1
|
Tang A, Xu M, Chen X, Liu J, Wang J, Wang Y, Cai S, Shu Y, Zheng D, Yu T, Wang Y, Luo T, Yu S. Somatostatin-expressing Neurons in the Medial Prefrontal Cortex Promote Sevoflurane Anesthesia in Mice. Anesthesiology 2025; 142:844-862. [PMID: 39869666 DOI: 10.1097/aln.0000000000005394] [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: 01/29/2025]
Abstract
BACKGROUND The medial prefrontal cortex plays a crucial role in regulating consciousness. However, the specific functions of its excitatory and inhibitory networks during anesthesia remain uncertain. Here, the authors explored the hypothesis that somatostatin interneurons in the medial prefrontal cortex enhance the effects of sevoflurane anesthesia by increasing γ-aminobutyric acid (GABA) transmission to pyramidal neurons. METHODS Electroencephalography was utilized to reflect the depth of anesthesia. Immunostaining and fiber photometry were employed to assess neuronal activities and GABA delivery. The regulation of neuronal activity was achieved by chemogenetics and optogenetics. RESULTS The expression of c-Fos was increased in somatostatin neurons of the medial prefrontal cortex during sevoflurane anesthesia (air vs. sevoflurane: 26.4 ± 6.5% vs. 48 ± 6.2%; P = 0.0007; n = 5 mice). Chemogenetic inhibition or activation of somatostatin neurons in the medial prefrontal cortex reduced (from 84 ± 24 s to 51 ± 18 s; P = 0.008; n = 7 mice) or prolonged (from 97 ± 31 s to 140 ± 30 s; P = 0.006; n = 7 mice) the sevoflurane anesthesia recovery time. Increased GABA input to pyramidal neurons in the medial prefrontal cortex precedes sevoflurane-induced loss of consciousness (baseline vs . pre-loss of the righting reflex: from 0.46 ± 0.57% to 2.25 ± 1.42%; P = 0.031; n = 10 mice). Activation of somatostatin neurons in the medial prefrontal cortex leads to a significant reduction in calcium signals within local pyramidal neurons (baseline vs . 20 Hz stimulation: from -0.14 ± 0.52% to -10.08 ± 4.44%; P = 0.002; n = 10 mice). Additionally,GABA input on pyramidal neurons increased (baseline vs . 20 Hz stimulation: from -0.001 ± 0.001% to 0.28 ± 0.03%; P = 0.002; n = 7 mice) in a time-locked manner. Chemogenetic inhibition of pyramidal neurons prolonged the recovery from sevoflurane anesthesia in mice (from 101 ± 46 s to 136 ± 54 s; P = 0.017; n = 19 mice). CONCLUSIONS Cortical somatostatin neurons may inhibit local pyramidal neurons by enhancing GABA transmission, which increases the effectiveness of sevoflurane anesthesia.
Collapse
Affiliation(s)
- Aichen Tang
- School of Anesthesiology, Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
| | - Mao Xu
- School of Anesthesiology, Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
| | - Xizu Chen
- School of Anesthesiology,Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
| | - Juan Liu
- School of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Jiamin Wang
- School of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Ying Wang
- School of Anesthesiology, Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
| | - Shuang Cai
- School of Anesthesiology, Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
| | - Yue Shu
- School of Anesthesiology, Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
| | - Danxu Zheng
- School of Anesthesiology, Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
| | - Tian Yu
- School of Anesthesiology, Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
| | - Yuan Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tianyuan Luo
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shouyang Yu
- School of Anesthesiology, Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
| |
Collapse
|
2
|
Fan BQ, Xia JM, Yi XW, Yuan TJ, Zhou Y, Xu R, Wang L, Wang D, Xia Y, Yi WJ, Ding JH, Qu WM, Li WX, Huang ZL, Han Y. Ventral pallidum GABAergic and glutamatergic neurons modulate arousal during sevoflurane general anaesthesia in male mice. Br J Pharmacol 2025. [PMID: 40205920 DOI: 10.1111/bph.70019] [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: 09/15/2024] [Revised: 01/17/2025] [Accepted: 02/12/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND AND PURPOSE The induction and emergence of general anaesthesia involve an altered process of states of consciousness, yet the central nervous system mechanisms remain inadequately understood. The ventral pallidum (VP) within the basal ganglia is crucial in sleep-wake modulation. However, its involvement in general anaesthesia and the underlying neuronal mechanisms are not well elucidated. EXPERIMENTAL APPROACH In vivo electrophysiological recordings were conducted to examine changes in the activity of different types of VP neurons before and after sevoflurane exposure. Fibre photometry, combined with electroencephalogram and electromyography recordings, was employed to analyse neuronal activity during both the induction and recovery phases of sevoflurane anaesthesia. Chemogenetics was implemented to investigate the impact of modulated neuronal activity on anaesthesia induction and emergence, whereas optogenetics was used for real time activation of neurons at different depths of anaesthesia. KEY RESULTS Sevoflurane exposure reduced the firing activity of both VP GABAergic (VPGABA) and VP glutamatergic (VPglu) neurons, without affecting cholinergic neurons. VPGABA and VPglu neuronal activity decreased during sevoflurane anaesthesia induction and increased during emergence. Manipulation of VPGABA neurons bidirectionally influenced the duration of induction and emergence. Inhibiting VPglu neurons accelerated induction. Real time activation of VPGABA neurons triggered cortical activation and behavioural emergence during steady-state sevoflurane anaesthesia and reduced the burst suppression ratio during deep anaesthesia. CONCLUSION AND IMPLICATIONS These findings highlight the role of VPGABA and VPglu neurons in modulating transitions between anaesthesia stages, providing valuable insights into the neuronal mechanisms underlying sevoflurane-induced anaesthesia.
Collapse
Affiliation(s)
- Bing-Qian Fan
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Centre for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anaesthesiology and Perioperative Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jun-Ming Xia
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Xiu-Wen Yi
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Tian-Jie Yuan
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Yu Zhou
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Rui Xu
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Lu Wang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Centre for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep, Shanghai Medical College, Fudan University, Shanghai, China
| | - Di Wang
- Department of Anaesthesiology and Perioperative Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Xia
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Wen-Jing Yi
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Jia-Hui Ding
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Centre for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Xian Li
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Centre for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anaesthesiology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Yuan Han
- Department of Anaesthesiology, Eye & ENT Hospital of Fudan University, Shanghai, China
| |
Collapse
|
3
|
Yang X, Zhu S, Xia M, Sun L, Li S, Xiang P, Li F, Deng Q, Chen L, Zhang W, Wang Y, Li Q, Lyu Z, Du X, Du J, Yang Q, Luo Y. The Serotonergic Dorsal Raphe Promotes Emergence from Propofol Anesthesia in Zebrafish. J Neurosci 2025; 45:e2125232025. [PMID: 39947921 PMCID: PMC11984078 DOI: 10.1523/jneurosci.2125-23.2025] [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: 11/07/2023] [Revised: 12/30/2024] [Accepted: 01/27/2025] [Indexed: 04/11/2025] Open
Abstract
The mechanisms through which general anesthetics induce loss of consciousness remain unclear. Previous studies have suggested that dorsal raphe nucleus serotonergic (DRN5-HT) neurons are involved in inhalational anesthesia, but the underlying neuronal and synaptic mechanisms are not well understood. In this study, we investigated the role of DRN5-HT neurons in propofol-induced anesthesia in larval zebrafish (sex undetermined at this developmental stage) using a combination of in vivo single-cell calcium imaging, two-photon laser ablation, optogenetic activation, in vivo glutamate imaging, and in vivo whole-cell recording. We found that calcium activity of DRN5-HT neurons reversibly decreased during propofol perfusion. Ablation of DRN5-HT neurons prolonged emergence from 30 µM propofol anesthesia, while induction times were not affected under concentrations of 1, 3, and 30 µM. Additionally, optogenetic activation of DRN5-HT neurons strongly promoted emergence from propofol anesthesia. Propofol application to DRN5-HT neurons suppressed both spontaneous and current injection-evoked spike firing, abolished spontaneous excitatory postsynaptic currents, and decreased membrane input resistance. Presynaptic glutamate release events in DRN5-HT neurons were also abolished by propofol. Furthermore, the hyperpolarization of DRN5-HT neurons caused by propofol was abolished by picrotoxin, a GABAA receptor antagonist, which shortened emergence time from propofol anesthesia when locally applied to the DRN. Our results reveal that DRN5-HT neurons in zebrafish are involved in the emergence from propofol anesthesia by inhibiting presynaptic excitatory glutamate inputs and inducing GABAA receptor-mediated hyperpolarization.
Collapse
Affiliation(s)
- Xiaoxuan Yang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shan Zhu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Miaoyun Xia
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Le Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sha Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Peishan Xiang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Funing Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiusui Deng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lijun Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ying Wang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qiang Li
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhuochen Lyu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xufei Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qianzi Yang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
4
|
Zhou K, Hou ZJ, Jiang XL, Xiao YJ, Zhang LC, Xu W, Xiong B, Qu WM, Huang YG, Huang ZL, Wang L. Striatal neurones expressing D1 dopamine receptors modulate consciousness in sevoflurane but not propofol anaesthesia in mice. Br J Anaesth 2025; 134:1105-1121. [PMID: 39915158 PMCID: PMC11947605 DOI: 10.1016/j.bja.2024.10.049] [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: 11/18/2023] [Revised: 09/09/2024] [Accepted: 10/02/2024] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Sevoflurane and propofol are the most widely used inhaled and i.v. general anaesthetics, respectively. The mechanisms by which sevoflurane and propofol induce loss of consciousness (LOC) remain unclear. Recent studies implicate the brain dopaminergic circuit in anaesthetic-induced LOC and the cortical-striatal-thalamic-cortical loop in decoding consciousness. We investigated the contribution of the dorsal striatum, which is a critical interface between the dopaminergic circuit and the cortical-striatal-thalamic-cortical loop, in sevoflurane and propofol anaesthesia. METHODS Electroencephalography and electromyography recordings and righting reflex tests were used to determine LOC and recovery of consciousness (ROC). The activity of D1 dopamine receptor (D1R)-expressing neurones in the dorsal striatum was monitored using fibre photometry, and regulated using optogenetic and chemogenetic methods in D1R-Cre mice. RESULTS Population activities of striatal D1R neurones began to decrease before LOC and gradually returned after ROC. During sevoflurane anaesthesia, optogenetic activation of striatal D1R neurones induced ROC at cortical and behavioural levels in steady-state anaesthesia and promoted cortical activation in deep burst suppression anaesthesia. Chemogenetic inhibition of striatal D1R neurones accelerated induction (from 242.0 [46.1] to 194.0 [26.9] s; P=0.010) and delayed emergence (from 93.5 [21.2] to 133.5 [33.9] s; P=0.005), whereas chemogenetic activation of these neurones accelerated emergence (from 107 [23.7] to 81.3 [16.1] s; P=0.011). However, neither optogenetic nor chemogenetic manipulation of striatal D1R neurones had any effects on propofol anaesthesia. CONCLUSIONS Striatal D1R neurones modulate the state of consciousness in sevoflurane anaesthesia, but not in propofol anaesthesia.
Collapse
Affiliation(s)
- Kang Zhou
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, Fudan University, Shanghai, China; Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zi-Jun Hou
- Department of Anesthesiology, Yijishan Hospital, Wannan Medical College, Wuhu, China
| | - Xu-Liang Jiang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Jie Xiao
- Department of Anesthesiology, Central South University, Changsha, Hunan, China
| | - Lin-Chen Zhang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, Fudan University, Shanghai, China
| | - Wei Xu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, Fudan University, Shanghai, China
| | - Bo Xiong
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, Fudan University, Shanghai, China
| | - Yu-Guang Huang
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, Fudan University, Shanghai, China; Department of Anesthesiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Lu Wang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Guillaumin MCC, Harding CD, Krone LB, Yamagata T, Kahn MC, Blanco-Duque C, Banks GT, Achermann P, Diniz Behn C, Nolan PM, Peirson SN, Vyazovskiy VV. Deficient synaptic neurotransmission results in a persistent sleep-like cortical activity across vigilance states in mice. Curr Biol 2025:S0960-9822(25)00267-2. [PMID: 40118064 DOI: 10.1016/j.cub.2025.02.053] [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: 08/14/2023] [Revised: 12/10/2024] [Accepted: 02/25/2025] [Indexed: 03/23/2025]
Abstract
Growing evidence suggests that brain activity during sleep, as well as sleep regulation, are tightly linked with synaptic function and network excitability at the local and global levels. We previously reported that a mutation in synaptobrevin 2 (Vamp2) in restless (rlss) mice results in a marked increase of wakefulness and suppression of sleep, in particular REM sleep (REMS), as well as increased consolidation of sleep and wakefulness. In this study, using finer-scale in vivo electrophysiology recordings, we report that spontaneous cortical activity in rlss mice during NREM sleep (NREMS) is characterized by an occurrence of abnormally prolonged periods of complete neuronal silence (OFF-periods), often lasting several seconds, similar to the burst suppression pattern typically seen under deep anesthesia. Increased incidence of prolonged network OFF-periods was not specific to NREMS but also present in REMS and wake in rlss mice. Slow-wave activity (SWA) was generally increased in rlss mice relative to controls, while higher frequencies, including theta-frequency activity, were decreased, further resulting in diminished differences between vigilance states. The relative increase in SWA after sleep deprivation was attenuated in rlss mice, suggesting either that rlss mice experience persistently elevated sleep pressure or, alternatively, that the intrusion of sleep-like patterns of activity into the wake state attenuates the accumulation of sleep drive. We propose that a deficit in global synaptic neurotransmitter release leads to "state inertia," reflected in an abnormal propensity of brain networks to enter and remain in a persistent "default state" resembling coma or deep anesthesia.
Collapse
Affiliation(s)
- Mathilde C C Guillaumin
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK; Medical Research Council Brain Network Dynamics Unit, Nuffield Department of Clinical Neurosciences, University of Oxford, Mansfield Road, Oxford OX1 3TH, UK; Sir Jules Thorn Sleep and Circadian Neuroscience Institute (SCNi), University of Oxford, South Parks Road, Oxford OX1 3QU, UK.
| | - Christian D Harding
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute (SCNi), University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Lukas B Krone
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute (SCNi), University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK; University Hospital of Psychiatry and Psychotherapy, University of Bern, Hochschulstrasse 6, Bern 3012, Switzerland
| | - Tomoko Yamagata
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK; Sir Jules Thorn Sleep and Circadian Neuroscience Institute (SCNi), University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Martin C Kahn
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute (SCNi), University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Cristina Blanco-Duque
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute (SCNi), University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Gareth T Banks
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Science and Innovation Campus, Didcot OX11 0RD, UK
| | - Peter Achermann
- Institute of Pharmacology and Toxicology, University of Zürich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Cecilia Diniz Behn
- Department of Applied Mathematics & Statistics, Colorado School of Mines, 1301 19(th) Street, Golden, CO 80401, USA; Department of Pediatrics, University of Colorado Anschutz Medical Campus, 13001 East 17(th) Place, Aurora, CO 80045, USA
| | - Patrick M Nolan
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Science and Innovation Campus, Didcot OX11 0RD, UK
| | - Stuart N Peirson
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK; Sir Jules Thorn Sleep and Circadian Neuroscience Institute (SCNi), University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Vladyslav V Vyazovskiy
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute (SCNi), University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford OX1 3QU, UK; Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK.
| |
Collapse
|
6
|
Yuan Y, Liu T, Wang J. Enhancing anesthetic techniques for improving whisker stimulation response in the barrel cortex. PLoS One 2025; 20:e0318306. [PMID: 39999042 DOI: 10.1371/journal.pone.0318306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/14/2025] [Indexed: 02/27/2025] Open
Abstract
This study adopts and validates an anesthetic protocol designed for rat whisker stimulation experiments, achieving significant enhancements in the neural response of the barrel field cortex. By combining alpha-chloralose, low-dose Isoflurane (0.5%) and Dexdomitor, the protocol not only maintains a stable anesthetic state but also markedly improves the amplitude and latency of local field potential (LFP) signals. Experimental results reveal that LFP amplitudes in the barrel field under this protocol are twice as high as those achieved with Isoflurane and four times as high as those with Ketamine-Xylazine, with significantly shortened latencies and reduced noise interference. For the first time, power spectral analysis reveals a distinct enhancement of oscillatory power in the alpha (8-13 Hz) and beta (13-30 Hz) bands under alpha-chloralose anesthesia, diverging from the traditional dominance of delta (0.5-4 Hz) oscillations observed with other anesthetics. Mechanistically, this phenomenon may be attributed to alpha-chloralose's unique modulation of GABAergic and glutamatergic pathways, promoting cortical desynchronization and enhanced sensory processing. This protocol offers new insights into optimizing sensory-evoked neural signal acquisition and provides a reference for future studies exploring neural modulation in sensory neuroscience.
Collapse
Affiliation(s)
- Ye Yuan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Tian Liu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jue Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
7
|
Maniaci A, Lentini M, Trombadore R, Gruppuso L, Milardi S, Scrofani R, Cuttone G, Sorbello M, Modica R, Lechien JR, Boscolo-Rizzo P, Paternò DS, La Via L. Neurological and Olfactory Disturbances After General Anesthesia. Life (Basel) 2025; 15:344. [PMID: 40141689 PMCID: PMC11944247 DOI: 10.3390/life15030344] [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: 01/08/2025] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Neurological and olfactory disturbances are increasingly recognized as potential complications of general anesthesia, particularly in vulnerable populations, such as the elderly, children, and individuals with comorbidities. Recent studies have highlighted the need for tailored anesthetic approaches in these high-risk groups to mitigate potential long-term effects. These disturbances, including postoperative cognitive dysfunction, delirium, and olfactory deficits, often arise from shared pathophysiological mechanisms, such as neuroinflammation, oxidative stress, and disruptions in cerebral perfusion. The olfactory system is particularly susceptible to anesthesia-induced neurotoxicity given its proximity to central nervous system structures and its role in sensory and cognitive processing. Furthermore, the unique regenerative capacity of olfactory neurons may be compromised by prolonged or repeated exposure to anesthetic agents, potentially leading to long-term olfactory dysfunction. Risk factors, such as advanced age, neurodegenerative diseases, diabetes, cardiovascular conditions, genetic predispositions, and the type and duration of anesthesia exposure, further exacerbate these complications. Preventive strategies, including comprehensive preoperative risk assessment, personalized anesthetic protocols based on genetic and physiological profiles, and proactive postoperative care with early intervention programs, are critical for reducing impairments and improving long-term patient outcomes. Emerging evidence highlights the potential role of neuroprotective agents, such as antioxidants and anti-inflammatory therapies, in mitigating the effects of anesthesia-induced neurotoxicity. Longitudinal studies are needed to evaluate the long-term effects of anesthesia on cognitive and sensory health, particularly in high-risk populations. These studies should incorporate advanced neuroimaging techniques and biomarker analysis to elucidate the underlying mechanisms of anesthesia-induced neurological and olfactory disturbances. This narrative review provides a comprehensive overview of the mechanisms, risk factors, and preventive strategies for neurological and olfactory disturbances after general anesthesia and highlights future directions for research to improve patient outcomes. We conducted a comprehensive literature search using databases, such as PubMed and Scopus, to identify relevant studies.
Collapse
Affiliation(s)
- Antonino Maniaci
- Deparment of Medicine and Surgery, University of Enna “Kore”, 94019 Enna, Italy; (M.L.); (G.C.); (M.S.)
- Asp 7 Ragusa, 97100 Ragusa, Italy; (R.T.); (L.G.); (S.M.); (R.S.); (R.M.); (D.S.P.)
- Division of Laryngology and Bronchoesophagology, Department of Otolaryngology Head Neck Surgery, EpiCURA Hospital, UMONS Research Institute for Health Sciences and Technology, Faculty of Medicine, University of Mons (UMons), 7000 Mons, Belgium;
| | - Mario Lentini
- Deparment of Medicine and Surgery, University of Enna “Kore”, 94019 Enna, Italy; (M.L.); (G.C.); (M.S.)
- Asp 7 Ragusa, 97100 Ragusa, Italy; (R.T.); (L.G.); (S.M.); (R.S.); (R.M.); (D.S.P.)
| | - Rosario Trombadore
- Asp 7 Ragusa, 97100 Ragusa, Italy; (R.T.); (L.G.); (S.M.); (R.S.); (R.M.); (D.S.P.)
| | - Loris Gruppuso
- Asp 7 Ragusa, 97100 Ragusa, Italy; (R.T.); (L.G.); (S.M.); (R.S.); (R.M.); (D.S.P.)
| | - Santo Milardi
- Asp 7 Ragusa, 97100 Ragusa, Italy; (R.T.); (L.G.); (S.M.); (R.S.); (R.M.); (D.S.P.)
| | - Rosario Scrofani
- Asp 7 Ragusa, 97100 Ragusa, Italy; (R.T.); (L.G.); (S.M.); (R.S.); (R.M.); (D.S.P.)
| | - Giuseppe Cuttone
- Deparment of Medicine and Surgery, University of Enna “Kore”, 94019 Enna, Italy; (M.L.); (G.C.); (M.S.)
| | - Massimiliano Sorbello
- Deparment of Medicine and Surgery, University of Enna “Kore”, 94019 Enna, Italy; (M.L.); (G.C.); (M.S.)
- Asp 7 Ragusa, 97100 Ragusa, Italy; (R.T.); (L.G.); (S.M.); (R.S.); (R.M.); (D.S.P.)
| | - Rodolfo Modica
- Asp 7 Ragusa, 97100 Ragusa, Italy; (R.T.); (L.G.); (S.M.); (R.S.); (R.M.); (D.S.P.)
| | - Jerome R. Lechien
- Division of Laryngology and Bronchoesophagology, Department of Otolaryngology Head Neck Surgery, EpiCURA Hospital, UMONS Research Institute for Health Sciences and Technology, Faculty of Medicine, University of Mons (UMons), 7000 Mons, Belgium;
| | - Paolo Boscolo-Rizzo
- Department of Medical, Surgical and Health Sciences, Section of Otolaryngology, University of Trieste, 34121 Trieste, Italy;
| | | | - Luigi La Via
- Department of Anesthesia and Intensive Care, University Hospital Policlinico “G. Rodolico-San Marco”, 95123 Catania, Italy;
| |
Collapse
|
8
|
Thong S, Doery E, Biabani M, Rogasch NC, Chong TTJ, Hendrikse J, Coxon JP. Disinhibition across Secondary Motor Cortical Regions during Motor Sequence Learning: A TMS-EEG Study. J Neurosci 2025; 45:e0443242024. [PMID: 39658257 PMCID: PMC11841768 DOI: 10.1523/jneurosci.0443-24.2024] [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: 03/07/2024] [Revised: 10/27/2024] [Accepted: 11/26/2024] [Indexed: 12/12/2024] Open
Abstract
Secondary motor cortical regions, such as the supplementary motor area (SMA), are involved in planning and learning motor sequences; however, the neurophysiological mechanisms across these secondary cortical networks remain poorly understood. In the primary motor cortex, changes in excitatory and inhibitory neurotransmission (E:I balance) accompany motor sequence learning. In particular, there is an early reduction in inhibition (i.e., disinhibition). Here, we investigated whether disinhibition occurs across secondary motor cortical regions during motor sequence learning using combined transcranial magnetic stimulation (TMS) and electroencephalography (EEG). Twenty-nine healthy adults (14 female) practiced a sequential motor task with TMS applied to the SMA during sequence planning. TMS-evoked potentials (TEPs) were measured with EEG before, during, and after practice. The N45 TEP peak was our primary measure of disinhibition, while we analyzed the slope of aperiodic EEG activity as an additional E:I balance measure. We observed a reduction in N45 amplitudes across an electrode cluster encompassing the SMA and nearby cortical regions as participants began learning new motor sequences, compared with a baseline rest phase (p < 0.01). Smaller N45 amplitudes during early learning were associated with improvements in reaction times across learning (p < 0.05). Intriguingly, aperiodic exponents increased as learning progressed and were associated with greater improvements in skill (p < 0.05). Overall, our results show that inhibition is modulated across SMA and secondary motor cortex during the planning phase of motor sequence learning and thus provide novel insight on the neurophysiological mechanisms within higher-order motor cortex that accompany new sequence learning.
Collapse
Affiliation(s)
- Sophie Thong
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, 3800 Victoria, Australia
| | - Elizabeth Doery
- Department of Health, School of Psychology, Deakin University, Melbourne, 3125 Victoria, Australia
| | - Mana Biabani
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, 3800 Victoria, Australia
| | - Nigel C Rogasch
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, 3800 Victoria, Australia
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, 5005 South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, 5001 South Australia, Australia
| | - Trevor T-J Chong
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, 3800 Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, 3004 Victoria, Australia
- Department of Clinical Neurosciences, St. Vincent's Hospital, Melbourne, 3065 Victoria, Australia
| | - Joshua Hendrikse
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, 3800 Victoria, Australia
| | - James P Coxon
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, 3800 Victoria, Australia
| |
Collapse
|
9
|
Li K, Li M, Liu W, Wu Y, Li F, Xie J, Zhou S, Wang S, Guo Y, Pan J, Wang X. Electroencephalographic differences between waking and sleeping periods in patients with prolonged disorders of consciousness at different levels of consciousness. Front Hum Neurosci 2025; 19:1521355. [PMID: 40034215 PMCID: PMC11872887 DOI: 10.3389/fnhum.2025.1521355] [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: 11/01/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Objective This study aimed to explore differences in sleep electroencephalogram (EEG) patterns in individuals with prolonged disorders of consciousness, utilizing polysomnography (PSG) to assist in distinguishing between the vegetative state (VS)/unresponsive wakefulness syndrome (UWS) and the minimally conscious state (MCS), thereby reducing misdiagnosis rates and enhancing the quality of medical treatment. Methods A total of 40 patients with prolonged disorders of consciousness (pDOC; 27 patients in the VS/UWS and 13 in the MCS) underwent polysomnography. We analyzed differential EEG indices between VS/UWS and MCS groups and performed correlation analyses between these indices and the Coma Recovery Scale-Revised (CRS-R) scores. The diagnostic accuracy of the differential indices was evaluated using receiver operating characteristic (ROC) curves. Results 1. The fractal dimension (Higuchi's fractal dimension (HFD)) of patients in the MCS tended to be higher than that of patients in the VS/UWS across all phases, with a significant difference only in the waking phase (p < 0.05). The HFD in the waking phase was positively correlated with the CRS-R score and exhibited the highest diagnostic accuracy at 88.3%. The Teager-Kaiser energy operator (TKEO) also showed higher levels in patients in the MCS compared to those in the VS/UWS, significantly so in the NREM2 phase (p < 0.05), with a positive correlation with the CRS-R score and diagnostic accuracy of 75.2%. The δ-band power spectral density [PSD(δ)] in the patients in the MCS was lower than that in those in the VS/UWS, significantly so in the waking phase (p < 0.05), and it was negatively correlated with the CRS-R score, with diagnostic accuracy of 71.5%. Conclusion Polysomnography for the VS/UWS and MCS revealed significant differences, aiding in distinguishing between the two patient categories and reducing misdiagnosis rates. Notably, the HFD and PSD(δ) showed significantly better performance during wakefulness compared to sleep, while the TKEO was more prominent in the NREM2 stage. Notably, the HFD exhibited a robust correlation with the CRS-R scores, the highest diagnostic accuracy, and immense promise in the clinical diagnosis of prolonged disorders of consciousness.
Collapse
Affiliation(s)
- Keke Li
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Man Li
- School of Automation Science and Engineering, South China University of Technology, Guangzhou, China
| | - Wanqing Liu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Neurosurgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanzhi Wu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Li
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingwei Xie
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Prevention and Treatment of Brain Injuries, Zhengzhou, China
- Henan Key Laboratory of Brain Science and Brain Computer Interface Technology, Zhengzhou, China
| | - Shaolong Zhou
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Prevention and Treatment of Brain Injuries, Zhengzhou, China
- Henan Key Laboratory of Brain Science and Brain Computer Interface Technology, Zhengzhou, China
| | - Sen Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongkun Guo
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Prevention and Treatment of Brain Injuries, Zhengzhou, China
- Henan Key Laboratory of Brain Science and Brain Computer Interface Technology, Zhengzhou, China
| | - Jiahui Pan
- School of Software, South China Normal University, Nanhai Software Technology Park, Foshan, Guangdong Province, China
| | - Xinjun Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Neurosurgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Prevention and Treatment of Brain Injuries, Zhengzhou, China
- Henan Key Laboratory of Brain Science and Brain Computer Interface Technology, Zhengzhou, China
| |
Collapse
|
10
|
Song X, Hu J. How does the brain emerge from anesthesia and regain consciousness. Chin Med J (Engl) 2025; 138:298-300. [PMID: 39512228 PMCID: PMC11771597 DOI: 10.1097/cm9.0000000000003378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Indexed: 11/15/2024] Open
Affiliation(s)
- Xuejun Song
- Department of Medical Neuroscience and SUSTech Center for Pain Medicine, Southern University of Science and Technology School of Medicine, Shenzhen, Guangdong 518000, China
| | - Jiangjian Hu
- Department of Medical Neuroscience and SUSTech Center for Pain Medicine, Southern University of Science and Technology School of Medicine, Shenzhen, Guangdong 518000, China
| |
Collapse
|
11
|
Zhou Y, Huang S, Zhang T, Deng D, Huang L, Chen X. Deciphering consciousness: The role of corticothalamocortical interactions in general anesthesia. Pharmacol Res 2025; 212:107593. [PMID: 39788339 DOI: 10.1016/j.phrs.2025.107593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/09/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
General anesthesia is administered to millions of individuals each year, however, the precise mechanism by which it induces unconsciousness remains unclear. While some theories suggest that anesthesia shares similarities with natural sleep, targeting sleep-promoting areas and inhibiting arousal nuclei, recent research indicates a more complex process. Emerging evidence highlights the critical role of corticothalamocortical circuits, which are involved in higher cognitive functions, in controlling arousal states and modulating transitions between different conscious states during anesthesia. The administration of general anesthetics disrupts connectivity within these circuits, resulting in a reversible state of unconsciousness. This review elucidates how anesthetics impair corticothalamocortical interactions, thereby affecting the flow of information across various cortical layers and disrupting higher-order cognitive functions while preserving basic sensory processing. Additionally, the role of the prefrontal cortex in regulating arousal through both top-down and bottom-up pathways was examined. These findings highlight the intricate interplay between the cortical and subcortical networks in maintaining and restoring consciousness under anesthesia, offering potential therapeutic targets for enhancing anesthesia management.
Collapse
Affiliation(s)
- Yuxi Zhou
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Shiqian Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Daling Deng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Li Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| |
Collapse
|
12
|
Ge D, Han C, Liu C, Meng Z. Neural Oscillations in the Somatosensory and Motor Cortex Distinguish Dexmedetomidine-Induced Anesthesia and Sleep in Rats. CNS Neurosci Ther 2025; 31:e70262. [PMID: 39963924 PMCID: PMC11833454 DOI: 10.1111/cns.70262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 01/06/2025] [Accepted: 01/27/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Anesthesia is featured by behavioral and physiological characteristics such as decreased sensory and motor function, loss of consciousness, etc. Some anesthetics such as dexmedetomidine (DEX), induce electroencephalogram signatures close to non-rapid eye movement sleep. Studies have shown that sleep is primarily driven by the activation of subcortical sleep-promoting neural pathways. AIMS However, the neuronal level electrophysiology features of anesthesia and how they differ from sleep is still not fully understood. MATERIALS AND METHODS In the present study, we recorded neuronal activity simultaneously from somatosensory cortex (S1) and motor cortex (M1) during awake, sleep, and DEX-induced anesthesia in rats. RESULTS The results show that DEX increased local field potential (LFP) power across a relatively wide band (1-25 Hz) in both S1 and M1. The coherence between S1 LFP and M1 LFP increased significantly in the delta and alpha bands. Power spectrum analysis during DEX-induced anesthesia revealed relatively high power in the delta and alpha bands, but low power in the theta and beta bands. Overall, the firing rate of individual neurons decreased after DEX. Correlation analysis of firing rate and LFP power indicate that more neurons were correlated, either positively or negatively, with LFPs during DEX-induced anesthesia compared to sleep. DISCUSSION Although these results showed enhancement of cortical LFP power in both DEX-induced anesthesia and sleep, different patterns of spike-field correlation suggest that the two states may be regulated by different cortical mechanisms. CONCLUSION Distinguishing anesthesia from sleep with neural oscillations could lead to more personalized, safer, and more effective approaches to managing consciousness in medical settings, with the potential for broad applications in neuroscience and clinical practice.
Collapse
Affiliation(s)
- Dengyun Ge
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Chuanliang Han
- School of Biomedical Sciences and Gerald Choa Neuroscience InstituteThe Chinese University of Hong KongHong KongSARChina
| | - Chang Liu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- CAS Key Laboratory of Brain Connectome and ManipulationChinese Academy of SciencesShenzhenChina
| | - Zhiqiang Meng
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- CAS Key Laboratory of Brain Connectome and ManipulationChinese Academy of SciencesShenzhenChina
| |
Collapse
|
13
|
Wu JY, Wang W, Dai XY, He S, Song FH, Gao SJ, Zhang LQ, Li DY, Liu L, Liu DQ, Zhou YQ, Zhang P, Tian B, Mei W. Regulation of states of consciousness by supramammillary nucleus glutamatergic neurones during sevoflurane anaesthesia in mice. Br J Anaesth 2025; 134:425-440. [PMID: 39645516 DOI: 10.1016/j.bja.2024.10.023] [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: 03/29/2024] [Revised: 08/25/2024] [Accepted: 10/03/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND The supramammillary nucleus (SuM), located in the caudal hypothalamus, includes wake-promoting glutamatergic neurones. Their potential role in regulating states of consciousness during general anaesthesia remains unknown. METHODS We used in vivo fibre photometry, c-Fos staining, chemogenetic and optogenetic manipulations, and electroencephalography/electromyography to explore the roles of glutamatergic SuM neurones (SuMVglut2 neurones) at different phases of sevoflurane anaesthesia. Rabies-mediated retrograde and anterograde tract tracing were used to investigate the monosynaptic glutamatergic inputs from the medial septum (MS) to SuM. Their roles in sevoflurane anaesthesia were investigated by in vivo fibre photometry and optogenetic manipulations. RESULTS The population activity of SuMVglut2 neurones decreased at loss of consciousness but increased during recovery of consciousness under sevoflurane anaesthesia. Their activity also decreased during suppression but increased during bursts in sevoflurane-induced burst-suppression oscillations. Activating SuMVglut2 neurones chemogenetically or optogenetically decreased sensitivity to sevoflurane, induced behavioural arousal and cortical activation during continuous steady-state anaesthesia, and stable burst-suppression oscillations under sevoflurane. In contrast, chemogenetic or optogenetic inhibition of SuMVglut2 neurones increased sensitivity to sevoflurane or intensified cortical inhibition during sevoflurane anaesthesia. Retrograde and anterograde tracing verified monosynaptic projections from MSVglut2 neurones to SuMVglut2 neurones. The activity of MSVglut2 SuM terminals increased during loss of consciousness but recovered during recovery of consciousness. Optogenetic activation or inhibition of MSVglut2 SuM terminals induced cortical activation or inhibition, respectively, during sevoflurane anaesthesia. CONCLUSIONS Activation of SuMVglut2 neurones or the glutamatergic septo-supramammillary circuit induces behavioural arousal and cortical activation during sevoflurane anaesthesia.
Collapse
Affiliation(s)
- Jia-Yi Wu
- Department of Anaesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anaesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anaesthesia, Tongji Hospital, China
| | - Wei Wang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin-Yi Dai
- Department of Anaesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anaesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anaesthesia, Tongji Hospital, China
| | - Si He
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan-He Song
- Department of Anaesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anaesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anaesthesia, Tongji Hospital, China
| | - Shao-Jie Gao
- Department of Anaesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anaesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anaesthesia, Tongji Hospital, China
| | - Long-Qing Zhang
- Department of Anaesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anaesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anaesthesia, Tongji Hospital, China
| | - Dan-Yang Li
- Department of Anaesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anaesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anaesthesia, Tongji Hospital, China
| | - Lin Liu
- Department of Anaesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anaesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anaesthesia, Tongji Hospital, China
| | - Dai-Qiang Liu
- Department of Anaesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anaesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anaesthesia, Tongji Hospital, China
| | - Ya-Qun Zhou
- Department of Anaesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anaesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anaesthesia, Tongji Hospital, China
| | - Pei Zhang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Tian
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wei Mei
- Department of Anaesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anaesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anaesthesia, Tongji Hospital, China.
| |
Collapse
|
14
|
Li D, Hudetz AG. Anesthesia alters complexity of spontaneous and stimulus-related neuronal firing patterns in rat visual cortex. Neuroscience 2025; 565:440-456. [PMID: 39631661 DOI: 10.1016/j.neuroscience.2024.11.076] [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: 08/27/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Complexity of neuronal firing patterns may serve as an indicator of sensory information processing across different states of consciousness. Recent studies have shown that spontaneous changes in brain states can occur during general anesthesia, which may influence neuronal complexity and the state of consciousness. In this study, we investigated how the firing patterns of cortical neurons, both at rest and during visual stimulation, are affected by spontaneously changing brain states under varying levels of anesthesia. Extracellular unit activity was measured in the primary visual cortex of unrestrained rats as the inhaled concentration of desflurane was incrementally reduced to 6%, 4%, 2%, and 0%. Using dimensionality reduction and density-based clustering on individual unit activities, we identified five distinct population states, which underwent dynamic transitions independent of the anesthetic level during both resting and stimulus conditions. One population state that occurred mainly in deep anesthesia exhibited a paradoxically increased number of active neurons and asynchronous spiking, suggesting a spontaneous reversal towards an awake-like condition. However, this was contradicted by the observation of low neuronal complexity in both spontaneous and stimulus-related spike activity, which more closely aligns with unconsciousness. Our findings reveal that transient neuronal states with distinct spiking patterns can emerge in visual cortex at constant anesthetic concentrations. The reduced complexity in states associated with deep anesthesia likely indicates a disruption of conscious sensory information processing.
Collapse
Affiliation(s)
- Duan Li
- Center for Consciousness Science, Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Anthony G Hudetz
- Center for Consciousness Science, Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
15
|
Zhang Q, Ke J, Cui G, Qian S, Qian W, Moon SW, Sun Y, Huang T, Qin Z. The neural ensembles activated by propofol and isoflurane anesthesia across the whole mouse brain. Neurosci Lett 2025; 846:138080. [PMID: 39662772 DOI: 10.1016/j.neulet.2024.138080] [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: 08/14/2024] [Revised: 12/07/2024] [Accepted: 12/07/2024] [Indexed: 12/13/2024]
Abstract
General anesthesia has been widely used in surgical procedures. Propofol and isoflurane are the most commonly used injectable and inhaled anesthetics, respectively. The various adverse effects induced by propofol and isoflurane are highly associated with the anesthetic-dependent change of brain activities. In this work, we aim to delineate a brain-wide neuronal activity landscape of injectable versus inhaled anesthetics to understand the neural basis underlying the different physiological effects induced by these two major types of anesthetics. Through detailed scanning of the whole mouse brain subjected to propofol or isoflurane anesthesia, in total, we identified 17 subcortical regions, 3 of which (anterodorsal preoptic nucleus, ADP; lateral habenular, LHb; inferior olivary nucleus, ION) were specifically activated by propofol, and 3 (ventral part of the lateral septum, LSV; the intermediate part of the lateral septum, LSI; the solitary tract nucleus, Sol) were specifically activated by isoflurane, with the remaining 11 were activated by both two anesthetics. Moreover, within the 17 brain regions, ADP, SubCV (subcoeruleus nucleus, ventral part), PCRtA (parvicellular reticular nucleus, alpba part) and ION were newly identified that activated by propofol or isoflurane, respectively. By using Targeted Recombination in Active Populations (TRAP) technique, we further showed that propofol and isoflurane largely activate the same group of neurons in supraoptic nucleus (SON), but activate different groups of neurons in central amygdala (CeA). Our results reveals the neural ensembles activated by injectable and inhaled anesthetics, and provides detailed anatomical references for future studies on general anesthesia.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong 510515, China; Department of Anesthesiology, Shenzhen University General Hospital and Shenzhen University Academy of Clinical Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Jin Ke
- CAS Key Laboratory of Brain Connectome and Manipulation, Institute of Brain Cognition and Brain Disease, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guangfu Cui
- CAS Key Laboratory of Brain Connectome and Manipulation, Institute of Brain Cognition and Brain Disease, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Shen Qian
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China
| | - Weixin Qian
- CAS Key Laboratory of Brain Connectome and Manipulation, Institute of Brain Cognition and Brain Disease, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sun-Wook Moon
- CAS Key Laboratory of Brain Connectome and Manipulation, Institute of Brain Cognition and Brain Disease, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Yanyan Sun
- Department of Anesthesiology, Shenzhen University General Hospital and Shenzhen University Academy of Clinical Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Tianwen Huang
- CAS Key Laboratory of Brain Connectome and Manipulation, Institute of Brain Cognition and Brain Disease, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; Shenzhen Key Laboratory of Drug Addiction, Shenzhen-Hong Kong Institute of Brain Science, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| | - Zaisheng Qin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
16
|
Zhang L, Ke Z, Zhang N, Wang D, Zhou L. Repeated sevoflurane exposure causes hypomyelination in the prefrontal cortex of adult male mice. Sci Rep 2025; 15:1546. [PMID: 39789243 PMCID: PMC11718107 DOI: 10.1038/s41598-025-85834-1] [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: 10/26/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025] Open
Abstract
As one of the most commonly used general anesthetics (GAs) in surgery, numerous studies have demonstrated the detrimental effects of sevoflurane exposure on myelination in the developing and elderly brain. However, the impact of sevoflurane exposure on intact myelin structure in the adult brain is barely discovered. Here, we show that repeated sevoflurane exposure, but not single exposure, causes hypomyelination and abnormal ultrastructure of myelin sheath in the prefrontal cortex (PFC) of adult male mice, which is considered as a critical brain region for general anesthesia mediated consciousness change. Furthermore, disrupted proliferation of oligodendrocyte precursor cells (OPCs) contributes to repeated sevoflurane exposure-induced myelin defect. This may be owing to an accumulated tuberous sclerosis complex 1 (TSC1) expression and inhibition of mammalian target of rapamycin (mTOR) signaling, leading to the unbalance of TSC1-mTORC1 activity after repeated sevoflurane exposure, which is critical for proper myelination of the central nervous system (CNS). Moreover, repeated sevoflurane exposure aggregates myelination defect in the cuprizone-induced demyelination model. Together, our present work establishes the role of sevoflurane exposure in myelin integrity in the PFC of the adult male mice and provides a new insight to elucidate the mechanism of GAs-induced brain dysfunctions.
Collapse
Affiliation(s)
- Linyong Zhang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Zhidan Ke
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of Education, Zunyi Medical University, Zunyi, 563003, China
| | - Ning Zhang
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of Education, Zunyi Medical University, Zunyi, 563003, China
| | - Dechuan Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Liang Zhou
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of Education, Zunyi Medical University, Zunyi, 563003, China.
| |
Collapse
|
17
|
Oestreicher D, Malpede AM, Reitmeier A, Bräuer CP, Schoch L, Strenzke N, Pangrsic T. Noise-induced ribbon synapse loss in the mouse basal cochlear region does not reduce inner hair cell exocytosis. Front Cell Neurosci 2025; 18:1523978. [PMID: 39839350 PMCID: PMC11747652 DOI: 10.3389/fncel.2024.1523978] [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: 11/06/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
Noise-induced hearing loss is one of the most common forms of hearing loss in adults and also one of the most common occupational diseases. Extensive previous work has shown that the highly sensitive synapses of the inner hair cells (IHCs) may be the first target for irreparable damage and permanent loss in the noise-exposed cochlea, more precisely in the cochlear base. However, how such synaptic loss affects the synaptic physiology of the IHCs in this particularly vulnerable part of the cochlea has not yet been investigated. To address this question, we exposed 3-4-week-old C57BL/6J mice to 8-16 kHz noise for 2 h under isoflurane anesthesia. We then employed hearing measurements, immunohistochemistry and patch-clamp to assess IHC synaptic function. Two noise sound pressure levels (SPLs) were used to evoke acute hearing threshold elevations with different levels of recovery 2 weeks post-exposure. Regardless of noise intensity, the exposure resulted in a loss of approximately 25-36% of ribbon synapses in the basal portions of the cochlea that persisted 2 weeks after exposure. Perforated patch-clamp recordings were made in the IHCs of the basal regions of the cochlea where the greatest synaptic losses were observed. Depolarization-evoked calcium currents in IHCs 2 weeks after exposure were slightly but not significantly smaller as compared to controls from age-matched non-exposed animals. Exocytic changes monitored as changes in membrane capacitance did not follow that trend and remained similar to controls despite significant loss of ribbons, likely reflecting increased exocytosis at the remaining synapses. Additionally, we report for the first time that acute application of isoflurane reduces IHC calcium currents, which may have implications for noise-induced IHC synaptic loss.
Collapse
Affiliation(s)
- David Oestreicher
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Alfonso Mauro Malpede
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Annalena Reitmeier
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Carolin Paula Bräuer
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Laura Schoch
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Nicola Strenzke
- Auditory Systems Physiology Group, Institute for Auditory Neuroscience, InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Tina Pangrsic
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| |
Collapse
|
18
|
Li J, Wei Y, Xiang J, Zhang D. Role of the ventral tegmental area in general anesthesia. Eur J Pharmacol 2025; 986:177145. [PMID: 39566814 DOI: 10.1016/j.ejphar.2024.177145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/22/2024]
Abstract
The ventral tegmental area (VTA), located in the midbrain, plays a pivotal role in the regulation of many important behaviors, such as reward, addiction, aversion, memory, learning, and sleep-wakefulness cycles. The majority of VTA neurons are dopaminergic neurons, although there is a significant proportion of GABAergic neurons and few glutamatergic neurons. These neuronal types project to different brain regions, thus mediating various biological functions. Therefore, the diverse roles of the VTA might depend on its heterogeneous neuronal types and projecting circuits. General anesthesia and sleep-wakefulness cycles share the feature of reversible loss of consciousness, and several common neural mechanisms underlie these two conditions. In addition to the well-known regulatory role of VTA in sleep-wakefulness, emerging evidence has demonstrated that VTA activity is also associated with promoting emergence from general anesthesia. Herein, we reviewed the literature and summarized the evidence regarding the modulation of the VTA by general anesthesia in rodents, which will improve the understanding of the modulatory mechanism of the VTA in general anesthesia.
Collapse
Affiliation(s)
- Jia Li
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430048, China.
| | - Yiyong Wei
- Department of Anesthesiology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518100, China
| | - Jiaxin Xiang
- Department of Anesthesiology, Weill Cornell Medicine, New York, 10065, USA
| | - Donghang Zhang
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430048, China; Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
19
|
Fleiner AS, Kolnier D, Hagger-Vaughan N, Ræder J, Storm JF. Effects of ketamine and propofol on muscarinic plateau potentials in rat neocortical pyramidal cells. PLoS One 2025; 20:e0316262. [PMID: 39746093 PMCID: PMC11695037 DOI: 10.1371/journal.pone.0316262] [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] [Received: 06/10/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Propofol and ketamine are widely used general anaesthetics, but have different effects on consciousness: propofol gives a deeply unconscious state, with little or no dream reports, whereas vivid dreams are often reported after ketamine anaesthesia. Ketamine is an N-methyl-D-aspartate (NMDA) receptor antagonist, while propofol is a γ-aminobutyric-acid (GABAA) receptor positive allosteric modulator, but these mechanisms do not fully explain how these drugs alter consciousness. Most previous in vitro studies of cellular mechanisms of anaesthetics have used brain slices or neurons in a nearly "comatose" state, because no "arousing" neuromodulators were added. Here we tested mechanisms of anaesthetics in rat medial prefrontal cortex (mPFC) slices after bath-applying the cholinergic agonist muscarine to partly mimic an "aroused-like" state, using whole-cell patch-clamp recordings from layer 2/3 pyramidal cells (L2/3PCs). According to leading theories of access consciousness and working memory, L2/3PCs are particularly important for these cognitive functions. We found that muscarine induced long-lasting depolarising plateau potentials (PPs) and spiking following brief depolarising current injections in the L2/3PCs. After 2 hours of pre-incubation with ketamine or propofol, the muscarine-induced PPs were altered in seemingly different ways: 3 μM propofol reduced the PPs and (significantly) spiking, whereas 20 μM ketamine seemed to enhance PPs and spiking (non-significantly). Brief wash-in of these drug concentrations failed to induce such effects, probably due to insufficient equilibration by diffusion in the slices. In contrast, pre-incubation with a high dose (100 μM) of ketamine suppressed the PPs and spiking. We discuss whether the apparently different effects on PPs may possibly be related to contrasting clinical effects: ketamine causing atypical anaesthesia with vivid, "psychedelic" dreaming while propofol causes less dreaming.
Collapse
Affiliation(s)
- Anne S. Fleiner
- Department of Molecular Medicine, Brain Signalling Laboratory, Institute of Basic Medical Sciences, Section for Physiology, University of Oslo, Oslo, Norway
| | - Daniel Kolnier
- Department of Molecular Medicine, Brain Signalling Laboratory, Institute of Basic Medical Sciences, Section for Physiology, University of Oslo, Oslo, Norway
| | - Nicholas Hagger-Vaughan
- Department of Molecular Medicine, Brain Signalling Laboratory, Institute of Basic Medical Sciences, Section for Physiology, University of Oslo, Oslo, Norway
| | - Johan Ræder
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Johan F. Storm
- Department of Molecular Medicine, Brain Signalling Laboratory, Institute of Basic Medical Sciences, Section for Physiology, University of Oslo, Oslo, Norway
| |
Collapse
|
20
|
Souza GMPR, Stornetta DS, Abbott SBG. Interactions between Arousal State and CO 2 Determine the Activity of Central Chemoreceptor Neurons That Drive Breathing. J Neurosci 2025; 45:e1587242024. [PMID: 39510833 DOI: 10.1523/jneurosci.1587-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/11/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
The homeostatic regulation of pulmonary ventilation, and ultimately arterial PCO2, depends on interactions between respiratory chemoreflexes and arousal state. The ventilatory response to CO2 is triggered by neurons in the retrotrapezoid nucleus (RTN) that function as sensors of central pH, which can be identified in adulthood by the expression of Phox2b and neuromedin B. Here, we examine the dynamic response of genetically defined RTN neurons to hypercapnia and arousal state in freely behaving adult male and female mice using the calcium indicator jGCaMP7 and fiber photometry. We found that hypercapnia vigorously activates RTN neurons with a low CO2 recruitment threshold and with response kinetics that match respiratory activity whereas hypoxia had little effect. RTN activity increased transiently during wakefulness and respiratory-related arousals and rose persistently during rapid eye movement sleep, and their CO2 response persisted under anesthesia. Complementary studies using inhibitory optogenetics show that RTN activity supports eupneic breathing under anesthesia as well as during states of high arousal, but their activity is redundant for voluntary breathing patterns. Collectively, this study demonstrates that CO2-activated RTN neurons are exquisitely sensitive to the arousal state, which determines their contribution to alveolar ventilation in relation to arterial PCO2.
Collapse
Affiliation(s)
- George M P R Souza
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| | - Daniel S Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| | - Stephen B G Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| |
Collapse
|
21
|
Dai W, Wang T, Li Y, Yang Y, Zhang Y, Wu Y, Zhou T, Yu H, Li L, Wang Y, Wang G, Xing D. Cortical direction selectivity increases from the input to the output layers of visual cortex. PLoS Biol 2025; 23:e3002947. [PMID: 39777916 PMCID: PMC11709279 DOI: 10.1371/journal.pbio.3002947] [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] [Received: 05/26/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
Sensitivity to motion direction is a feature of visual neurons that is essential for motion perception. Recent studies have suggested that direction selectivity is re-established at multiple stages throughout the visual hierarchy, which contradicts the traditional assumption that direction selectivity in later stages largely derives from that in earlier stages. By recording laminar responses in areas 17 and 18 of anesthetized cats of both sexes, we aimed to understand how direction selectivity is processed and relayed across 2 successive stages: the input layers and the output layers within the early visual cortices. We found a strong relationship between the strength of direction selectivity in the output layers and the input layers, as well as the preservation of preferred directions across the input and output layers. Moreover, direction selectivity was enhanced in the output layers compared to the input layers, with the response strength maintained in the preferred direction but reduced in other directions and under blank stimuli. We identified a direction-tuned gain mechanism for interlaminar signal transmission, which likely originated from both feedforward connections across the input and output layers and recurrent connections within the output layers. This direction-tuned gain, coupled with nonlinearity, contributed to the enhanced direction selectivity in the output layers. Our findings suggest that direction selectivity in later cortical stages partially inherits characteristics from earlier cortical stages and is further refined by intracortical connections.
Collapse
Affiliation(s)
- Weifeng Dai
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Tian Wang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yang Li
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Yi Yang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Yange Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Yujie Wu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Tingting Zhou
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Hongbo Yu
- School of Life Sciences, State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Liang Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yizheng Wang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Gang Wang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Dajun Xing
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| |
Collapse
|
22
|
Baron M, Vaso K, Ibraheem A, Minert A, Devor M. Molecular and cellular targets of GABAergic anesthetics in the mesopontine tegmentum that enable pain-free surgery. Pain 2024:00006396-990000000-00798. [PMID: 39792547 DOI: 10.1097/j.pain.0000000000003504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/27/2024] [Indexed: 01/12/2025]
Abstract
ABSTRACT The mesopontine tegmental anesthesia area (MPTA) is a focal brainstem locus which, when exposed to GABAergic agents, induces brain-state transitioning from wakefulness to unconsciousness. Correspondingly, MPTA lesions render animals relatively insensitive to GABAergic anesthetics delivered systemically. Using chemogenetics, we recently identified a neuronal subpopulation within the MPTA whose excitation induces this same pro-anesthetic effect. However, very few of these "effector-neurons" express synaptic γ2-containing GABAA receptor isoforms and none express extrasynaptic δ-subunit containing receptors, suggesting that they are not the direct cellular target of GABAergic agents. Here we used pharmacological tools in rats to define the molecular target(s) of GABAergics in the MPTA. GABA microinjected into the MPTA at nanomolar concentrations, selective for GABAAδ-Rs, proved to be pro-anesthetic as was blocking GABA reuptake. Likewise, low-concentration gaboxadol/THIP, also selective for GABAAδ-Rs, was effective, whereas benzodiazepines and zolpidem, which selectively target GABAAγ2-Rs, were not. The GABAergic anesthetics pentobarbital and propofol proved pro-anesthetic when applied to the MPTA at the low concentrations present in the brain after systemic dosing. Glycinergic agonists which are inhibitory, but infective on GABAAδ-Rs, and other non-GABAergic agonists tested, were at most only marginally effective. We conclude that GABAAδ-Rs are the primary molecular target of GABAergic anesthetics in the MPTA. Immunolabeling revealed that this GABAA-R isoform is expressed exclusively by a distinct subpopulation of MPTA "δ-cells" that reside in close apposition to effector neurons. This suggests that during wakefulness, δ-cells serve as inhibitory interneurons which, when silenced by GABAergic agents, disinhibit (excite) the effector-neurons, triggering transition to unconsciousness.
Collapse
Affiliation(s)
- Mark Baron
- Department of Cell and Developmental Biology, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kristina Vaso
- Department of Cell and Developmental Biology, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Angham Ibraheem
- Department of Cell and Developmental Biology, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Anne Minert
- Department of Cell and Developmental Biology, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Marshall Devor
- Department of Cell and Developmental Biology, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
- Center for Research on Pain, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
23
|
Manohara N, Ferrari A, Greenblatt A, Berardino A, Peixoto C, Duarte F, Moyiaeri Z, Robba C, Nascimento F, Kreuzer M, Vacas S, Lobo FA. Electroencephalogram monitoring during anesthesia and critical care: a guide for the clinician. J Clin Monit Comput 2024:10.1007/s10877-024-01250-2. [PMID: 39704777 DOI: 10.1007/s10877-024-01250-2] [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: 10/21/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024]
Abstract
Perioperative anesthetic, surgical and critical careinterventions can affect brain physiology and overall brain health. The clinical utility of electroencephalogram (EEG) monitoring in anesthesia and intensive care settings is multifaceted, offering critical insights into the level of consciousness and depth of anesthesia, facilitating the titration of anesthetic doses, and enabling the detection of ischemic events and epileptic activity. Additionally, EEG monitoring can aid in predicting perioperative neurocognitive disorders, assessing the impact of systemic insults on cerebral function, and informing neuroprognostication. This review provides a comprehensive overview of the fundamental principles of electroencephalography, including the foundations of processed and quantitative electroencephalography. It further explores the characteristic EEG signatures associated wtih anesthetic drugs, the interpretation of the EEG data during anesthesia, and the broader clinical benefits and applications of EEG monitoring in both anesthetic practice and intensive care environments.
Collapse
Affiliation(s)
- Nitin Manohara
- Division of Anesthesiology, Cleveland Clinic Abu Dhabi, Integrated Hospital Care Institute, Abu Dhabi, United Arab Emirates
| | | | - Adam Greenblatt
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Andrea Berardino
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | | | - Flávia Duarte
- Department of Anesthesiology, Hospital Garcia de Orta, Almada, Portugal
| | - Zahra Moyiaeri
- Division of Anesthesiology, Cleveland Clinic Abu Dhabi, Integrated Hospital Care Institute, Abu Dhabi, United Arab Emirates
| | | | - Fabio Nascimento
- Department of Neurology, Washington University in St Louis, St Louis, MO, USA
| | - Matthias Kreuzer
- Department of Anesthesiology and Intensive Care Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Susana Vacas
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Francisco A Lobo
- Division of Anesthesiology, Cleveland Clinic Abu Dhabi, Integrated Hospital Care Institute, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
24
|
Grigis A, Gomez C, Frouin V, Duchesnay E, Uhrig L, Jarraya B. Revisiting the standard for modeling functional brain network activity: Application to consciousness. PLoS One 2024; 19:e0314598. [PMID: 39680526 DOI: 10.1371/journal.pone.0314598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
Functional connectivity (FC) of resting-state fMRI time series can be estimated using methods that differ in their temporal sensitivity (static vs. dynamic) and the number of regions included in the connectivity estimation (derived from a prior atlas). This paper presents a novel framework for identifying and quantifying resting-state networks using resting-state fMRI recordings. The study employs a linear latent variable model to generate spatially distinct brain networks and their associated activities. It specifically addresses the atlas selection problem, and the statistical inference and multivariate analysis of the obtained brain network activities. The approach is demonstrated on a dataset of resting-state fMRI recordings from monkeys under different anesthetics using static FC. Our results suggest that two networks, one fronto-parietal and cingular and another temporo-parieto-occipital (posterior brain) strongly influences shifts in consciousness, especially between anesthesia and wakefulness. Interestingly, this observation aligns with the two prominent theories of consciousness: the global neural workspace and integrated information theories of consciousness. The proposed method is also able to decipher the level of anesthesia from the brain network activities. Overall, we provide a framework that can be effectively applied to other datasets and may be particularly useful for the study of disorders of consciousness.
Collapse
Affiliation(s)
- Antoine Grigis
- Université Paris-Saclay, CEA, NeuroSpin, Gif-sur-Yvette, France
| | - Chloé Gomez
- Université Paris-Saclay, CEA, NeuroSpin, Gif-sur-Yvette, France
- Cognitive Neuroimaging Unit, Institut National de la Santé et de la Recherche Médicale U992, Gif-sur-Yvette, France
| | - Vincent Frouin
- Université Paris-Saclay, CEA, NeuroSpin, Gif-sur-Yvette, France
| | | | - Lynn Uhrig
- Université Paris-Saclay, CEA, NeuroSpin, Gif-sur-Yvette, France
- Cognitive Neuroimaging Unit, Institut National de la Santé et de la Recherche Médicale U992, Gif-sur-Yvette, France
| | - Béchir Jarraya
- Université Paris-Saclay, CEA, NeuroSpin, Gif-sur-Yvette, France
- Cognitive Neuroimaging Unit, Institut National de la Santé et de la Recherche Médicale U992, Gif-sur-Yvette, France
- Université Paris-Saclay (UVSQ), Neuroscience Pole, Foch Hospital, Suresnes, France
| |
Collapse
|
25
|
Chen H, Liu C, Liu J, Yuan C, He H, Zhang Y, Yu S, Luo T, Shen W, Yu T. Zona Incerta GABAergic Neurons Facilitate Emergence from Isoflurane Anesthesia in Mice. Neurochem Res 2024; 49:3297-3307. [PMID: 39312079 PMCID: PMC11502554 DOI: 10.1007/s11064-024-04230-9] [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: 05/28/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 10/25/2024]
Abstract
The zona incerta (ZI) predominantly consists of gamma-aminobutyric acid (GABAergic) neurons, located adjacent to the lateral hypothalamus. GABA, acting on GABAA receptors, serves as a crucial neuromodulator in the initiation and maintenance of general anesthesia. In this study, we aimed to investigate the involvement of ZI GABAergic neurons in the general anesthesia process. Utilizing in-vivo calcium signal optical fiber recording, we observed a decrease in the activity of ZI GABAergic neurons during isoflurane anesthesia, followed by a significant increase during the recovery phase. Subsequently, we selectively ablated ZI GABAergic neurons to explore their role in general anesthesia, revealing no impact on the induction of isoflurane anesthesia but a prolonged recovery time, accompanied by a reduction in delta-band power in mice under isoflurane anesthesia. Finally, through optogenetic activation/inhibition of ZI GABAergic neurons during isoflurane anesthesia, we discovered that activation of these neurons facilitated emergence without affecting the induction process, while inhibition delayed emergence, leading to fluctuations in delta band activity. In summary, these findings highlight the involvement of ZI GABAergic neurons in modulating the emergence of isoflurane anesthesia.
Collapse
Affiliation(s)
- Hong Chen
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Department of Anesthesiology, KweiChow Moutai Hospital, Renhuai, Guizhou, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Chengxi Liu
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Junxiao Liu
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Department of Anesthesiology, Zunyi Hospital of Traditional Chinese Medicine, Zunyi, Guizhou, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Chengdong Yuan
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Department of Anesthesiology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Haifeng He
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Yu Zhang
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
| | - Shouyang Yu
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Tianyuan Luo
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Wei Shen
- School of Life Science and Technology and Shanghai Institute of Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Tian Yu
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China.
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
26
|
Chen G, Xu M, Chen Z, Yang F. Clinical applications of small-molecule GABA AR modulators for neurological disorders. Bioorg Chem 2024; 153:107983. [PMID: 39581171 DOI: 10.1016/j.bioorg.2024.107983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/31/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024]
Abstract
Gamma-aminobutyric acid type A receptor (GABAAR) modulators are crucial in treating neurological and psychiatric disorders, including epilepsy, anxiety, insomnia, and depression. This review examines the synthetic approaches and clinical applications of representative small-molecule GABAAR modulators. Benzodiazepines, such as Diazepam, are well-known positive allosteric modulators (PAMs) that enhance GABAAR function, providing therapeutic effects but also associated with side effects like sedation and dependence. Non-benzodiazepine modulators, including Z-drugs like Zolpidem and Zaleplon, offer improved selectivity for the α1 subunit of GABAAR, reducing some of these side effects. Neurosteroids such as allopregnanolone and its synthetic analogs, including Brexanolone, have emerged as potent GABAAR modulators with applications in conditions like postpartum depression and refractory epilepsy. Advances in molecular biology and pharmacology have facilitated the development of isoform-specific modulators, potentially reducing off-target effects and enhancing therapeutic profiles. Additionally, combining GABAAR modulators with other therapeutic agents has shown promise in enhancing efficacy and minimizing side effects. This review highlights the design strategies, pharmacodynamics, clinical efficacy, and safety profiles of these compounds, emphasizing the opportunities for developing novel GABAAR modulators with improved therapeutic outcomes.
Collapse
Affiliation(s)
- Guangyong Chen
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meiling Xu
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhuo Chen
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Fuwei Yang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
27
|
Li W, Xu B, Huang Y, Wang X, Yu D. Rodent models in sensorineural hearing loss research: A comprehensive review. Life Sci 2024; 358:123156. [PMID: 39442868 DOI: 10.1016/j.lfs.2024.123156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Sensorineural hearing loss (SNHL) constitutes a major global health challenge, affecting millions of individuals and substantially impairing social integration and quality of life. The complexity of the auditory system and the multifaceted nature of SNHL necessitate advanced methodologies to understand its etiology, progression, and potential therapeutic interventions. This review provides a comprehensive overview of the current animal models used in SNHL research, focusing on their selection based on specific characteristics and their contributions to elucidating pathophysiological mechanisms and evaluating novel treatment strategies. It discusses the most commonly used rodent models in hearing research, including mice, rats, guinea pigs, Mongolian gerbils, and chinchillas. Through a comparative analysis, this review underscores the importance of selecting models that align with specific research objectives in SNHL studies, discussing the advantages and limitations of each model. By advocating for a multidisciplinary approach that leverages the strengths of various animal models with technological advancements, this review aims to facilitate significant advancements in the prevention, diagnosis, and treatment of sensorineural hearing loss.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200100, PR China
| | - Baoying Xu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Yuqi Huang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Xueling Wang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200100, PR China
| | - Dehong Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China.
| |
Collapse
|
28
|
Bruschetta G, Leonardi F, Licata P, Iannelli NM, Fernàndez-Parra R, Bruno F, Messina L, Costa GL. Oxidative stress in relation to serotonin under general anaesthesia in dogs undergoing ovariectomy. Vet Q 2024; 44:1-8. [PMID: 39028214 PMCID: PMC11262201 DOI: 10.1080/01652176.2024.2379319] [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: 03/07/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024] Open
Abstract
Abdominal surgery such as ovariectomy is a traumatic event that can cause oxidative stress. The aim of the present study was to evaluate the concentration of serotonin in relation to ovariectomy-induced oxidative stress in dogs undergoing general anesthesia. Thirty-two female dogs, under general anesthesia, received meloxicam before surgery (0.2 mgkg-1 SC) and after surgery (0.1 mgkg-1 OS every 24 h). The physiological, hematological, and biochemical parameters: glycemia, aspartate transaminase (AST), alanine aminotransferase (ALT), total protein, albumin and BUN were evaluated. Oxidative stress was determined by malondialdehyde (MDA) assay, catalase (CAT), superoxide dismutase (SOD), myeloperoxidase (MPO) and butyrylcholinesterase (BuChe) at baseline, 36 and 48 h after the last administration of meloxicam. Serotonin (5-HT) concentration was also evaluated at baseline, 36 and 48 h after the last administration of meloxicam. Responses to surgical stimulus were evaluated. Physiological and hematological parameters they fell within the normal ranges for anesthetized dogs. Glycemia increased, albumin levels decreased after surgery. No rescue analgesia was required. MDA and 5-HT concentrations significantly increased from the baseline at 36 and 48 h after surgery (p < .001). 5-HT levels could be used as an indicator for oxidative stress induced by surgery and it might be employed for objectively quantifying the well-being of the surgical patient.
Collapse
Affiliation(s)
| | - Fabio Leonardi
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Patrizia Licata
- Department of Veterinary Sciences, University of Messina, Messina, Italy
| | | | | | - Fabio Bruno
- Department of Veterinary Sciences, University of Messina, Messina, Italy
| | - Laura Messina
- Department of Veterinary Sciences, University of Messina, Messina, Italy
| | | |
Collapse
|
29
|
Parajuli B, Koizumi S. Unexpected role of microglia and P2Y 12 in the induction of and emergence from anesthesia. Purinergic Signal 2024; 20:573-575. [PMID: 38724707 PMCID: PMC11554972 DOI: 10.1007/s11302-024-10014-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/26/2024] [Indexed: 11/13/2024] Open
Affiliation(s)
- Bijay Parajuli
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan.
- GLIA Center, University of Yamanashi, Yamanashi, 409-3898, Japan.
| |
Collapse
|
30
|
Pavlovič A. Touch, light, wounding: how anaesthetics affect plant sensing abilities. PLANT CELL REPORTS 2024; 43:293. [PMID: 39580775 PMCID: PMC11586303 DOI: 10.1007/s00299-024-03369-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024]
Abstract
KEY MESSAGE Anaesthetics affect not only humans and animals but also plants. Plants exposed to certain anaesthetics lose their ability to respond adequately to various stimuli such as touch, injury or light. Available results indicate that anaesthetics modulate ion channel activities in plants, e.g. Ca2+ influx. The word anaesthesia means loss of sensation. Plants, as all living creatures, can also sense their environment and they are susceptible to anaesthesia. Although some anaesthetics are often known as drugs with well-defined target to their animal/human receptors, some other are promiscuous in their binding. Both have effects on plants. Application of general volatile anaesthetics (GVAs) inhibits plant responses to different stimuli but also induces strong cellular response. Of particular interest is the ability of GVAs inhibit long-distance electrical and Ca2+ signalling probably through inhibition of GLUTAMATE RECEPTOR-LIKE proteins (GLRs), the effect which is surprisingly very similar to inhibition of nerve impulse transmission in animals or human. However, GVAs act also as a stressor for plants and can induce their own Ca2+ signature, which strongly reprograms gene expression . Down-regulation of genes encoding enzymes of chlorophyll biosynthesis and pigment-protein complexes are responsible for inhibited de-etiolation and photomorphogenesis. Vesicle trafficking, germination, and circumnutation movement of climbing plants are also strongly inhibited. On the other hand, other cellular processes can be upregulated, for example, heat shock response and generation of reactive oxygen species (ROS). Upregulation of stress response by GVAs results in preconditioning/priming and can be helpful to withstand abiotic stresses in plants. Thus, anaesthetic drugs may become a useful tool for scientists studying plant responses to environmental stimuli.
Collapse
Affiliation(s)
- Andrej Pavlovič
- Department of Biophysics, Faculty of Science, Palacký University, Šlechtitelů 27, 78371, Olomouc, Czech Republic.
| |
Collapse
|
31
|
Baron M, Devor M. Neurosteroids foster sedation by engaging tonic GABA A-Rs within the mesopontine tegmental anesthesia area (MPTA). Neurosci Lett 2024; 843:138030. [PMID: 39490574 DOI: 10.1016/j.neulet.2024.138030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/11/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Neurosteroids are endogenous molecules with anxiolytic, anticonvulsant, sleep-promoting and sedative effects. They are biosynthesized de novo within the brain, among other tissues, and are thought to act primarily as positive allosteric modulators of high-affinity extrasynaptic GABAAδ-receptors. The location of action of neurosteroids in the brain, however, remains unknown. We have demonstrated that GABAergic anesthetics act within the brainstem mesopontine tegmental anesthesia area (MPTA) to induce and maintain anesthetic loss-of-consciousness. Here we asked whether endogenous and synthetic neurosteroids might also act in the MPTA to induce their suppressive effects. Direct exposure of the MPTA to the endogenous neurosteroids pregnenolone and progesterone, their metabolites testosterone, allopregnanolone and 3α5α-THDOC, and the synthetic neurosteroids ganaxolone and alphaxalone, was found to be pro-anesthetic. Although we cannot rule out additional sites of action, results of this study suggest that the suppressive effects of neurosteroids are due, at least in part, to actions within the MPTA, presumably by recruitment of dedicated neuronal circuitry. This undermines the usual presumption that neurosteroids, like other sedatives, endogenous somnogens and anesthetics, act by nonspecific global distribution.
Collapse
Affiliation(s)
- Mark Baron
- Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Marshall Devor
- Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; Center for Research on Pain, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
32
|
Hollingworth D, Herold KF, Kelly G, Mykhaylyk VB, Xiang J, Zhang D, Wallace BA, Hemmings HC. Functionally important binding site for a volatile anesthetic in a voltage-gated sodium channel identified by X-ray crystallography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621342. [PMID: 39574657 PMCID: PMC11580951 DOI: 10.1101/2024.11.04.621342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Volatile general anesthetics are used for inhalational anesthesia in hundreds of millions of surgical procedures annually, yet their mechanisms of action remain unclear. Membrane proteins involved in cell signaling are major targets for anesthetics, and voltage-gated ion channels that mediate neurotransmission, movement, and cognition are sensitive to volatile anesthetics (VAs). In many cases, the effects produced by VAs on mammalian ion channels are reproduced in prokaryotic orthologues, providing an opportunity to investigate VA interactions at high resolution using these structurally simpler prokaryotic proteins. We utilized the bacterial voltage-gated sodium channel (VGSC) NavMs from Magnetococcus marinus to investigate its interaction with the widely used VA sevoflurane. Sevoflurane interacted directly with NavMs, producing effects consistent with multisite binding models for VA actions on their targets. We report the identification of one of these interactions at atomic detail providing the first high-resolution structure of a VA bound to a voltage-gated ion channel. The X-ray crystal structure shows sevoflurane binding to NavMs within an intramembrane hydrophobic pocket formed by residues from the voltage sensor and channel pore, domains essential for channel gating. Mutation of the dominant sevoflurane binding-site residue within this pocket, and analogous residues found in similar sites in human VGSCs, profoundly affected channel properties, supporting a critical role for this site in VGSC function. These findings provide the basis for future work to understand the role of VA interactions with VGSCs in both the anesthetic and toxic effects associated with general anesthesia.
Collapse
Affiliation(s)
| | - Karl F Herold
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Geoff Kelly
- The Medical Research Council Biomedical NMR Centre, The Francis Crick Institute, London, UK
| | - Vitaliy B Mykhaylyk
- Diamond Light Source, Harwell Science and Innovation Campus, Chilton, Didcot, UK
| | - Jiaxin Xiang
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Donghang Zhang
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - B A Wallace
- School of Natural Sciences, Birkbeck University of London, London, UK
| | - Hugh C Hemmings
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
33
|
Chen X, Cramer SR, Chan DC, Han X, Zhang N. Sequential Deactivation Across the Hippocampus-Thalamus-mPFC Pathway During Loss of Consciousness. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406320. [PMID: 39248326 PMCID: PMC11558098 DOI: 10.1002/advs.202406320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/12/2024] [Indexed: 09/10/2024]
Abstract
How consciousness is lost in states such as sleep or anesthesia remains a mystery. To gain insight into this phenomenon, concurrent recordings of electrophysiology signals in the anterior cingulate cortex and whole-brain functional magnetic resonance imaging (fMRI) are conducted in rats exposed to graded propofol, undergoing the transition from consciousness to unconsciousness. The results reveal that upon the loss of consciousness (LOC), there is a sharp increase in low-frequency power of the electrophysiological signal. Additionally, fMRI signals exhibit a cascade of deactivation across a pathway including the hippocampus, thalamus, and medial prefrontal cortex (mPFC) surrounding the moment of LOC, followed by a broader increase in brain activity across the cortex during sustained unconsciousness. Furthermore, sliding window analysis demonstrates a temporary increase in synchrony of fMRI signals across the hippocampus-thalamus-mPFC pathway preceding LOC. These data suggest that LOC may be triggered by sequential activities in the hippocampus, thalamus, and mPFC, while wide-spread activity increases in other cortical regions commonly observed during anesthesia-induced unconsciousness may be a consequence, rather than a cause of LOC. Taken together, the study identifies a cascade of neural events unfolding as the brain transitions into unconsciousness, offering insight into the systems-level neural mechanisms underpinning LOC.
Collapse
Affiliation(s)
- Xiaoai Chen
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Samuel R. Cramer
- The Neuroscience Graduate ProgramThe Huck Institutes of the Life SciencesThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Dennis C.Y. Chan
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Xu Han
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Nanyin Zhang
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- The Neuroscience Graduate ProgramThe Huck Institutes of the Life SciencesThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neurotechnology in Mental Health ResearchThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| |
Collapse
|
34
|
Lin J, Hou X, Liu Y, Cai Y, Pan J, Liao J. Elevated peripheral glutamate and upregulated expression of NMDA receptor NR1 subunit in insomnia disorder. Front Psychiatry 2024; 15:1436024. [PMID: 39435127 PMCID: PMC11491378 DOI: 10.3389/fpsyt.2024.1436024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/18/2024] [Indexed: 10/23/2024] Open
Abstract
Background The present study explored the serum glutamate (Glu), glutamine (Gln), glutamic acid dehydrogenase (GAD) concentrations and the mRNA expression levels of the N-methyl-D-aspartate receptor (NMDAR) NR1 subunit in the peripheral blood of patients with insomnia disorder (ID). To our knowledge, this is the first study showing an increase in the mRNA expression levels of the NMDAR NR1 subunit in patients with ID. Methods This study included 30 ID patients and 30 matched healthy controls. We investigated the demographic and illness information and assessed subjective sleep quality using the Pittsburgh Sleep Quality Index. The Hamilton Depression Scale-17 and Hamilton Anxiety Scale were used to evaluate the patients' symptoms of depression and anxiety, respectively. The quantifications of Glu, Gln and GAD concentrations were performed by Enzyme-linked immunosorbent assay (ELISA). Real-time PCR was used to detect the mRNA expression levels of the NMDAR NR1 subunit in peripheral blood. Results Compared with the healthy control group, the serum Glu concentrations and the mRNA expression levels of the NMDAR NR1 subunit in the ID group were significantly higher. However, there was no significant difference in Gln and GAD between the two groups. The receiver operating characteristic (ROC) analysis showed that the mRNA expression levels of the NMDAR NR1 subunit could distinguish ID patients from healthy individuals (area under the curve: 0.758; sensitivity: 73.3%; specificity: 76.7%). A negative correlation was found between the mRNA expression levels of the NMDAR NR1 subunit for age, total duration of illness, and age of first onset in the ID group, whereas a positive correlation was detected for daytime dysfunction. Conclusion Glutamatergic neurotransmission was abnormal in ID patients. Additionally, the mRNA expression levels of the NMDAR NR1 subunit appeared to have potential as a clinical biomarker for ID. However, the sample size of our study was limited, and future studies with larger sample sizes are needed to further validate and explore the mechanisms involved and to assess the reliability of the biomarker.
Collapse
Affiliation(s)
- Jingjing Lin
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Department of Psychiatry, Sleep Medicine Centre, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiaohui Hou
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yaxi Liu
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Department of Psychiatry, Sleep Medicine Centre, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yixian Cai
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Department of Psychiatry, Sleep Medicine Centre, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jiyang Pan
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Department of Psychiatry, Sleep Medicine Centre, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jiwu Liao
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
35
|
Zhou Y, Dong W, Qiu YK, Shao KJ, Zhang ZX, Yao JQ, Chen TQ, Li ZY, Zhou CR, Jiao XH, Chen Y, Lu H, Wu YQ. Regulating the activity of GABAergic neurons in the ventral pallidum alters the general anesthesia effect of propofol. Neuropharmacology 2024; 257:110032. [PMID: 38852839 DOI: 10.1016/j.neuropharm.2024.110032] [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: 03/07/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
The full mechanism of action of propofol, a commonly administered intravenous anesthetic drug in clinical practice, remains elusive. The focus of this study was the role of GABAergic neurons which are the main neuron group in the ventral pallidum (VP) closely associated with anesthetic effects in propofol anesthesia. The activity of VP GABAergic neurons following propofol anesthesia in Vgat-Cre mice was observed via detecting c-Fos immunoreactivity by immunofluorescence and western blotting. Subsequently, chemogenetic techniques were employed in Vgat-Cre mice to regulate the activity of VP GABAergic neurons. The role of VP GABAergic neurons in generating the effects of general anesthesia induced by intravenous propofol was further explored through behavioral tests of the righting reflex. The results revealed that c-Fos expression in VP GABAergic neurons in Vgat-Cre mice dramatically decreased after propofol injection. Further studies demonstrated that chemogenetic activation of VP GABAergic neurons during propofol anesthesia shortened the duration of anesthesia and promoted wakefulness. Conversely, the inhibition of VP GABAergic neurons extended the duration of anesthesia and facilitated the effects of anesthesia. The results obtained in this study suggested that regulating the activity of GABAergic neurons in the ventral pallidum altered the effect of propofol on general anesthesia.
Collapse
Affiliation(s)
- Yue Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Wei Dong
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Yong-Kang Qiu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Ke-Jie Shao
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Zi-Xin Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Jia-Qi Yao
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Tian-Qi Chen
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Zi-Yi Li
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Chen-Rui Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Xin-Hao Jiao
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Yu Chen
- Department of Anesthesiology, Liyang People's Hospital, Jiangsu Province, Liyang, China; Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Han Lu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yu-Qing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
36
|
Kimura A. Cross-modal sensitivities to auditory and visual stimulations in the first-order somatosensory thalamic nucleus. Eur J Neurosci 2024; 60:5621-5657. [PMID: 39192569 DOI: 10.1111/ejn.16510] [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: 04/10/2024] [Revised: 07/15/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024]
Abstract
The ventral posterolateral nucleus (VPL), being categorized as the first-order thalamic nucleus, is considered to be dedicated to uni-modal somatosensory processing. Cross-modal sensory interactions on thalamic reticular nucleus cells projecting to the VPL, on the other hand, suggest that VPL cells are subject to cross-modal sensory influences. To test this possibility, the effects of auditory or visual stimulation on VPL cell activities were examined in anaesthetized rats, using juxta-cellular recording and labelling techniques. Recordings were obtained from 70 VPL cells, including 65 cells responsive to cutaneous electrical stimulation of the hindpaw. Auditory or visual alone stimulation did not elicit cell activity except in three bi-modal cells and one auditory cell. Cross-modal alterations of somatosensory response by auditory and/or visual stimulation were recognized in 61 cells with regard to the response magnitude, latency (time and jitter) and/or burst spiking properties. Both early (onset) and late responses were either suppressed or facilitated, and de novo cell activity was also induced. Cross-modal alterations took place depending on the temporal interval between the preceding counterpart and somatosensory stimulations, the intensity and frequency of sound. Alterations were observed mostly at short intervals (< 200 ms) and up to 800 ms intervals. Sounds of higher intensities and lower frequencies were more effective for modulation. The susceptibility to cross-modal influences was related to cell location and/or morphology. These and previously reported similar findings in the auditory and visual thalamic nuclei suggest that cross-modal sensory interactions pervasively take place in the first-order sensory thalamic nuclei.
Collapse
Affiliation(s)
- Akihisa Kimura
- Department of Physiology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
37
|
Dai S, Ren Y, Chen L, Wu M, Wang R, Zhou Q. Machine learning-based prediction of the risk of moderate-to-severe catheter-related bladder discomfort in general anaesthesia patients: a prospective cohort study. BMC Anesthesiol 2024; 24:334. [PMID: 39300332 PMCID: PMC11411741 DOI: 10.1186/s12871-024-02720-5] [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: 05/01/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Catheter-related bladder discomfort (CRBD) commonly occurs in patients who have indwelling urinary catheters while under general anesthesia. And moderate-to-severe CRBD can lead to significant adverse events and negatively impact patient health outcomes. However, current screening studies for patients experiencing moderate-to-severe CRBD after waking from general anesthesia are insufficient. Constructing predictive models with higher accuracy using multiple machine learning techniques for early identification of patients at risk of experiencing moderate-to-severe CRBD during general anesthesia resuscitation. METHODS Eight hundred forty-six patients with indwelling urinary catheters who were resuscitated in a post-anesthesia care unit (PACU). Trained researchers used the CRBD 4-level assessment method to evaluate the severity of a patient's CRBD. They then inputted 24 predictors into six different machine learning algorithms. The performance of the models was evaluated using metrics like the area under the curve (AUC). RESULTS The AUCs of the six models ranged from 0.82 to 0.89. Among them, the RF model displayed the highest predictive ability, with an AUC of 0.89 (95%CI: 0.87, 0.91). Additionally, it achieved an accuracy of 0.93 (95%CI: 0.91, 0.95), 0.80 sensitivity, 0.98 specificity, 0.94 positive predictive value (PPV), 0.92 negative predictive value (NPV), 0.87 F1 score, and 0.07 Brier score. The logistic regression (LR) model has achieved good results (AUC:0.87) and converted into a nomogram. CONCLUSIONS The study has successfully developed a machine learning prediction model that exhibits excellent predictive capabilities in identifying patients who may develop moderate-to-severe CRBD after undergoing general anesthesia. Furthermore, the study also presents a nomogram, which serves as a valuable tool for clinical healthcare professionals, enabling them to intervene at an early stage for better patient outcomes.
Collapse
Affiliation(s)
- Suwan Dai
- Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, Zhejiang, China
| | - Yingchun Ren
- College of Data Science, Jiaxing University, Jiaxing, Zhejiang, China
| | - Lingyan Chen
- Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, Zhejiang, China
| | - Min Wu
- Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, Zhejiang, China
| | - Rong Wang
- Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, Zhejiang, China.
| | - Qinghe Zhou
- Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, Zhejiang, China.
| |
Collapse
|
38
|
Zhou P, Deng H, Zeng J, Ran H, Yu C. Unconscious classification of quantitative electroencephalogram features from propofol versus propofol combined with etomidate anesthesia using one-dimensional convolutional neural network. Front Med (Lausanne) 2024; 11:1447951. [PMID: 39359920 PMCID: PMC11445052 DOI: 10.3389/fmed.2024.1447951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024] Open
Abstract
Objective Establishing a convolutional neural network model for the recognition of characteristic raw electroencephalogram (EEG) signals is crucial for monitoring consciousness levels and guiding anesthetic drug administration. Methods This trial was conducted from December 2023 to March 2024. A total of 40 surgery patients were randomly divided into either a propofol group (1% propofol injection, 10 mL: 100 mg) (P group) or a propofol-etomidate combination group (1% propofol injection, 10 mL: 100 mg, and 0.2% etomidate injection, 10 mL: 20 mg, mixed at a 2:1 volume ratio) (EP group). In the P group, target-controlled infusion (TCI) was employed for sedation induction, with an initial effect site concentration set at 5-6 μg/mL. The EP group received an intravenous push with a dosage of 0.2 mL/kg. Six consciousness-related EEG features were extracted from both groups and analyzed using four prediction models: support vector machine (SVM), Gaussian Naive Bayes (GNB), artificial neural network (ANN), and one-dimensional convolutional neural network (1D CNN). The performance of the models was evaluated based on accuracy, precision, recall, and F1-score. Results The power spectral density (94%) and alpha/beta ratio (72%) demonstrated higher accuracy as indicators for assessing consciousness. The classification accuracy of the 1D CNN model for anesthesia-induced unconsciousness (97%) surpassed that of the SVM (83%), GNB (81%), and ANN (83%) models, with a significance level of p < 0.05. Furthermore, the mean and mean difference ± standard error of the primary power values for the EP and P groups during the induced period were as follows: delta (23.85 and 16.79, 7.055 ± 0.817, p < 0.001), theta (10.74 and 8.743, 1.995 ± 0.7045, p < 0.02), and total power (24.31 and 19.72, 4.588 ± 0.7107, p < 0.001). Conclusion Large slow-wave oscillations, power spectral density, and the alpha/beta ratio are effective indicators of changes in consciousness during intravenous anesthesia with a propofol-etomidate combination. These indicators can aid anesthesiologists in evaluating the depth of anesthesia and adjusting dosages accordingly. The 1D CNN model, which incorporates consciousness-related EEG features, represents a promising tool for assessing the depth of anesthesia. Clinical Trial Registration https://www.chictr.org.cn/index.html.
Collapse
Affiliation(s)
- Pan Zhou
- Department of Anesthesiology, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Haixia Deng
- Department of Anesthesiology, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jie Zeng
- Department of Anesthesiology, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Haosong Ran
- College of Artificial Intelligent, Chongqing University of Technology, Chongqing, China
| | - Cong Yu
- Department of Anesthesiology, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
39
|
Cheaha D, Basor N, Manor R, Hayeemasae N, Samerphob N. Sedative and hypnotic effects with cortical EEG sleep-wake profiles of Millingtonia hortensis dried flower aqueous in mice. Heliyon 2024; 10:e37531. [PMID: 39296192 PMCID: PMC11409071 DOI: 10.1016/j.heliyon.2024.e37531] [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: 04/17/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/21/2024] Open
Abstract
The ethnopharmacological relevance of the Millingtonia hortensis (M. hortensis) flower's aqueous extract lies in its traditional use as a herbal remedy in Southeast Asia. With a rich history in folk medicine, this aqueous has been esteemed for its purported sedative and anxiolytic properties. Our research delves into the scientific basis of these traditional claims, exploring the potential mechanisms underlying the observed effects of M. hortensis flower's aqueous extract on sleep promotion and mood regulation. This study aimed to explore the sleep-promoting effects of M. hortensis dried flower in mice, using an aqueous concentration equivalent to a human concentration of 2.7 mg/mL. Anxiolytic and antidepressant properties were evaluated using behavioural tests, while electroencephalography (EEG) analysis probed the neural mechanisms underlying sleep promotion post-consumption. The aqueous extract of M. hortensis dried flower administered to mice showed a decrease in immobility in the forced swimming test, demonstrating antidepressant-like effects. Moreover, mice treated with M. hortensis aqueous exhibited increased non-rapid eye movement (NREM) sleep duration, corroborating sleep-promoting potential. EEG analysis of mice treated with M. hortensis aqueous revealed heightened beta oscillations in the frontal and parietal cortex, while pre-treatment with M. hortensis aqueous or diazepam enhanced rapid eye movement (REM) sleep after thiopental administration. Interestingly, M. hortensis aqueous pre-treatment augmented delta frequency ranges in the frontal cortex. Overall, these findings indicate that M. hortensis dried flower's aqueous extract, at a human-equivalent dosage, exerts significant behavioural and neural effects specifically, sedative and hypnotic aspects in mice, corroborating its potential as a natural remedy to promote sleep and regulate mood.
Collapse
Affiliation(s)
- Dania Cheaha
- Division of Biological Science, Faculty of Science, Prince of Songkla University, Thailand
| | - Nurulhuda Basor
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Thailand
| | - Rodiya Manor
- Faculty of Science and Technology, Prince of Songkla University, Thailand
| | - Nabil Hayeemasae
- Faculty of Science and Technology, Prince of Songkla University, Thailand
| | - Nifareeda Samerphob
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Thailand
| |
Collapse
|
40
|
Wang D, Bao C, Wu H, Li J, Zhang X, Wang S, Zhou F, Li H, Dong H. A hypothalamus-lateral periaqueductal gray GABAergic neural projection facilitates arousal following sevoflurane anesthesia in mice. CNS Neurosci Ther 2024; 30:e70047. [PMID: 39317457 PMCID: PMC11421888 DOI: 10.1111/cns.70047] [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: 04/29/2024] [Revised: 08/01/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND The lateral hypothalamus (LHA) is an evolutionarily conserved structure that regulates basic functions of an organism, particularly wakefulness. To clarify the function of LHAGABA neurons and their projections on regulating general anesthesia is crucial for understanding the excitatory and inhibitory effects of anesthetics on the brain. The aim of the present study is to investigate whether LHAGABA neurons play either an inhibitory or a facilitatory role in sevoflurane-induced anesthetic arousal regulation. METHODS We used fiber photometry and immunofluorescence staining to monitor changes in neuronal activity during sevoflurane anesthesia. Opto-/chemogenetic modulations were employed to study the effect of neurocircuit modulations during the anesthesia. Anterograde tracing was used to identify a GABAergic projection from the LHA to a periaqueductal gray (PAG) subregion. RESULTS c-Fos staining showed that LHAGABA activity was inhibited by induction of sevoflurane anesthesia. Anterograde tracing revealed that LHAGABA neurons project to multiple arousal-associated brain areas, with the lateral periaqueductal gray (LPAG) being one of the dense projection areas. Optogenetic experiments showed that activation of LHAGABA neurons and their downstream target LPAG reduced the burst suppression ratio (BSR) during continuous sevoflurane anesthesia. Chemogenetic experiments showed that activation of LHAGABA and its projection to LPAG neurons prolonged the anesthetic induction time and promoted wakefulness. CONCLUSIONS In summary, we show that an inhibitory projection from LHAGABA to LPAGGABA neurons promotes arousal from sevoflurane-induced loss of consciousness, suggesting a complex control of wakefulness through intimate interactions between long-range connections.
Collapse
Affiliation(s)
- Dan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Chang Bao
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Huimin Wu
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Jiannan Li
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Xinxin Zhang
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Sa Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Fang Zhou
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Huiming Li
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| |
Collapse
|
41
|
Liu P, Li Q, Tang YF, Cui CY, Liu Q, Zhang Y, Tang B, Lai QC. Multiple algorithms highlight key brain genes driven by multiple anesthetics. Comput Biol Med 2024; 179:108805. [PMID: 38991319 DOI: 10.1016/j.compbiomed.2024.108805] [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: 04/07/2024] [Revised: 05/05/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
Anesthesia serves as a pivotal tool in modern medicine, creating a transient state of sensory deprivation to ensure a pain-free surgical or medical intervention. While proficient in alleviating pain, anesthesia significantly modulates brain dynamics, metabolic processes, and neural signaling, thereby impairing typical cognitive functions. Furthermore, anesthesia can induce notable impacts such as memory impairment, decreased cognitive function, and diminished intelligence, emphasizing the imperative need to explore the concealed repercussions of anesthesia on individuals. In this investigation, we aggregated gene expression profiles (GSE64617, GSE141242, GSE161322, GSE175894, and GSE178995) from public repositories following second-generation sequencing analysis of various anesthetics. Through scrutinizing post-anesthesia brain tissue gene expression utilizing Gene Set Enrichment Analysis (GSEA), Robust Rank Aggregation (RRA), and Weighted Gene Co-expression Network Analysis (WGCNA), this research aims to pinpoint pivotal genes, pathways, and regulatory networks linked to anesthesia. This undertaking not only enhances comprehension of the physiological changes brought about by anesthesia but also lays the groundwork for future investigations, cultivating new insights and innovative perspectives in medical practice.
Collapse
Affiliation(s)
- Ping Liu
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qun Li
- Department of Pain, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yi-Fan Tang
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chun-Yan Cui
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China; Department of Pain, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qing Liu
- Department of Pain, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China; Department of Anesthesiology, Hejiang Hospital of Traditional Chinese Medicine, Southwest Medical University, China
| | - Ying Zhang
- Department of Anesthesiology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China; Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, 646000, Sichuan, China; Department of Anesthesiology, Hejiang Hospital of Traditional Chinese Medicine, Southwest Medical University, China.
| | - Bo Tang
- Department of Pathology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qian-Cheng Lai
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
42
|
Massimini M, Corbetta M, Sanchez-Vives MV, Andrillon T, Deco G, Rosanova M, Sarasso S. Sleep-like cortical dynamics during wakefulness and their network effects following brain injury. Nat Commun 2024; 15:7207. [PMID: 39174560 PMCID: PMC11341729 DOI: 10.1038/s41467-024-51586-1] [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: 12/29/2023] [Accepted: 08/07/2024] [Indexed: 08/24/2024] Open
Abstract
By connecting old and recent notions, different spatial scales, and research domains, we introduce a novel framework on the consequences of brain injury focusing on a key role of slow waves. We argue that the long-standing finding of EEG slow waves after brain injury reflects the intrusion of sleep-like cortical dynamics during wakefulness; we illustrate how these dynamics are generated and how they can lead to functional network disruption and behavioral impairment. Finally, we outline a scenario whereby post-injury slow waves can be modulated to reawaken parts of the brain that have fallen asleep to optimize rehabilitation strategies and promote recovery.
Collapse
Grants
- The authors thank Dr Ezequiel Mikulan, Dr Silvia Casarotto, Dr Andrea Pigorini, Dr Simone Russo, and Dr Pilleriin Sikka for their help and comments on the manuscript draft and illustrations. This work was financially supported by the following entities: ERC-2022-SYG Grant number 101071900 Neurological Mechanisms of Injury and Sleep-like Cellular Dynamics (NEMESIS); Italian National Recovery and Resilience Plan (NRRP), M4C2, funded by the European Union - NextGenerationEU (Project IR0000011, CUP B51E22000150006, “EBRAINS-Italy”); European Union’s Horizon 2020 Framework Program for Research and Innovation under the Specific Grant Agreement No.945539 (Human Brain Project SGA3); Tiny Blue Dot Foundation; Canadian Institute for Advanced Research (CIFAR), Canada; Italian Ministry for Universities and Research (PRIN 2022); Fondazione Regionale per la Ricerca Biomedica (Regione Lombardia), Project ERAPERMED2019–101, GA 779282; CORTICOMOD PID2020-112947RB-I00 financed by MCIN/ AEI /10.13039/501100011033; Fondazione Cassa di Risparmio di Padova e Rovigo (CARIPARO) Grant Agreement number 55403; Ministry of Health, Italy (RF-2008 -12366899) Brain connectivity measured with high-density electroencephalography: a novel neurodiagnostic tool for stroke- NEUROCONN; BIAL foundation grant (Grant Agreement number 361/18); H2020 European School of Network Neuroscience (euSNN); H2020 Visionary Nature Based Actions For Heath, Wellbeing & Resilience in Cities (VARCITIES); Ministry of Health Italy (RF-2019-12369300): Eye-movement dynamics during free viewing as biomarker for assessment of visuospatial functions and for closed-loop rehabilitation in stroke (EYEMOVINSTROKE).
Collapse
Affiliation(s)
- Marcello Massimini
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy.
| | - Maurizio Corbetta
- Department of Neuroscience and Padova Neuroscience Center (PNC), University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Maria V Sanchez-Vives
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Institució Catalana de la Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Thomas Andrillon
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Mov'it team, Inserm, CNRS, Paris, France
- Monash Centre for Consciousness and Contemplative Studies, Faculty of Arts, Monash University, Melbourne, VIC, Australia
| | - Gustavo Deco
- Institució Catalana de la Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Information and Communication Technologies, Universitat Pompeu Fabra, Center for Brain and Cognition, Computational Neuroscience Group, Barcelona, Spain
| | - Mario Rosanova
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Simone Sarasso
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
43
|
Dong K, Zhang L, Zhong Y, Xu T, Zhao Y, Chen S, Mahmoud SS, Fang Q. Meso-scale reorganization of local-global brain networks under mild sedation of propofol anesthesia. Neuroimage 2024; 297:120744. [PMID: 39033791 DOI: 10.1016/j.neuroimage.2024.120744] [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: 05/16/2024] [Revised: 06/30/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024] Open
Abstract
The fragmentation of the functional brain network has been identified through the functional connectivity (FC) analysis in studies investigating anesthesia-induced loss of consciousness (LOC). However, it remains unclear whether mild sedation of anesthesia can cause similar effects. This paper aims to explore the changes in local-global brain network topology during mild anesthesia, to better understand the macroscopic neural mechanism underlying anesthesia sedation. We analyzed high-density EEG from 20 participants undergoing mild and moderate sedation of propofol anesthesia. By employing a local-global brain parcellation in EEG source analysis, we established binary functional brain networks for each participant. Furthermore, we investigated the global-scale properties of brain networks by estimating global efficiency and modularity, and examined the changes in meso-scale properties of brain networks by quantifying the distribution of high-degree and high-betweenness hubs and their corresponding rich-club coefficients. It is evident from the results that the mild sedation of anesthesia does not cause a significant change in the global-scale properties of brain networks. However, network components centered on SomMot L show a significant decrease, while those centered on Default L, Vis L and Limbic L exhibit a significant increase during the transition from wakefulness to mild sedation (p<0.05). Compared to the baseline state, mild sedation almost doubled the number of high-degree hubs in Vis L, DorsAttn L, Limbic L, Cont L, and reduced by half the number of high-degree hubs in SomMot R, DorsAttn R, SalVentAttn R. Further, mild sedation almost doubled the number of high-betweenness hubs in Vis L, Vis R, Limbic R, Cont R, and reduced by half the number of high-betweenness hubs in SomMot L, SalVentAttn L, Default L, and SomMot R. Our results indicate that mild anesthesia cannot affect the global integration and segregation of brain networks, but influence meso-scale function for integrating different resting-state systems involved in various segregation processes. Our findings suggest that the meso-scale brain network reorganization, situated between global integration and local segregation, could reflect the autonomic compensation of the brain for drug effects. As a direct response and adjustment of the brain network system to drug administration, this spontaneous reorganization of the brain network aims at maintaining consciousness in the case of sedation.
Collapse
Affiliation(s)
- Kangli Dong
- Department of Biomedical Engineering, College of Engineering, Shantou University, Shantou 515063, Guangdong, China.
| | - Lu Zhang
- Department of Rehabilitation, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310027, Zhejiang, China.
| | - Yuming Zhong
- Department of Biomedical Engineering, College of Engineering, Shantou University, Shantou 515063, Guangdong, China.
| | - Tao Xu
- Department of Biomedical Engineering, College of Engineering, Shantou University, Shantou 515063, Guangdong, China.
| | - Yue Zhao
- Department of Urology, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen 361102, Fujian, China.
| | - Siya Chen
- Department of Computer Science, City University of Hong Kong, Hong Kong 999077, Hong Kong, China.
| | - Seedahmed S Mahmoud
- Department of Biomedical Engineering, College of Engineering, Shantou University, Shantou 515063, Guangdong, China.
| | - Qiang Fang
- Department of Biomedical Engineering, College of Engineering, Shantou University, Shantou 515063, Guangdong, China.
| |
Collapse
|
44
|
Kinateder T, Kratzer S, Husemann C, Hautmann H, García PS, Schneider G, Kreuzer M. Electroencephalogram-Based Anesthesia Indices Differently React to Modulations of Alpha-Oscillatory Activity. Anesth Analg 2024:00000539-990000000-00892. [PMID: 39093724 DOI: 10.1213/ane.0000000000007042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
BACKGROUND The electroencephalographic (EEG) provides the anesthesiologist with information regarding the level of anesthesia. Processed EEG indices are available that reflect the level of anesthesia as a single number. Strong oscillatory EEG activity in the alpha-band may be associated with an adequate level of anesthesia and a lower incidence of cognitive sequelae. So far, we do not know how the processed indices would react to changes in the alpha-band activity. Hence, we modulated the alpha-oscillatory activity of intraoperative EEG to assess possible index changes. METHODS We performed our analyses based on data from 2 studies. Intraoperative EEG was extracted, and we isolated the alpha-band activity by band-pass filtering (8-12 Hz). We added or subtracted this activity to the original EEG in different steps with different amplifications of the alpha signal. We then replayed these signals to the bispectral index (BIS), the Entropy Module (state entropy [SE]), the CONOX (qCON), and the SEDLine (patient state index [PSI]); and evaluated the alpha-band modulation's impact on the respective index. RESULTS The indices behaved differently to the modulation. In general, indices decreased with stronger alpha-band activity, but the rate of change was different with SE showing the strongest change (9% per step) and PSI and BIS (<5% per step) showing the weakest change. A simple regression analysis revealed a decrease of 0.02 to 0.09 index points with increasing alpha amplification. CONCLUSIONS While the alpha-band in the intraoperative EEG seems to carry information regarding the quality of anesthesia, changes in the alpha-band activity do neither strongly nor uniformly influence processed EEG indices. Hence, to assess alpha-oscillatory activity's strength, the user needs to focus on the raw EEG or its spectral representation also displayed on the monitoring systems.
Collapse
Affiliation(s)
- Thomas Kinateder
- From the Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Stephan Kratzer
- Department of Anesthesia, Critical Care and Pain Medicine, Hessing Foundation, Augsburg, Germany
| | - Cornelius Husemann
- From the Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Hubert Hautmann
- Department of Internal Medicine and Pneumology, Klinik Ottobeuren, Ottobeuren, Germany
| | - Paul S García
- Department of Anesthesiology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Gerhard Schneider
- From the Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Matthias Kreuzer
- From the Department of Anesthesiology and Intensive Care, School of Medicine and Health, Technical University of Munich, Munich, Germany
| |
Collapse
|
45
|
Zhang S, Zhang X, Li H, Wang D, Wang S, Wang Y, Zhao G, Dong H, Li J. Ventral Tegmental Area Glutamatergic Neurons Facilitated Emergence From Isoflurane Anesthesia Involves Excitation of Lateral Septum GABA-ergic Neurons in Mice. Anesth Analg 2024; 139:397-410. [PMID: 38048607 DOI: 10.1213/ane.0000000000006739] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
BACKGROUND Ventral tegmental area (VTA) glutamatergic neurons promote wakefulness in the sleep-wake cycle; however, their roles and neural circuit mechanisms during isoflurane (ISO) anesthesia remain unclear. METHODS Fiber photometry and in vivo electrophysiology were used to observe the changes in neuronal or terminal activity during ISO anesthesia and arousal processes. Optogenetic and anesthesia behaviors were used to investigate the effects of VTA glutamatergic neurons and their projections to the lateral septum (LS) during ISO anesthesia and arousal. Anterograde and retrograde tracings were performed to identify the connections between VTA glutamatergic neurons and the LS. RESULTS Population activity and firing rates of VTA glutamatergic neurons decreased during ISO anesthesia (ISO: 95% confidence interval [CI], 0.83-2.06 Spikes.s -1 vs wake: 95% CI, 3.53-7.83 Spikes.s -1 ; P =.0001; n = 34 from 4 mice). Optogenetic activation of VTA glutamatergic neurons reduced the burst-suppression ratio in electroencephalography (laser: 95% CI, 13.09%-28.76% vs pre: 95% CI, 52.85%-71.59%; P =.0009; n = 6) and facilitated emergence (ChR2: 95% CI, 343.3-388.0 seconds vs mCherry: 95% CI, 447.6-509.8 seconds; P < .0001; n = 11/12) from ISO anesthesia. VTA glutamatergic neurons monosynaptically innervated LS γ-aminobutyric acid (GABA)-ergic neurons. The activity of VTA glutamatergic terminals in the LS decreased during ISO anesthesia, and optogenetic activation of the VTA glutamatergic terminals in the LS facilitated emergence from ISO anesthesia. Furthermore, optogenetic activation of VTA glutamatergic terminals increased the firing rates of LS γ-aminobutyric acid-ergic (GABAergic) neurons (laser: 95% CI, 0.85-4.03 Spikes.s -1 vs pre: 95% CI, 0.24-0.78 Spikes.s -1 ; P =.008; n = 23 from 4 mice) during ISO anesthesia. CONCLUSIONS VTA glutamatergic neurons facilitated emergence from ISO anesthesia involving excitation of LS GABAergic neurons.
Collapse
Affiliation(s)
- Simin Zhang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Xinxin Zhang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huiming Li
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dan Wang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Sa Wang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuhao Wang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Guangchao Zhao
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hailong Dong
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiannan Li
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
46
|
Wang X, Yi R, Liang X, Zhang N, Zhong F, Lu Y, Chen W, Yu T, Zhang L, Wang H, Zhou L. Myelin modulates the process of isoflurane anesthesia through the regulation of neural activity. CNS Neurosci Ther 2024; 30:e14922. [PMID: 39138640 PMCID: PMC11322027 DOI: 10.1111/cns.14922] [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: 03/14/2024] [Revised: 07/01/2024] [Accepted: 08/04/2024] [Indexed: 08/15/2024] Open
Abstract
AIMS The mechanism underlying the reversible unconsciousness induced by general anesthetics (GA) remains unclear. Recent studies revealed the critical roles of myelin and oligodendrocytes (OLs) in higher functions of the brain. However, it is unknown whether myelin actively participates in the regulation of GA. The aim of this study is to investigate the roles and possible mechanisms of myelin in the regulation of consciousness alterations induced by isoflurane anesthesia. METHODS First, demyelination models for the entire brain and specific neural nuclei were established to investigate the potential role of myelination in the regulation of GA, as well as its possible regional specificity. c-Fos staining was then performed on the demyelinated nuclei to verify the impact of myelin loss on neuronal activity. Finally, the activity of neurons during isoflurane anesthesia in demyelinated mice was recorded by optical fiber photometric calcium signal. The related behavioral indicators and EEG were recorded and analyzed. RESULTS A prolonged emergence time was observed from isoflurane anesthesia in demyelinated mice, which suggested the involvement of myelin in regulating GA. The demyelination in distinct nuclei by LPC further clarified the region-specific roles of isoflurane anesthesia regulation by myelin. The effect of demyelination on isoflurane anesthesia in the certain nucleus was consistent with that in neurons towards isoflurane anesthesia. Finally, we found that the mechanism of myelin in the modulation of isoflurane anesthesia is possibly through the regulation of neuronal activity. CONCLUSIONS In brief, myelin in the distinct neural nucleus plays an essential role in regulating the process of isoflurane anesthesia. The possible mechanism of myelin in the regulation of isoflurane anesthesia is neuronal activity modification by myelin integrity during GA. Our findings enhanced the comprehension of myelin function, and offered a fresh perspective for investigating the neural mechanisms of GA.
Collapse
Affiliation(s)
- Xu Wang
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
| | - Rulan Yi
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Xiaoling Liang
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
| | - Ning Zhang
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
| | - Fuwang Zhong
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
| | - Yali Lu
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
| | - Wenjia Chen
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
| | - Tian Yu
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Linyong Zhang
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Haiying Wang
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Liang Zhou
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| |
Collapse
|
47
|
Wang R, Zhang L, Wang X, Li W, Jian T, Yin P, Wang X, Chen Q, Chen X, Qin H. Electrophysiological activity pattern of mouse hippocampal CA1 and dentate gyrus under isoflurane anesthesia. Front Cell Neurosci 2024; 18:1392498. [PMID: 39104439 PMCID: PMC11299216 DOI: 10.3389/fncel.2024.1392498] [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: 02/27/2024] [Accepted: 07/03/2024] [Indexed: 08/07/2024] Open
Abstract
General anesthesia can impact a patient's memory and cognition by influencing hippocampal function. The CA1 and dentate gyrus (DG), serving as the primary efferent and gateway of the hippocampal trisynaptic circuit facilitating cognitive learning and memory functions, exhibit significant differences in cellular composition, molecular makeup, and responses to various stimuli. However, the effects of isoflurane-induced general anesthesia on CA1 and DG neuronal activity in mice are not well understood. In this study, utilizing electrophysiological recordings, we examined neuronal population dynamics and single-unit activity (SUA) of CA1 and DG in freely behaving mice during natural sleep and general anesthesia. Our findings reveal that isoflurane anesthesia shifts local field potential (LFP) to delta frequency and reduces the firing rate of SUA in both CA1 and DG, compared to wakefulness. Additionally, the firing rates of DG neurons are significantly lower than CA1 neurons during isoflurane anesthesia, and the recovery of theta power is slower in DG than in CA1 during the transition from anesthesia to wakefulness, indicating a stronger and more prolonged impact of isoflurane anesthesia on DG. This work presents a suitable approach for studying brain activities during general anesthesia and provides evidence for distinct effects of isoflurane anesthesia on hippocampal subregions.
Collapse
Affiliation(s)
- Rui Wang
- Department of Anesthesiology, Shanxi Medical University and Second Hospital of Shanxi Medical University, Taiyuan, China
- Guangyang Bay Laboratory, Chongqing Institute for Brain and Intelligence, Chongqing, China
| | - Linzhong Zhang
- Department of Anesthesiology, Shanxi Medical University and Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xia Wang
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, China
| | - Wen Li
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Tingliang Jian
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Pengcheng Yin
- Department of Anesthesiology, Shanxi Medical University and Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xinzhi Wang
- Department of Anesthesiology, Shanxi Medical University and Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Qianwei Chen
- Department of Rehabilitation Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaowei Chen
- Guangyang Bay Laboratory, Chongqing Institute for Brain and Intelligence, Chongqing, China
- Brain Research Center and State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Han Qin
- Guangyang Bay Laboratory, Chongqing Institute for Brain and Intelligence, Chongqing, China
| |
Collapse
|
48
|
Hudetz AG. Microstimulation reveals anesthetic state-dependent effective connectivity of neurons in cerebral cortex. Front Neurosci 2024; 18:1387098. [PMID: 39035779 PMCID: PMC11258030 DOI: 10.3389/fnins.2024.1387098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/07/2024] [Indexed: 07/23/2024] Open
Abstract
Introduction Complex neuronal interactions underlie cortical information processing that can be compromised in altered states of consciousness. Here intracortical microstimulation was applied to investigate anesthetic state-dependent effective connectivity of neurons in rat visual cortex in vivo. Methods Extracellular activity was recorded at 32 sites in layers 5/6 while stimulating with charge-balanced discrete pulses at each electrode in random order. The same stimulation pattern was applied at three levels of anesthesia with desflurane and in wakefulness. Spikes were sorted and classified by their waveform features as putative excitatory and inhibitory neurons. Network motifs were identified in graphs of effective connectivity constructed from monosynaptic cross-correlograms. Results Microstimulation caused early (<10 ms) increase followed by prolonged (11-100 ms) decrease in spiking of all neurons throughout the electrode array. The early response of excitatory but not inhibitory neurons decayed rapidly with distance from the stimulation site over 1 mm. Effective connectivity of neurons with significant stimulus response was dense in wakefulness and sparse under anesthesia. The number of network motifs, especially those of higher order, increased rapidly as the anesthesia was withdrawn indicating a substantial increase in network connectivity as the animals woke up. Conclusion The results illuminate the impact of anesthesia on functional integrity of local cortical circuits affecting the state of consciousness.
Collapse
Affiliation(s)
- Anthony G Hudetz
- Department of Anesthesiology, Center for Consciousness Science, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
49
|
Zhou L, Ran Q, Yi R, Tang H, Zhang Y, Yu T. Glutamatergic neurons of piriform cortex delay induction of inhalational general anesthesia. FUNDAMENTAL RESEARCH 2024; 4:829-840. [PMID: 39156577 PMCID: PMC11330113 DOI: 10.1016/j.fmre.2022.12.014] [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: 04/30/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 01/04/2023] Open
Abstract
Since their clinical application in the 1840s, the greatest mystery surrounding general anesthesia (GA) is how different kinds of general anesthetics cause reversible unconsciousness, and the precise neural mechanisms underlying the processes. Over past years, although many studies revealed the roles of cortex, thalamus, brainstem, especially the sleep-wake circuits in GA-induced loss of consciousness (LOC),the full picture of the neural circuit mechanism of GA is still largely unknown. Recent studies have focused on the importance of other brain regions. Here, we report that the activity of glutamatergic (Glu) neurons in the piriform cortex (PC), a critical brain region for odor encoding, began to increase during the LOC of GA and gradually recovered after recovery of consciousness. Chemical lesions of the anterior PC (APC) neurons accelerated the induction time of isoflurane anesthesia. Chemogenetic and optogenetic activation of APCGlu neurons prolonged isoflurane and sevoflurane anesthesia induction, whereas APCGlu neuron inhibition displayed the opposite effects. Moreover, the modification of APCGlu neurons did not affect the induction or emergence time of propofol GA. In addition, odor processing may be partially involved in the induction of isoflurane and sevoflurane GA regulated by APCGlu neurons. In conclusion, our findings reveal a critical role of APCGlu neurons in inhalational GA induction.
Collapse
Affiliation(s)
- Liang Zhou
- Key Laboratory of Brain Science, Zunyi Medical University, Zunyi 563003, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi 563003, China
| | - Qipeng Ran
- Key Laboratory of Brain Science, Zunyi Medical University, Zunyi 563003, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi 563003, China
| | - Rulan Yi
- Key Laboratory of Brain Science, Zunyi Medical University, Zunyi 563003, China
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Huanyao Tang
- Key Laboratory of Brain Science, Zunyi Medical University, Zunyi 563003, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi 563003, China
| | - Yu Zhang
- Key Laboratory of Brain Science, Zunyi Medical University, Zunyi 563003, China
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi 563003, China
| | - Tian Yu
- Key Laboratory of Brain Science, Zunyi Medical University, Zunyi 563003, China
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi 563003, China
| |
Collapse
|
50
|
Wu Y, Zhang D, Liu J, Jiang J, Xie K, Wu L, Leng Y, Liang P, Zhu T, Zhou C. Activity of the Sodium Leak Channel Maintains the Excitability of Paraventricular Thalamus Glutamatergic Neurons to Resist Anesthetic Effects of Sevoflurane in Mice. Anesthesiology 2024; 141:56-74. [PMID: 38625708 DOI: 10.1097/aln.0000000000005015] [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
BACKGROUND Stimulation of the paraventricular thalamus has been found to enhance anesthesia recovery; however, the underlying molecular mechanism by which general anesthetics modulate paraventricular thalamus is unclear. This study aimed to test the hypothesis that the sodium leak channel (NALCN) maintains neuronal activity in the paraventricular thalamus to resist anesthetic effects of sevoflurane in mice. METHODS Chemogenetic and optogenetic manipulations, in vivo multiple-channel recordings, and electroencephalogram recordings were used to investigate the role of paraventricular thalamus neuronal activity in sevoflurane anesthesia. Virus-mediated knockdown and/or overexpression was applied to determine how NALCN influenced excitability of paraventricular thalamus glutamatergic neurons under sevoflurane. Viral tracers and local field potentials were used to explore the downstream pathway. RESULTS Single neuronal spikes in the paraventricular thalamus were suppressed by sevoflurane anesthesia and recovered during emergence. Optogenetic activation of paraventricular thalamus glutamatergic neurons shortened the emergence period from sevoflurane anesthesia, while chemogenetic inhibition had the opposite effect. Knockdown of the NALCN in the paraventricular thalamus delayed the emergence from sevoflurane anesthesia (recovery time: from 24 ± 14 to 64 ± 19 s, P < 0.001; concentration for recovery of the righting reflex: from 1.13% ± 0.10% to 0.97% ± 0.13%, P < 0.01). As expected, the overexpression of the NALCN in the paraventricular thalamus produced the opposite effects. At the circuit level, knockdown of the NALCN in the paraventricular thalamus decreased the neuronal activity of the nucleus accumbens, as indicated by the local field potential and decreased single neuronal spikes in the nucleus accumbens. Additionally, the effects of NALCN knockdown in the paraventricular thalamus on sevoflurane actions were reversed by optical stimulation of the nucleus accumbens. CONCLUSIONS Activity of the NALCN maintains the excitability of paraventricular thalamus glutamatergic neurons to resist the anesthetic effects of sevoflurane in mice. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Yujie Wu
- Department of Anesthesiology, West China Hospital, 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, Sichuan University, Chengdu, China
| | - Donghang Zhang
- Department of Anesthesiology, West China Hospital, 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, Sichuan University, Chengdu, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, 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, Sichuan University, Chengdu, China
| | - Jingyao Jiang
- Department of Anesthesiology, West China Hospital, 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, Sichuan University, Chengdu, China
| | - Keyu Xie
- Department of Anesthesiology, West China Hospital, 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, Sichuan University, Chengdu, China
| | - Lin Wu
- Department of Anesthesiology, West China Hospital, 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, Sichuan University, Chengdu, China
| | - Yu Leng
- Department of Anesthesiology, West China Hospital, 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, Sichuan University, Chengdu, China
| | - Peng Liang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, 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, Sichuan University, Chengdu, China
| |
Collapse
|