitin-proteasomal degradation is responsible for KCC2 downregulation, which is driven by ubiquitin ligase Fbxl4. Phosphorylation of KCC2 at Thr1007 promotes interaction between KCC2 and Fbxl4. KCC2 downregulation leads to γ-aminobutyric acid type A receptor-mediated disinhibition, enabling accelerated recovery of VPM neuron excitability and emergence of consciousness from anesthetic inhibition. This pathway to recovery is an active process and occurs independent of anesthetic choice. The present study demonstrates that ubiquitin degradation of KCC2 in the VPM is an important intermediate step en route to emergence of consciousness from anesthesia.
Collapse
|
24
|
Koch S, Blankertz B, Windmann V, Spies C, Radtke FM, Röhr V. Desflurane is risk factor for postoperative delirium in older patients' independent from intraoperative burst suppression duration. Front Aging Neurosci 2023; 15:1067268. [PMID: 36819718 PMCID: PMC9929347 DOI: 10.3389/fnagi.2023.1067268] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/09/2023] [Indexed: 02/04/2023] Open
Abstract
Background Postoperative Delirium (POD) is the most frequent neurocognitive complication after general anesthesia in older patients. The development of POD is associated with prolonged periods of burst suppression activity in the intraoperative electroencephalogram (EEG). The risk to present burst suppression activity depends not only on the age of the patient but is also more frequent during propofol anesthesia as compared to inhalative anesthesia. The aim of our study is to determine, if the risk to develop POD differs depending on the anesthetic agent given and if this correlates with a longer duration of intraoperative burst suppression. Methods In this secondary analysis of the SuDoCo trail [ISRCTN 36437985] 1277 patients, older than 60 years undergoing general anesthesia were included. We preprocessed and analyzed the raw EEG files from each patient and evaluated the intraoperative burst suppression duration. In a logistic regression analysis, we assessed the impact of burst suppression duration and anesthetic agent used for maintenance on the risk to develop POD. Results 18.7% of patients developed POD. Burst suppression duration was prolonged in POD patients (POD 27.5 min ± 21.3 min vs. NoPOD 21.4 ± 16.2 min, p < 0.001), for each minute of prolonged intraoperative burst suppression activity the risk to develop POD increased by 1.1% (OR 1.011, CI 95% 1.000-1.022, p = 0.046). Burst suppression duration was prolonged under propofol anesthesia as compared to sevoflurane and desflurane anesthesia (propofol 32.5 ± 20.3 min, sevoflurane 17.1 ± 12.6 min and desflurane 20.1 ± 16.0 min, p < 0.001). However, patients receiving desflurane anesthesia had a 1.8fold higher risk to develop POD, as compared to propofol anesthesia (OR 1.766, CI 95% 1.049-2.974, p = 0.032). Conclusion We found a significantly increased risk to develop POD after desflurane anesthesia in older patients, even though burst suppression duration was shorter under desflurane anesthesia as compared to propofol anesthesia. Our finding might help to explain some discrepancies in studies analyzing the impact of burst suppression duration and EEG-guided anesthesia on the risk to develop POD.
Collapse
Affiliation(s)
- Susanne Koch
- Department of Anaesthesiology and Operative Intensive Care Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany,*Correspondence: Susanne Koch, ✉
| | | | - Victoria Windmann
- Department of Anaesthesiology and Operative Intensive Care Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Spies
- Department of Anaesthesiology and Operative Intensive Care Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Finn M. Radtke
- Department of Anesthesia, Hospital of Nykobing, University of Southern Denmark, Odense, Denmark
| | - Vera Röhr
- Neurotechnology Group, Technische Universität Berlin, Berlin, Germany
| |
Collapse
|
25
|
Mitzelfelt T, Bao X, Barnes P, Lomber SG. Visually evoked potentials (VEPs) across the visual field in hearing and deaf cats. Front Neurosci 2023; 17:997357. [PMID: 36937669 PMCID: PMC10020186 DOI: 10.3389/fnins.2023.997357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 01/24/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Congenitally deaf cats perform better on visual localization tasks than hearing cats, and this advantage has been attributed to the posterior auditory field. Successful visual localization requires both visual processing of the target and timely generation of an action to approach the target. Activation of auditory cortex in deaf subjects during visual localization in the peripheral visual field can occur either via bottom-up stimulus-driven and/or top-down goal-directed pathways. Methods In this study, we recorded visually evoked potentials (VEPs) in response to a reversing checkerboard stimulus presented in the hemifield contralateral to the recorded hemisphere in both hearing and deaf cats under light anesthesia. Results Although VEP amplitudes and latencies were systematically modulated by stimulus eccentricity, we found little evidence of changes in VEP in deaf cats that can explain their behavioral advantage. A statistical trend was observed, showing larger peak amplitudes and shorter peak latencies in deaf subjects for stimuli in the near- and mid-peripheral field. Additionally, latency of the P1 wave component had a larger inter-sweep variation in deaf subjects. Discussion Our results suggested that cross-modal plasticity following deafness does not play a major part in cortical processing of the peripheral visual field when the "vision for action" system is not recruited.
Collapse
Affiliation(s)
| | - Xiaohan Bao
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Paisley Barnes
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Stephen G. Lomber
- Department of Physiology, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- *Correspondence: Stephen G. Lomber,
| |
Collapse
|
26
|
Luo M, Fei X, Liu X, Jin Z, Wang Y, Xu M. Divergent Neural Activity in the VLPO During Anesthesia and Sleep. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2203395. [PMID: 36461756 PMCID: PMC9839870 DOI: 10.1002/advs.202203395] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/10/2022] [Indexed: 05/27/2023]
Abstract
The invention of general anesthesia (GA) represents a significant advance in modern clinical practices. However, the exact mechanisms of GA are not entirely understood. Because of the multitude of similarities between GA and sleep, one intriguing hypothesis is that anesthesia may engage the sleep-wake regulation circuits. Here, using fiber photometry and micro-endoscopic imaging of Ca2+ signals at both population and single-cell levels, it investigates how various anesthetics modulate the neural activity in the ventrolateral preoptic nucleus (vLPO), a brain region essential for the initiation of sleep. It is found that different anesthetics primarily induced suppression of neural activity and tended to recruit a similar group of vLPO neurons; however, each anesthetic caused comparable modulations of both wake-active and sleep-active neurons. These results demonstrate that anesthesia creates a different state of neural activity in the vLPO than during natural sleep, suggesting that anesthesia may not engage the same vLPO circuits for sleep generation.
Collapse
Affiliation(s)
- Mengqiang Luo
- Department of AnesthesiologyHuashan HospitalFudan UniversityShanghai200040China
| | - Xiang Fei
- Institute of NeuroscienceState Key Laboratory of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
| | - Xiaotong Liu
- Institute of NeuroscienceState Key Laboratory of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
| | - Zikang Jin
- Institute of NeuroscienceState Key Laboratory of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
| | - Yingwei Wang
- Department of AnesthesiologyHuashan HospitalFudan UniversityShanghai200040China
| | - Min Xu
- Institute of NeuroscienceState Key Laboratory of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
- Shanghai Center for Brain Science and Brain‐Inspired Intelligence TechnologyShanghai201210China
| |
Collapse
|
27
|
Vedaei F, Alizadeh M, Tantawi M, Romo V, Mohamed FB, Wu C. Vascular and neuronal effects of general anesthesia on the brain: An fMRI study. J Neuroimaging 2023; 33:109-120. [PMID: 36097249 DOI: 10.1111/jon.13049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND AND PURPOSE A number of functional magnetic resonance imaging (fMRI) studies rely on application of anesthetic agents during scanning that can modulate and complicate interpretation of the measured hemodynamic blood oxygenation level-dependent (BOLD) response. The purpose of the present study was to investigate the effect of general anesthesia on two main components of BOLD signal including neuronal activity and vascular response. METHODS Breath-holding (BH) fMRI was conducted in wakefulness and under anesthesia states in 9 patients with drug-resistant epilepsy who needed to get scanned under anesthesia during laser interstitial thermal therapy. BOLD and BOLD cerebrovascular reactivity (BOLD-CVR) maps were compared using t-test between two states to assess the effect of anesthesia on neuronal activity and vascular factors (p < .05). RESULTS Overall, our findings revealed an increase in BOLD-CVR and decrease in BOLD response under anesthesia in several brain regions. The results proposed that the modulatory mechanism of anesthetics on neuronal and vascular components of BOLD signal may work in different ways. CONCLUSION This experiment for the first human study showed that anesthesia may play an important role in dissociation between neuronal and vascular responses contributed to hemodynamic BOLD signal using BH fMRI imaging that may assist the implication of general anesthesia and interpretation of outcomes in clinical setting.
Collapse
Affiliation(s)
- Faezeh Vedaei
- Jefferson Integrated Magnetic Resonance Imaging Center, Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Mahdi Alizadeh
- Jefferson Integrated Magnetic Resonance Imaging Center, Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Mohamed Tantawi
- Jefferson Integrated Magnetic Resonance Imaging Center, Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Victor Romo
- Department of Anesthesiology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Feroze B Mohamed
- Jefferson Integrated Magnetic Resonance Imaging Center, Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Chengyuan Wu
- Jefferson Integrated Magnetic Resonance Imaging Center, Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
28
|
Zhang M, Chen Y, Liu J, Yang Y, Wang R, Zhang D, Zhu T. Development of NMDA receptors contributes to the enhancement of electroencephalogram oscillations under volatile anesthetics in rats. Front Neural Circuits 2022; 16:1065374. [PMID: 36589861 PMCID: PMC9797678 DOI: 10.3389/fncir.2022.1065374] [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: 10/09/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Background Volatile anesthetics including sevoflurane and isoflurane enhance oscillations of cortical electroencephalogram (EEG), partly by their modulations on glutamate-mediated excitatory synaptic transmission. Expression of NMDA receptors is increased during neonatal development. However, how the development of NMDA receptors influences EEG under volatile anesthesia remains unclear. Methods Expressions of NMDA receptor subtypes (NR1, NR2A, and NR2B) during neonatal development were measured by Western blotting. MAC (minimal alveolar concentration) of isoflurane and sevoflurane that inducing loss of righting reflex (LORR) and no response to tail-clamp (immobility) were measured to verify the effect of NR1 expression on anesthetic potency during neonatal development. Cortical electroencephalogram recording was used to examine the influence of NR1 expression on the power density of EEG. Results The expressions of GluNR1, GluNR2A and GluNR2B receptors were gradually increased during neonatal development in cortex, hippocampus and thalamus of rats. Knockdown of NR1 enhanced the sedative potency of volatile anesthetics but not on immobility potency in postnatal day 14 (P14)-P17 rats. For cortical EEG, along with the increased concentration of volatile anesthetics, cortical slow-delta oscillations of P5 rats were inhibited, theta and alpha oscillations were not changed significantly; while these oscillations were enhanced until high anesthetic concentrations in P21 rats. Knockdown of NR1 in forebrain suppressed the enhancement of cortical EEG oscillations in P21 rats. Conclusion The development of NMDA receptors may contribute to the enhancement of cortical EEG oscillations under volatile anesthetics.
Collapse
Affiliation(s)
- Mingyue Zhang
- 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
| | - Yali Chen
- 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
- 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
| | - Rurong Wang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Donghang Zhang
- 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,*Correspondence: Donghang Zhang,
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China,Tao Zhu,
| |
Collapse
|
29
|
Jung J, Kim T. General anesthesia and sleep: like and unlike. Anesth Pain Med (Seoul) 2022; 17:343-351. [DOI: 10.17085/apm.22227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
General anesthesia and sleep have long been discussed in the neurobiological context owingto their commonalities, such as unconsciousness, immobility, non-responsiveness to externalstimuli, and lack of memory upon returning to consciousness. Sleep is regulated bycomplex interactions between wake-promoting and sleep-promoting neural circuits. Anestheticsexert their effects partly by inhibiting wake-promoting neurons or activating sleep-promotingneurons. Unconscious but arousable sedation is more related to sleep-wake circuitries,whereas unconscious and unarousable anesthesia is independent of them. Generalanesthesia is notable for its ability to decrease sleep propensity. Conversely, increasedsleep propensity due to insufficient sleep potentiates anesthetic effects. Taken together, it isplausible that sleep and anesthesia are closely related phenomena but not the same ones.Further investigations on the relationship between sleep and anesthesia are warranted.
Collapse
|
30
|
Li Z, Zhang H, Wang Y, Li Y, Li Q, Zhang L. The distinctive role of menthol in pain and analgesia: Mechanisms, practices, and advances. Front Mol Neurosci 2022; 15:1006908. [PMID: 36277488 PMCID: PMC9580369 DOI: 10.3389/fnmol.2022.1006908] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Menthol is an important flavoring additive that triggers a cooling sensation. Under physiological condition, low to moderate concentrations of menthol activate transient receptor potential cation channel subfamily M member 8 (TRPM8) in the primary nociceptors, such as dorsal root ganglion (DRG) and trigeminal ganglion, generating a cooling sensation, whereas menthol at higher concentration could induce cold allodynia, and cold hyperalgesia mediated by TRPM8 sensitization. In addition, the paradoxical irritating properties of high concentrations of menthol is associated with its activation of transient receptor potential cation channel subfamily A member 1 (TRPA1). Under pathological situation, menthol activates TRPM8 to attenuate mechanical allodynia and thermal hyperalgesia following nerve injury or chemical stimuli. Recent reports have recapitulated the requirement of central group II/III metabotropic glutamate receptors (mGluR) with endogenous κ-opioid signaling pathways for menthol analgesia. Additionally, blockage of sodium channels and calcium influx is a determinant step after menthol exposure, suggesting the possibility of menthol for pain management. In this review, we will also discuss and summarize the advances in menthol-related drugs for pathological pain treatment in clinical trials, especially in neuropathic pain, musculoskeletal pain, cancer pain and postoperative pain, with the aim to find the promising therapeutic candidates for the resolution of pain to better manage patients with pain in clinics.
Collapse
Affiliation(s)
- Ziping Li
- The Graduate School, Tianjin Medical University, Tianjin, China
| | - Haoyue Zhang
- The Graduate School, Tianjin Medical University, Tianjin, China
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yigang Wang
- The Graduate School, Tianjin Medical University, Tianjin, China
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yize Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qing Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Qing Li,
| | - Linlin Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Linlin Zhang,
| |
Collapse
|
31
|
Vedaei F, Alizadeh M, Romo V, Mohamed FB, Wu C. The effect of general anesthesia on the test–retest reliability of resting-state fMRI metrics and optimization of scan length. Front Neurosci 2022; 16:937172. [PMID: 36051647 PMCID: PMC9425911 DOI: 10.3389/fnins.2022.937172] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/27/2022] [Indexed: 01/01/2023] Open
Abstract
Resting-state functional magnetic resonance imaging (rs-fMRI) has been known as a powerful tool in neuroscience. However, exploring the test–retest reliability of the metrics derived from the rs-fMRI BOLD signal is essential, particularly in the studies of patients with neurological disorders. Here, two factors, namely, the effect of anesthesia and scan length, have been estimated on the reliability of rs-fMRI measurements. A total of nine patients with drug-resistant epilepsy (DRE) requiring interstitial thermal therapy (LITT) were scanned in two states. The first scan was performed in an awake state before surgery on the same patient. The second scan was performed 2 weeks later under general anesthesia necessary for LITT surgery. At each state, two rs-fMRI sessions were obtained that each one lasted 15 min, and the effect of scan length was evaluated. Voxel-wise rs-fMRI metrics, including the amplitude of low-frequency fluctuation (ALFF), the fractional amplitude of low-frequency fluctuation (fALFF), functional connectivity (FC), and regional homogeneity (ReHo), were measured. Intraclass correlation coefficient (ICC) was calculated to estimate the reliability of the measurements in two states of awake and under anesthesia. Overall, it appeared that the reliability of rs-fMRI metrics improved under anesthesia. From the 15-min data, we found mean ICC values in awake state including 0.81, 0.51, 0.65, and 0.84 for ALFF, fALFF, FC, and ReHo, respectively, as well as 0.80, 0.59, 0.83, and 0.88 for ALFF, fALFF, FC, and ReHo, respectively, under anesthesia. Additionally, our findings revealed that reliability increases as the function of scan length. We showed that the optimized scan length to achieve less variability of rs-fMRI measurements was 3.1–7.5 min shorter in an anesthetized, compared to a wakeful state.
Collapse
Affiliation(s)
- Faezeh Vedaei
- Department of Radiology, Jefferson Integrated Magnetic Resonance Imaging Center, Thomas Jefferson University, Philadelphia, PA, United States
- *Correspondence: Faezeh Vedaei
| | - Mahdi Alizadeh
- Department of Radiology, Jefferson Integrated Magnetic Resonance Imaging Center, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | - Victor Romo
- Department of Anesthesiology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Feroze B. Mohamed
- Department of Radiology, Jefferson Integrated Magnetic Resonance Imaging Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Chengyuan Wu
- Department of Radiology, Jefferson Integrated Magnetic Resonance Imaging Center, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
32
|
Jiang J, Zhao Y, Liu J, Yang Y, Liang P, Huang H, Wu Y, Kang Y, Zhu T, Zhou C. Signatures of Thalamocortical Alpha Oscillations and Synchronization With Increased Anesthetic Depths Under Isoflurane. Front Pharmacol 2022; 13:887981. [PMID: 35721144 PMCID: PMC9204038 DOI: 10.3389/fphar.2022.887981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Electroencephalography (EEG) recordings under propofol exhibit an increase in slow and alpha oscillation power and dose-dependent phase–amplitude coupling (PAC), which underlie GABAA potentiation and the central role of thalamocortical entrainment. However, the exact EEG signatures elicited by volatile anesthetics and the possible neurophysiological mechanisms remain unclear.Methods: Cortical EEG signals and thalamic local field potential (LFP) were recorded in a mouse model to detect EEG signatures induced by 0.9%, 1.5%, and 2.0% isoflurane. Then, the power of the EEG spectrum, thalamocortical coherence, and slow–alpha phase–amplitude coupling were analyzed. A computational model based on the thalamic network was used to determine the primary neurophysiological mechanisms of alpha spiking of thalamocortical neurons under isoflurane anesthesia.Results: Isoflurane at 0.9% (light anesthesia) increased the power of slow and delta oscillations both in cortical EEG and in thalamic LFP. Isoflurane at 1.5% (surgery anesthesia) increased the power of alpha oscillations both in cortical EEG and in thalamic LFP. Isoflurane at 2% (deep anesthesia) further increased the power of cortical alpha oscillations, while thalamic alpha oscillations were unchanged. Thalamocortical coherence of alpha oscillation only exhibited a significant increase under 1.5% isoflurane. Isoflurane-induced PAC modulation remained unchanged throughout under various concentrations of isoflurane. By adjusting the parameters in the computational model, isoflurane-induced alpha spiking in thalamocortical neurons was simulated, which revealed the potential molecular targets and the thalamic network involved in isoflurane-induced alpha spiking in thalamocortical neurons.Conclusion: The EEG changes in the cortical alpha oscillation, thalamocortical coherence, and slow–alpha PAC may provide neurophysiological signatures for monitoring isoflurane anesthesia at various depths.
Collapse
Affiliation(s)
- Jingyao Jiang
- 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
| | - Yi Zhao
- 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
- 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
| | - Peng Liang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Han Huang
- Department of Anesthesiology, West China Second Hospital of Sichuan University, Chengdu, China
| | - Yongkang Wu
- Intelligent Manufacturing Institute, Chengdu Jincheng College, Chengdu, China
| | - Yi Kang
- 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
- *Correspondence: Tao Zhu, ; Cheng Zhou,
| | - 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
- *Correspondence: Tao Zhu, ; Cheng Zhou,
| |
Collapse
|
33
|
Hasselager RP, Madsen SS, Møller K, Gögenur I, Asghar MS. Effect of sevoflurane versus propofol on neutrophil-to-lymphocyte ratio in healthy individuals: a sub-study of a randomised crossover trial. BJA OPEN 2022; 2:100005. [PMID: 37588265 PMCID: PMC10430840 DOI: 10.1016/j.bjao.2022.100005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 03/07/2022] [Indexed: 08/18/2023]
Abstract
Background Sevoflurane and propofol are commonly used drugs in general anaesthesia. However, their effects on perioperative immune function are incompletely understood. We hypothesised that sevoflurane and propofol differentially affect immune function in healthy individuals. Therefore, we investigated the effect of sevoflurane and propofol on neutrophil-to-lymphocyte ratio before, during, and after general anaesthesia. Methods In this randomised crossover study, 19 healthy individuals underwent 2 h of general anaesthesia with either propofol or sevoflurane. After 4 weeks, anaesthesia was repeated using the other drug. Blood samples were obtained before, during, 1 h after, and 1 day after anaesthesia. The primary outcome was whole-blood neutrophil-to-lymphocyte ratio, and secondary outcomes were specific white blood cell differential counts. A linear mixed-effects model was used to estimate effect sizes. Results The neutrophil-to-lymphocyte ratio was higher in the propofol compared with the sevoflurane group during anaesthesia, 2.8 (confidence interval [CI]: 2.3-3.3) vs 1.6 (CI: 1.1-2.1), and 1 day after anaesthesia, 2.6 (CI: 2.1-3.1) vs 1.9 (CI: 1.4-2.4). In all patients, we observed transient lymphopaenia during propofol anaesthesia, 1.1 × 109 cells × L-1 (CI: 0.9-1.4), compared with sevoflurane anaesthesia, 1.9 × 109 cells × L-1 (CI: 1.7-2.1). In addition, neutrophil counts were higher 1 day after propofol anaesthesia, 4.4 × 109 cells × L-1 (CI: 4.0-4.9), compared with sevoflurane anaesthesia, 3.5 × 109 cells × L-1 (CI: 3.1-4.0). We observed no differences in the remaining white blood cell subgroups. Conclusions In healthy individuals undergoing general anaesthesia without surgery, the neutrophil-to-lymphocyte ratio was affected by the type of hypnotic used. Transient lymphopaenia was observed in all participants during propofol anaesthesia.
Collapse
Affiliation(s)
| | - Signe Sloth Madsen
- Department of Neuroanaesthesiology, Neuroscience Centre, Rigshospitalet, Copenhagen, Denmark
| | - Kirsten Møller
- Department of Neuroanaesthesiology, Neuroscience Centre, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Roskilde, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
34
|
Shimizu M, Mi X, Toyoda F, Kojima A, Ding WG, Fukushima Y, Omatsu-Kanbe M, Kitagawa H, Matsuura H. Propofol, an Anesthetic Agent, Inhibits HCN Channels through the Allosteric Modulation of the cAMP-Dependent Gating Mechanism. Biomolecules 2022; 12:biom12040570. [PMID: 35454159 PMCID: PMC9032835 DOI: 10.3390/biom12040570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 11/16/2022] Open
Abstract
Propofol is a broadly used intravenous anesthetic agent that can cause cardiovascular effects, including bradycardia and asystole. A possible mechanism for these effects is slowing cardiac pacemaker activity due to inhibition of the hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels. However, it remains unclear how propofol affects the allosteric nature of the voltage- and cAMP-dependent gating mechanism in HCN channels. To address this aim, we investigated the effect of propofol on HCN channels (HCN4 and HCN2) in heterologous expression systems using a whole-cell patch clamp technique. The extracellular application of propofol substantially suppressed the maximum current at clinical concentrations. This was accompanied by a hyperpolarizing shift in the voltage dependence of channel opening. These effects were significantly attenuated by intracellular loading of cAMP, even after considering the current modification by cAMP in opposite directions. The differential degree of propofol effects in the presence and absence of cAMP was rationalized by an allosteric gating model for HCN channels, where we assumed that propofol affects allosteric couplings between the pore, voltage-sensor, and cyclic nucleotide-binding domain (CNBD). The model predicted that propofol enhanced autoinhibition of pore opening by unliganded CNBD, which was relieved by the activation of CNBD by cAMP. Taken together, these findings reveal that propofol acts as an allosteric modulator of cAMP-dependent gating in HCN channels, which may help us to better understand the clinical action of this anesthetic drug.
Collapse
Affiliation(s)
- Morihiro Shimizu
- Department of Anesthesiology, Shiga University of Medical Science, Otsu 520-2192, Japan; (M.S.); (A.K.); (Y.F.); (H.K.)
| | - Xinya Mi
- Department of Physiology, Shiga University of Medical Science, Otsu 520-2192, Japan; (X.M.); (F.T.); (M.O.-K.); (H.M.)
| | - Futoshi Toyoda
- Department of Physiology, Shiga University of Medical Science, Otsu 520-2192, Japan; (X.M.); (F.T.); (M.O.-K.); (H.M.)
| | - Akiko Kojima
- Department of Anesthesiology, Shiga University of Medical Science, Otsu 520-2192, Japan; (M.S.); (A.K.); (Y.F.); (H.K.)
| | - Wei-Guang Ding
- Department of Physiology, Shiga University of Medical Science, Otsu 520-2192, Japan; (X.M.); (F.T.); (M.O.-K.); (H.M.)
- Correspondence: ; Tel.: +81-77-548-2152; Fax: +81-77-548-2348
| | - Yutaka Fukushima
- Department of Anesthesiology, Shiga University of Medical Science, Otsu 520-2192, Japan; (M.S.); (A.K.); (Y.F.); (H.K.)
| | - Mariko Omatsu-Kanbe
- Department of Physiology, Shiga University of Medical Science, Otsu 520-2192, Japan; (X.M.); (F.T.); (M.O.-K.); (H.M.)
| | - Hirotoshi Kitagawa
- Department of Anesthesiology, Shiga University of Medical Science, Otsu 520-2192, Japan; (M.S.); (A.K.); (Y.F.); (H.K.)
| | - Hiroshi Matsuura
- Department of Physiology, Shiga University of Medical Science, Otsu 520-2192, Japan; (X.M.); (F.T.); (M.O.-K.); (H.M.)
| |
Collapse
|
35
|
Wang Q, Li Y, Tan H, Wang Y. Sevoflurane-Induced Apoptosis in the Mouse Cerebral Cortex Follows Similar Characteristics of Physiological Apoptosis. Front Mol Neurosci 2022; 15:873658. [PMID: 35465098 PMCID: PMC9024292 DOI: 10.3389/fnmol.2022.873658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
General anesthetics are capable of inducing neuronal apoptosis during the rapid synaptogenesis of immature mammalian brains. In this vulnerable time window, physiological apoptosis also occurs to eliminate excess and inappropriately integrated neurons. We previously showed that physiological and ketamine-induced apoptosis in mouse primary somatosensory cortex (S1) followed similar developmental patterns. However, since sevoflurane is more widely used in pediatric anesthesia, and targets mainly on different receptors, as compared with ketamine, it is important to determine whether sevoflurane-induced apoptosis also follows similar developmental patterns as physiological apoptosis or not. Mice at postnatal days 5 (P5) and P9 were anesthetized with 1.5% sevoflurane for 4 h, and the apoptotic neurons in S1 were quantitated by immunohistochemistry. The results showed that sevoflurane raised the levels of apoptosis in S1 without interfering with the developmental patterns of physiological apoptosis. The cells more vulnerable to both physiological and sevoflurane-induced apoptosis shifted from layer V pyramidal neurons at P5 to layers II–IV GABAergic neurons by P9. The magnitude of both sevoflurane-induced and physiological apoptosis was more attenuated at P9 than P5. To determine whether the Akt-FoxO1-PUMA pathway contributes to the developmental decrease in magnitude of both physiological and sevoflurane-induced apoptosis, Western blot was used to measure the levels of related proteins in S1 of P5 and P9 mice. We observed higher levels of antiapoptotic phosphorylated Akt (p-Akt) and phosphorylated FoxO1 (p-FoxO1), and lower levels of the downstream proapoptotic factor PUMA in control and anesthetized mice at P9 than P5. In addition, the Akt-FoxO1-PUMA pathway may also be responsible for sevoflurane-induced apoptosis. Together, these results suggest that magnitude, lamination pattern and cell-type specificity to sevoflurane-induced apoptosis are age-dependent and follow physiological apoptosis pattern. Moreover, The Akt-FoxO1-PUMA pathway may mediate the developmental decreases in magnitude of both physiological and sevoflurane-induced apoptosis in neonatal mouse S1.
Collapse
|
36
|
Oda R, Shou J, Zhong W, Ozeki Y, Yasui M, Nuriya M. Direct visualization of general anesthetic propofol on neurons by stimulated Raman scattering microscopy. iScience 2022; 25:103936. [PMID: 35252821 PMCID: PMC8894261 DOI: 10.1016/j.isci.2022.103936] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/28/2022] [Accepted: 02/14/2022] [Indexed: 11/29/2022] Open
Abstract
The consensus for the precise mechanism of action of general anesthetics is through allosteric interactions with GABA receptors in neurons. However, it has been speculated that these anesthetics may also interact with the plasma membrane on some level. Owing to the small size of anesthetics, direct visualization of these interactions is difficult to achieve. We demonstrate the ability to directly visualize a deuterated analog of propofol in living cells using stimulated Raman scattering (SRS) microscopy. Our findings support the theory that propofol is highly concentrated and interacts primarily through non-specific binding to the plasma membrane of neurons. Additionally, we show that SRS microscopy can be used to monitor the dynamics of propofol binding using real-time, live-cell imaging. The strategy used to visualize propofol can be applied to other small molecule drugs that have been previously invisible to traditional imaging techniques Multi-modal SRS developed for real-time biological imaging of small molecule substances Propofol primarily concentrates at the cell membrane of neurons Anesthesia dynamics can be monitored in real-time with SRS
Collapse
Affiliation(s)
- Robert Oda
- Department of Pharmacology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
- Department of Electrical Engineering and Information Systems, Graduate School of Engineering, 7-3-1 Hongo, Bunkyo, Tokyo 113-8656, Japan
- Corresponding author
| | - Jingwen Shou
- Department of Electrical Engineering and Information Systems, Graduate School of Engineering, 7-3-1 Hongo, Bunkyo, Tokyo 113-8656, Japan
| | - Wenying Zhong
- Department of Pharmacology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Yasuyuki Ozeki
- Department of Electrical Engineering and Information Systems, Graduate School of Engineering, 7-3-1 Hongo, Bunkyo, Tokyo 113-8656, Japan
| | - Masato Yasui
- Department of Pharmacology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Mutsuo Nuriya
- Department of Pharmacology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
- Graduate School of Environment and Information Sciences, Yokohama National University, 79-1 Tokiwadai, Hodogaya, Yokohama, Kanagawa 240-8501, Japan
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
- Corresponding author
| |
Collapse
|
37
|
Uchida Y, Hashimoto T, Saito H, Takita K, Morimoto Y. Neonatal isoflurane exposure disturbs granule cell migration in the rat dentate gyrus. Biomed Res 2022; 43:1-9. [PMID: 35173111 DOI: 10.2220/biomedres.43.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
It has been reported that neonatal isoflurane exposure causes behavioral abnormalities following neurodegeneration in animals and gamma-aminobutyric acid type A (GABAA) receptor activation during the synaptogenesis is considered to be one possible trigger. Additionally, the inhibitory effect of excitatory GABAA receptor signaling on the granule cell (GC) migration in the neonatal rat dentate gyrus (DG) was reported in a febrile seizure model. Then, we hypothesized that neonatal isoflurane exposure, which activates GABAA receptor, causes GC migration disturbances in the neonatal rat. Rat pups were injected with 5-bromo-2'-deoxyuridine (BrdU) and divided into five treatment groups, and double immunofluorescent staining targeting BrdU and homeobox prospero-like protein 1 (Prox1) was performed to examine the localization of BrdU/Prox1 colabeled cells, and then the GC migration was assessed. As a result, we found that the ectopic migration of GC after 2% isoflurane exposure on postnatal day 7 significantly increased after P21. The number of hilar ectopic GCs was influenced by the concentration of isoflurane and the exposure day but not by carbon dioxide exposure. Our main finding is that neonatal isoflurane anesthesia disturbs the migration of GCs in the rat DG, which may be one possible mechanism underlying the neurotoxicity following neonatal isoflurane anesthesia.
Collapse
Affiliation(s)
- Yosuke Uchida
- Department of Anesthesiology, Hokkaido University Hospital
| | | | - Hitoshi Saito
- Department of Anesthesiology, Hokkaido University Hospital
| | - Koichi Takita
- Department of Anesthesiology, Hokkaido University Hospital
| | - Yuji Morimoto
- Department of Anesthesiology, Hokkaido University Hospital
| |
Collapse
|
38
|
Wang C, Liu S, Liu F, Bhutta A, Patterson TA, Slikker W. Application of Nonhuman Primate Models in the Studies of Pediatric Anesthesia Neurotoxicity. Anesth Analg 2022; 134:1203-1214. [PMID: 35147575 DOI: 10.1213/ane.0000000000005926] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Numerous animal models have been used to study developmental neurotoxicity associated with short-term or prolonged exposure of common general anesthetics at clinically relevant concentrations. Pediatric anesthesia models using the nonhuman primate (NHP) may more accurately reflect the human condition because of their phylogenetic similarity to humans with regard to reproduction, development, neuroanatomy, and cognition. Although they are not as widely used as other animal models, the contribution of NHP models in the study of anesthetic-induced developmental neurotoxicity has been essential. In this review, we discuss how neonatal NHP animals have been used for modeling pediatric anesthetic exposure; how NHPs have addressed key data gaps and application of the NHP model for the studies of general anesthetic-induced developmental neurotoxicity. The appropriate application and evaluation of the NHP model in the study of general anesthetic-induced developmental neurotoxicity have played a key role in enhancing the understanding and awareness of the potential neurotoxicity associated with pediatric general anesthetics.
Collapse
Affiliation(s)
- Cheng Wang
- From the Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, Arkansas
| | - Shuliang Liu
- From the Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, Arkansas
| | - Fang Liu
- From the Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, Arkansas
| | - Adnan Bhutta
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Tucker A Patterson
- Office of the Director, National Center for Toxicological Research/FDA, Jefferson, Arkansas
| | - William Slikker
- Office of the Director, National Center for Toxicological Research/FDA, Jefferson, Arkansas
| |
Collapse
|
39
|
Zhou C, Chen X, Zhang X. Progress in Neuropharmacology of Anesthetics and Analgesics for the
Improvement of Medical Treatment. Curr Neuropharmacol 2022; 20:3. [PMID: 35060453 PMCID: PMC9199551 DOI: 10.2174/1570159x20666220105121554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Affiliation(s)
- Cheng Zhou
- West China Hospital
Sichuan University
Chengdu, 610041
China
| | - Xiangdong Chen
- Union Hospital, Tongji Medical College
Huazhong University of Science and Technology
Wuhan, 430022
China
| | - Xia Zhang
- Institute of Neuropsychiatric Diseases
Qingdao University
Qingdao, 266001
China
| |
Collapse
|
40
|
Procópio IM, Pereira-Sampaio MA, Costa WS, Sampaio FJB, Souza DBD. Histomorphometric comparison of the corpus cavernosum of rats submitted to euthanasia with ketamine and xylazine or isoflurane. Acta Cir Bras 2021; 36:e361103. [PMID: 34909838 PMCID: PMC8664359 DOI: 10.1590/acb361103] [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: 07/12/2021] [Accepted: 10/09/2021] [Indexed: 11/22/2022] Open
Abstract
PURPOSE To compare the penile histoarchitecture of rats euthanized with isoflurane or with ketamine and xylazine. METHODS Fourteen male rats were divided into two groups: ISO, with animals euthanized with isoflurane; and K+X, with animals euthanized with ketamine (150 mg/kg) associated with xylazine (15 mg/kg). Immediately after the death, the penises were dissected, fixed in a 4% buffered formalin solution, and processed for histomorphometric analysis. The surface densities (Sv) of the corpus cavernosum structures (connective tissue, smooth muscle, sinusoidal space, and elastic fibers) were evaluated using Image J software. The distribution of collagen types I and III was qualitatively assessed. Statistical analyses were performed using the Student's t test for data comparison, considering it statistically significant when p < 0.05. RESULTS Regarding the Sv of connective tissue, smooth muscle and sinusoidal space, there were no differences between animals in both groups. On the other hand, the animals euthanized with the association of ketamine and xylazine showed the Sv of elastic fibers 24.8% higher in relation to animals euthanized with isoflurane. CONCLUSIONS The euthanasia method affected one of the morphological parameters of the rat penises. The choice of euthanasia method must be standardized to reduce bias and to obtain reliable and reproducible results.
Collapse
|
41
|
Zhang L, Fang K, Tao S, Deng L, Li H, Cao Y, Wang L, Yu F, Gu E. Electroencephalography-demonstrated mechanisms of dexmedetomidine-mediated deepening of propofol anesthesia: an observational study. Perioper Med (Lond) 2021; 10:44. [PMID: 34879867 PMCID: PMC8656083 DOI: 10.1186/s13741-021-00213-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 08/08/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although dexmedetomidine (Dex) is known to reduce bispectral index (BIS) values and propofol dosage, there is little information regarding raw electroencephalography (EEG) changes related to Dex deepening of propofol general anesthesia (GA). This study investigated the Dex effects on propofol GA via analysis of EEG changes. METHODS A study cohort of 21 surgical patients (age range, 20-60 years) categorized as American Society of Anesthesiologists (ASA) class I or II was enrolled. We used time-varying spectral and bicoherence methods to compare electroencephalogram signatures 5 min before versus 10 min after intravenous Dex injection under propofol GA. The means and medians are reported with 95% confidence intervals (CIs) and inter-quartile ranges (IQRs), respectively. RESULTS Dex augmented the slow waves power and theta (θ) oscillation bicoherence peak from a mean (95% CI) of 22.1% (19.0, 25.2) to 25.2% (21.8, 28.6). Meanwhile, Dex reduced alpha (α) peak power and bicoherence from 3.5 dB (1.0, 6.0) and 41.5% (34.0, 49.0) to 1.7 dB (- 0.6, 4.0) and 35.4% (29.0, 41.8), respectively, while diminishing the median frequency of α oscillation peak values and the mean frequency of α peaks in bicoherence spectra from 12.0 Hz (IQR 11.2, 12.6) and 11.7 Hz (11.3, 12.2) to 11.1 Hz (IQR 10.3, 11.8) and 11.2 Hz (10.9, 11.6), respectively. CONCLUSIONS Profound EEG changes support the supposition that Dex enhances propofol-induced GA from a moderate to a deeper state. The present findings provide a theoretical basis and reference regarding protocols aimed at reducing anesthetic/sedative dosage while maintaining sufficient depth of GA. CLINICAL TRIAL REGISTRATION ChiCTR, ChiCTR1900026955 . Registered on 27 October 2019.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Kun Fang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China.,First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Shengwei Tao
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China.,First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Liyun Deng
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China.,First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Hua Li
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China.,First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Yuanyuan Cao
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Lei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
| | - Fengqiong Yu
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Erwei Gu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
42
|
Anesthesia-Induced Oxidative Stress: Are There Differences between Intravenous and Inhaled Anesthetics? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8782387. [PMID: 34873432 PMCID: PMC8643269 DOI: 10.1155/2021/8782387] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/11/2021] [Accepted: 11/12/2021] [Indexed: 12/11/2022]
Abstract
Agents used for the induction of anesthesia have been shown to either promote or mitigate oxidative stress. A fine balance between the presence of reactive oxygen species (ROS) and antioxidants is crucial for the proper normal functioning of the cell. A basal concentration of ROS is essential for the manifestation of cellular functions, whereas disproportionate levels of ROS cause damage to cellular macromolecules such as DNA, lipids, and proteins, eventually leading to necrosis and apoptosis. Increased ROS has been linked with numerous illnesses, such as cardiovascular, immune system, liver, and kidney, and has been shown to promote cancer and accelerate aging. Knowledge of the various pharmacologic agents that increase or reduce oxidative stress may promote a safer way of inducing anesthesia. Furthermore, surgery itself leads to increased ROS production and ischemia/reperfusion injury. Indeed, increased perioperative oxidative stress has been correlated with increased postoperative complications and prolonged recovery. Anesthesiologists care for patients during the whole spectrum of perioperative care and thus are in a unique position to deliver countermeasures to oxidative stress. Using preferentially an induction agent which reduces oxidative stress might lead to better clinical outcomes and fewer postoperative complications. Propofol has been shown in several studies to reduce oxidative stress, which reduces postoperative complications and leads to a faster recovery, and thus might represent the preferred induction agent in the right clinical setting.
Collapse
|
43
|
Han L, Zhao S, Xu F, Wang Y, Zhou R, Huang S, Ding Y, Deng D, Mao W, Chen X. Sevoflurane Increases Hippocampal Theta Oscillations and Impairs Memory Via TASK-3 Channels. Front Pharmacol 2021; 12:728300. [PMID: 34776954 PMCID: PMC8581481 DOI: 10.3389/fphar.2021.728300] [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: 06/21/2021] [Accepted: 10/12/2021] [Indexed: 11/21/2022] Open
Abstract
Sevoflurane can induce memory impairment during clinical anesthesia; however, the underlying mechanisms are largely unknown. TASK-3 channels are one of the potential targets of sevoflurane. Accumulating evidence supports a negative role of intracranial theta rhythms (4–12 Hz) in memory formation. Here, we investigated whether TASK-3 channels contribute to sevoflurane-induced memory impairment by regulating hippocampal theta rhythms. In this study, the memory performance of mice was tested by contextual fear conditioning and inhibitory avoidance experiments. The hippocampal local field potentials (LFPs) were recorded from chronically implanted electrodes located in CA3 region. The results showed that sevoflurane concentration-dependently impaired the memory function of mice, as evidenced by the decreased time mice spent on freezing and reduced latencies for mice to enter the shock compartment. Our electrophysiological results revealed that sevoflurane also enhanced the spectral power of hippocampal LFPs (1–30 Hz), particularly in memory-related theta rhythms (4–12 Hz). These effects were mitigated by viral-mediated knockdown of TASK-3 channels in the hippocampal CA3 region. The knockdown of hippocampal TASK-3 channels significantly reduced the enhancing effect of sevoflurane on hippocampal theta rhythms and alleviated sevoflurane-induced memory impairment. Our data indicate that sevoflurane can increase hippocampal theta oscillations and impair memory function via TASK-3 channels.
Collapse
Affiliation(s)
- Linlin Han
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Zhao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Xu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruihui Zhou
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiqian Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Ding
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Daling Deng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weike Mao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Coulter I, Timic Stamenic T, Eggan P, Fine BR, Corrigan T, Covey DF, Yang L, Pan JQ, Todorovic SM. Different roles of T-type calcium channel isoforms in hypnosis induced by an endogenous neurosteroid epipregnanolone. Neuropharmacology 2021; 197:108739. [PMID: 34339750 PMCID: PMC8478885 DOI: 10.1016/j.neuropharm.2021.108739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/09/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Many neuroactive steroids induce sedation/hypnosis by potentiating γ-aminobutyric acid (GABAA) currents. However, we previously demonstrated that an endogenous neuroactive steroid epipregnanolone [(3β,5β)-3-hydroxypregnan-20-one] (EpiP) exerts potent peripheral analgesia and blocks T-type calcium currents while sparing GABAA currents in rat sensory neurons. This study seeks to investigate the behavioral effects elicited by systemic administration of EpiP and to characterize its use as an adjuvant agent to commonly used general anesthetics (GAs). METHODS Here, we utilized electroencephalographic (EEG) recordings to characterize thalamocortical oscillations, as well as behavioral assessment and mouse genetics with wild-type (WT) and different knockout (KO) models of T-channel isoforms to investigate potential sedative/hypnotic and immobilizing properties of EpiP. RESULTS Consistent with increased oscillations in slower EEG frequencies, EpiP induced an hypnotic state in WT mice when injected alone intra-peritoneally (i.p.) and effectively facilitated anesthetic effects of isoflurane (ISO) and sevoflurane (SEVO). The CaV3.1 (Cacna1g) KO mice demonstrated decreased sensitivity to EpiP-induced hypnosis when compared to WT mice, whereas no significant difference was noted between CaV3.2 (Cacna1h), CaV3.3 (Cacna1i) and WT mice. Finally, when compared to WT mice, onset of EpiP-induced hypnosis was delayed in CaV3.2 KO mice but not in CaV3.1 and CaV3.3 KO mice. CONCLUSION We posit that EpiP may have an important role as novel hypnotic and/or adjuvant to volatile anesthetic agents. We speculate that distinct hypnotic effects of EpiP across all three T-channel isoforms is due to their differential expression in thalamocortical circuitry.
Collapse
Affiliation(s)
- Ian Coulter
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Tamara Timic Stamenic
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Pierce Eggan
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Brier R. Fine
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Timothy Corrigan
- Department of Pediatrics, Division of Neurology,
Translational Epilepsy Research Program, University of Colorado, Anschutz Medical
Campus, Aurora, CO 80045, USA
| | - Douglas F. Covey
- Department of Developmental Biology, Washington University
School of Medicine, St. Louis, MO 63110, USA;,Taylor Family Institute for Innovative Psychiatric
Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lingling Yang
- Stanley Center for Psychiatric Research, Broad Institute of
Harvard and MIT
| | - Jen Q. Pan
- Stanley Center for Psychiatric Research, Broad Institute of
Harvard and MIT
| | - Slobodan M. Todorovic
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045;,Neuroscience, University of Colorado, Anschutz Medical
Campus, Aurora 80045;,Pharmacology Graduate Programs, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| |
Collapse
|
45
|
He E, Xu S, Dai Y, Wang Y, Xiao G, Xie J, Xu S, Fan P, Mo F, Wang M, Song Y, Yin H, Li Y, Wang Y, Cai X. SWCNTs/PEDOT:PSS-Modified Microelectrode Arrays for Dual-Mode Detection of Electrophysiological Signals and Dopamine Concentration in the Striatum under Isoflurane Anesthesia. ACS Sens 2021; 6:3377-3386. [PMID: 34410704 DOI: 10.1021/acssensors.1c01241] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Accurate detection of the degree of isoflurane anesthesia during a surgery is important to avoid the risk of overdose isoflurane anesthesia timely. To address this challenge, a four-shank implantable microelectrode array (MEA) was fabricated for the synchronous real-time detection of dual-mode signals [electrophysiological signal and dopamine (DA) concentration] in rat striatum. The SWCNTs/PEDOT:PSS nanocomposites were modified onto the MEAs, which significantly improved the electrical and electrochemical performances of the MEAs. The electrical performance of the modified MEAs with a low impedance (16.20 ± 1.68 kΩ) and a small phase delay (-27.76 ± 0.82°) enabled the MEAs to detect spike firing with a high signal-to-noise ratio (> 3). The electrochemical performance of the modified MEAs with a low oxidation potential (160 mV), a low detection limit (10 nM), high sensitivity (217 pA/μM), and a wide linear range (10 nM-72 μM) met the specific requirements for DA detection in vivo. The anesthetic effect of isoflurane was mediated by inhibiting the spike firing of D2_SPNs (spiny projection neurons expressing the D2-type DA receptor) and the broadband oscillation rhythm of the local field potential (LFP). Therefore, the spike firing rate of D2_SPNs and the power of LFP could reflect the degree of isoflurane anesthesia together. During the isoflurane anesthesia-induced death procedure, we found that electrophysiological activities and DA release were strongly inhibited, and changes in the DA concentration provided more details regarding this procedure. The dual-mode recording MEA provided a detection method for the degree of isoflurane anesthesia and a prediction method for fatal overdose isoflurane anesthesia.
Collapse
Affiliation(s)
- Enhui He
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shengwei Xu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuchuan Dai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiding Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guihua Xiao
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingyu Xie
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shihong Xu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Penghui Fan
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fan Mo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mixia Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yilin Song
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huabing Yin
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Oakfield Avenue, Glasgow G12 8LT, United Kingdom
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, China
| | - Ying Wang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
46
|
Belelli D, Hales TG, Lambert JJ, Luscher B, Olsen R, Peters JA, Rudolph U, Sieghart W. GABA A receptors in GtoPdb v.2021.3. IUPHAR/BPS GUIDE TO PHARMACOLOGY CITE 2021; 2021. [PMID: 35005623 DOI: 10.2218/gtopdb/f72/2021.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The GABAA receptor is a ligand-gated ion channel of the Cys-loop family that includes the nicotinic acetylcholine, 5-HT3 and strychnine-sensitive glycine receptors. GABAA receptor-mediated inhibition within the CNS occurs by fast synaptic transmission, sustained tonic inhibition and temporally intermediate events that have been termed 'GABAA, slow' [45]. GABAA receptors exist as pentamers of 4TM subunits that form an intrinsic anion selective channel. Sequences of six α, three β, three γ, one δ, three ρ, one ε, one π and one θ GABAA receptor subunits have been reported in mammals [278, 235, 236, 283]. The π-subunit is restricted to reproductive tissue. Alternatively spliced versions of many subunits exist (e.g. α4- and α6- (both not functional) α5-, β2-, β3- and γ2), along with RNA editing of the α3 subunit [71]. The three ρ-subunits, (ρ1-3) function as either homo- or hetero-oligomeric assemblies [359, 50]. Receptors formed from ρ-subunits, because of their distinctive pharmacology that includes insensitivity to bicuculline, benzodiazepines and barbiturates, have sometimes been termed GABAC receptors [359], but they are classified as GABA A receptors by NC-IUPHAR on the basis of structural and functional criteria [16, 235, 236]. Many GABAA receptor subtypes contain α-, β- and γ-subunits with the likely stoichiometry 2α.2β.1γ [168, 235]. It is thought that the majority of GABAA receptors harbour a single type of α- and β - subunit variant. The α1β2γ2 hetero-oligomer constitutes the largest population of GABAA receptors in the CNS, followed by the α2β3γ2 and α3β3γ2 isoforms. Receptors that incorporate the α4- α5-or α 6-subunit, or the β1-, γ1-, γ3-, δ-, ε- and θ-subunits, are less numerous, but they may nonetheless serve important functions. For example, extrasynaptically located receptors that contain α6- and δ-subunits in cerebellar granule cells, or an α4- and δ-subunit in dentate gyrus granule cells and thalamic neurones, mediate a tonic current that is important for neuronal excitability in response to ambient concentrations of GABA [209, 272, 83, 19, 288]. GABA binding occurs at the β+/α- subunit interface and the homologous γ+/α- subunits interface creates the benzodiazepine site. A second site for benzodiazepine binding has recently been postulated to occur at the α+/β- interface ([254]; reviewed by [282]). The particular α-and γ-subunit isoforms exhibit marked effects on recognition and/or efficacy at the benzodiazepine site. Thus, receptors incorporating either α4- or α6-subunits are not recognised by 'classical' benzodiazepines, such as flunitrazepam (but see [356]). The trafficking, cell surface expression, internalisation and function of GABAA receptors and their subunits are discussed in detail in several recent reviews [52, 140, 188, 316] but one point worthy of note is that receptors incorporating the γ2 subunit (except when associated with α5) cluster at the postsynaptic membrane (but may distribute dynamically between synaptic and extrasynaptic locations), whereas as those incorporating the δ subunit appear to be exclusively extrasynaptic. NC-IUPHAR [16, 235, 3, 2] class the GABAA receptors according to their subunit structure, pharmacology and receptor function. Currently, eleven native GABAA receptors are classed as conclusively identified (i.e., α1β2γ2, α1βγ2, α3βγ2, α4βγ2, α4β2δ, α4β3δ, α5βγ2, α6βγ2, α6β2δ, α6β3δ and ρ) with further receptor isoforms occurring with high probability, or only tentatively [235, 236]. It is beyond the scope of this Guide to discuss the pharmacology of individual GABAA receptor isoforms in detail; such information can be gleaned in the reviews [16, 95, 168, 173, 143, 278, 216, 235, 236] and [9, 10]. Agents that discriminate between α-subunit isoforms are noted in the table and additional agents that demonstrate selectivity between receptor isoforms, for example via β-subunit selectivity, are indicated in the text below. The distinctive agonist and antagonist pharmacology of ρ receptors is summarised in the table and additional aspects are reviewed in [359, 50, 145, 223]. Several high-resolution cryo-electron microscopy structures have been described in which the full-length human α1β3γ2L GABAA receptor in lipid nanodiscs is bound to the channel-blocker picrotoxin, the competitive antagonist bicuculline, the agonist GABA (γ-aminobutyric acid), and the classical benzodiazepines alprazolam and diazepam [198].
Collapse
|
47
|
Towards Quantum-Chemical Modeling of the Activity of Anesthetic Compounds. Int J Mol Sci 2021; 22:ijms22179272. [PMID: 34502179 PMCID: PMC8431746 DOI: 10.3390/ijms22179272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022] Open
Abstract
The modeling of the activity of anesthetics is a real challenge because of their unique electronic and structural characteristics. Microscopic approaches relevant to the typical features of these systems have been developed based on the advancements in the theory of intermolecular interactions. By stressing the quantum chemical point of view, here, we review the advances in the field highlighting differences and similarities among the chemicals within this group. The binding of the anesthetics to their partners has been analyzed by Symmetry-Adapted Perturbation Theory to provide insight into the nature of the interaction and the modeling of the adducts/complexes allows us to rationalize their anesthetic properties. A new approach in the frame of microtubule concept and the importance of lipid rafts and channels in membranes is also discussed.
Collapse
|
48
|
Lin D, Liu J, Florveus A, Ganesan V, Cottrell JE, Kass IS. Exposure to Sevoflurane, But Not Ketamine, During Early-life Brain Development has Long-Lasting Effects on GABA A Receptor Mediated Inhibitory Neurotransmission. Neuroscience 2021; 472:116-127. [PMID: 34384844 DOI: 10.1016/j.neuroscience.2021.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 11/19/2022]
Abstract
Understanding the different mechanisms associated with different anesthetic targeted receptors is critical towards identifying accurate long-term outcome measures as a result of early-life anesthetic exposure. We examined changes in GABAA receptor mediated neurotransmission by a predominately GABAA receptor targeted anesthetic, sevoflurane or a predominately NMDA receptor targeted anesthetic, ketamine. Postnatal day 7 male mice were exposed to sevoflurane or ketamine and examined as adults for changes in inhibitory neurotransmission and its associated change in induced seizure activity. Paired pulse stimulation experiment showed that early-life sevoflurane treated mice had significantly less hippocampal CA1 inhibition later in life. There was significantly increased CA1 excitatory output in the sevoflurane treated group compared to the no sevoflurane treated group after the GABA agonist muscimol. Similar to our previously established data for early-life sevoflurane, here we established early-life ketamine administration resulted in neurodevelopmental behavioral changes later in life. However, muscimol did not produce a significant difference on the excitatory CA1 output between early-life ketamine group and saline group. While sevoflurane treated mice showed significantly higher induced seizure intensities and shorter latency periods to reach seizure intensity stage 5 (Racine score) compared with no sevoflurane treated mice, this phenomenon was not observed in the ketamine vs. saline treated groups. Early-life sevoflurane, but not ketamine, exposure reduced GABAergic inhibition and enhanced seizure activity later in life. The results indicate that early-life exposure to different anesthetics lead to distinct long-term effects and their unique pathways require mechanistic studies to understand induced long-lasting changes in the brain.
Collapse
Affiliation(s)
- Daisy Lin
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA; Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
| | - Jinyang Liu
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
| | - Alizna Florveus
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
| | - Vanathi Ganesan
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
| | - James E Cottrell
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA
| | - Ira S Kass
- Department of Anesthesiology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA; Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203-2098, USA.
| |
Collapse
|
49
|
Isoflurane Suppresses Hippocampal High-frequency Ripples by Differentially Modulating Pyramidal Neurons and Interneurons in Mice. Anesthesiology 2021; 135:122-135. [PMID: 33951177 DOI: 10.1097/aln.0000000000003803] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Isoflurane can induce anterograde amnesia. Hippocampal ripples are high-frequency oscillatory events occurring in the local field potentials of cornu ammonis 1 involved in memory processes. The authors hypothesized that isoflurane suppresses hippocampal ripples at a subanesthetic concentration by modulating the excitability of cornu ammonis 1 neurons. METHODS The potencies of isoflurane for memory impairment and anesthesia were measured in mice. Hippocampal ripples were measured by placing recording electrodes in the cornu ammonis 1. Effects of isoflurane on the excitability of hippocampal pyramidal neurons and interneurons were measured. A simulation model of ripples based on the firing frequency of hippocampal cornu ammonis 1 neurons was used to validate the effects of isoflurane on neuronal excitability in vitro and on ripples in vivo. RESULTS Isoflurane at 0.5%, which did not induce loss of righting reflex, impaired hippocampus-dependent fear memory by 97.4 ± 3.1% (mean ± SD; n = 14; P < 0.001). Isoflurane at 0.5% reduced ripple amplitude (38 ± 13 vs. 42 ± 13 μV; n = 9; P = 0.003), rate (462 ± 66 vs. 538 ± 81 spikes/min; n = 9; P = 0.002) and duration (36 ± 5 vs. 48 ± 9 ms; n = 9; P < 0.001) and increased the interarrival time (78 ± 7 vs. 69 ± 6 ms; n = 9; P < 0.001) and frequency (148.2 ± 3.9 vs. 145.0 ± 2.9 Hz; n = 9; P = 0.001). Isoflurane at the same concentration depressed action potential frequency in fast-spiking interneurons while slightly enhancing action potential frequency in cornu ammonis 1 pyramidal neurons. The simulated effects of isoflurane on hippocampal ripples were comparable to recordings in vivo. CONCLUSIONS The authors' results suggest that a subanesthetic concentration of isoflurane can suppress hippocampal ripples by differentially modulating the excitability of pyramidal neurons and interneurons, which may contribute to its amnestic action. EDITOR’S PERSPECTIVE
Collapse
|
50
|
Liu Y, Yang H, Fu Y, Pan Z, Qiu F, Xu Y, Yang X, Chen Q, Ma D, Liu Z. TRPV1 Antagonist Prevents Neonatal Sevoflurane-Induced Synaptic Abnormality and Cognitive Impairment in Mice Through Regulating the Src/Cofilin Signaling Pathway. Front Cell Dev Biol 2021; 9:684516. [PMID: 34307363 PMCID: PMC8293754 DOI: 10.3389/fcell.2021.684516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/14/2021] [Indexed: 11/13/2022] Open
Abstract
Long-term neurodevelopmental disorders following neonatal anesthesia have been reported both in young animals and in children. The activation of transient receptor potential vanilloid 1 (TRPV1) channels in hippocampus adversely affects neurodevelopment. The current study explored the underlying mechanism of TRPV1 channels on long-lasting cognitive dysfunction induced by anesthetic exposure to the developing brain. we demonstrated that TRPV1 expression was increased after sevoflurane exposure both in vitro and in vivo. Sevoflurane exposure to hippocampal neurons decreased the synaptic density and the surface GluA1 expression, as well as increased co-localization of internalized AMPAR in early and recycling endosomes. Sevoflurane exposure to newborn mice impaired learning and memory in adulthood, and reduced AMPAR subunit GluA1, 2 and 3 expressions in the crude synaptosomal fractions from mouse hippocampus. The inhibition of TRPV1 reversed the phenotypic changes induced by sevoflurane. Moreover, sevoflurane exposure increased Src phosphorylation at tyrosine 416 site thereby reducing cofilin phosphorylation. TRPV1 blockade reversed these suppressive effects of sevoflurane. Our data suggested that TRPV1 antagonist may protect against synaptic damage and cognitive dysfunction induced by sevoflurane exposure during the brain developing stage.
Collapse
Affiliation(s)
- Yuqiang Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Han Yang
- Department of Anesthesiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yifei Fu
- Department of Anesthesiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhenglong Pan
- Department of Anesthesiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Fang Qiu
- Department of Anesthesiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yanwen Xu
- Shenzhen Key Laboratory of Neurosurgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Xinping Yang
- Department of Anesthesiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qian Chen
- 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
| | - 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
| | - Zhiheng Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|