1
|
Ding X, Hwang FJ, Silverman D, Zhong P, Li B, Ma C, Lu L, Jiang G, Zhang Z, Huang X, Tu X, Tian ZM, Ding J, Dan Y. Neuroendocrine circuit for sleep-dependent growth hormone release. Cell 2025:S0092-8674(25)00626-9. [PMID: 40562026 DOI: 10.1016/j.cell.2025.05.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 04/30/2025] [Accepted: 05/28/2025] [Indexed: 06/28/2025]
Abstract
Sleep is known to promote tissue growth and regulate metabolism, partly by enhancing growth hormone (GH) release, but the underlying circuit mechanism is unknown. We demonstrate how GH release, which is enhanced during both rapid eye movement (REM) and non-REM (NREM) sleep, is regulated by sleep-wake-dependent activity of distinct hypothalamic neurons expressing GH-releasing hormone (GHRH) and somatostatin (SST). SST neurons in the arcuate nucleus suppress GH release by inhibiting nearby GHRH neurons that stimulate GH release, whereas periventricular SST neurons inhibit GH release by projecting to the median eminence. GH release is associated with strong surges of both GHRH and SST activity during REM sleep but moderately increased GHRH and decreased SST activity during NREM sleep. Furthermore, we identified a negative feedback pathway in which GH enhances the excitability of locus coeruleus neurons and increases wakefulness. These results elucidate a circuit mechanism underlying bidirectional interactions between sleep and hormone regulation.
Collapse
Affiliation(s)
- Xinlu Ding
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Fuu-Jiun Hwang
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Daniel Silverman
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Peng Zhong
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Bing Li
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Chenyan Ma
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Lihui Lu
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Grace Jiang
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Zhe Zhang
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Xiaolin Huang
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Xun Tu
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Zhiyu Melissa Tian
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA
| | - Jun Ding
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
| | - Yang Dan
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
2
|
Imamura T, Wasilczuk AZ, Reitz SL, Lian J, Imamura M, Keenan BT, Shimizu N, Pack AI, Kelz MB. Parafacial GABAergic neurone ablation induces behavioural resistance to volatile anaesthetic-induced hypnosis without reducing sleep. Br J Anaesth 2025; 134:1696-1708. [PMID: 40240218 PMCID: PMC12106870 DOI: 10.1016/j.bja.2025.02.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND It is hypothesised that general anaesthetics co-opt the neural circuits regulating endogenous sleep and wakefulness to produce hypnosis. To further probe this association, we focused on the GABAergic neurones of the parafacial zone (PZGABA), a brainstem site capable of promoting non-rapid eye movement sleep. METHODS To determine whether PZ neurones are activated by a hypnotic dose of anaesthetics, c-Fos immunohistochemistry was performed. The behavioural and physiological contributions of PZGABA neurones to anaesthetic sensitivity were assessed in mice transfected with an adeno-associated virus (AAV)-driving expression of an mCherry fluorescent control or a caspase that irreversibly eliminates PZGABA neurones. EEG-defined sleep was measured in PZGABA-ablated and mCherry control mice, as was the homeostatic drive to sleep after sleep deprivation. RESULTS Consistent with anaesthetic-induced depolarisation, hypnotic doses of isoflurane significantly increased c-Fos expression three-fold in PZGABA neurones compared with oxygen-exposed mice. PZGABA-ablated mice developed significant and durable behavioural resistance to both isoflurane- and sevoflurane-induced hypnosis, with roughly 50% higher likelihood of intact righting than controls. PZGABA-ablated mice emerged from isoflurane significantly faster than mCherry controls with purposeful movements. The degree of anaesthetic resistance was inversely correlated with the number of surviving PZGABA neurones. Despite confirming that PZGABA ablation reduced the potency of two distinct volatile anaesthetics behaviourally, ablation did not alter the amount of endogenous sleep or wakefulness, nor did it affect the homeostatic sleep drive after sleep deprivation, and it did not produce EEG signatures of anaesthetic resistance during isoflurane exposure. CONCLUSIONS There was an unexpected dissociation in which destruction of up to 70-80% of PZGABA neurones was sufficient to alter anaesthetic susceptibility behaviourally without causing insomnia or altering sleep pressure. These findings suggest that PZGABA neurones are more critical to drug-induced hypnosis than to the regulation of natural sleep and arousal.
Collapse
Affiliation(s)
- Toshihiro Imamura
- Chronobiology and Sleep Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Division of Pulmonary and Sleep Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Sleep Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Andrzej Z Wasilczuk
- Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah L Reitz
- Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jie Lian
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Miyoko Imamura
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Brendan T Keenan
- Chronobiology and Sleep Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Division of Sleep Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Naoki Shimizu
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Allan I Pack
- Chronobiology and Sleep Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Division of Sleep Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Max B Kelz
- Chronobiology and Sleep Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Anesthesiology and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Li X, Ma Z, Liu X, Chen C, Yu Z, Sang D, Wang T, Zhang EE, Duan G, Ju D, Huang H. Activation of CaMKII + neurons in the paramedian raphe nucleus promotes general anesthesia in male mice. Cell Biol Toxicol 2025; 41:83. [PMID: 40360778 PMCID: PMC12075403 DOI: 10.1007/s10565-025-10037-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
General anesthesia (GA) is an essential clinical and surgical adjunct, widely recognized as the result of coordinated networks among numerous brain regions. Anesthetic drugs with different characteristics are associated with distinct networks of brain regions involved in anesthesia. Ciprofol, a novel intravenous anesthetic derived from structural modifications of propofol, has shown promise in clinical applications. However, the specific neuronal circuits and brain regions mediating their actions may differ. Moreover, the core brain regions that mediate the common anesthetic effects of these drugs remain unclear. In this research, we identified a central ensemble of brainstem neurons within the paramedian raphe nucleus (PMnR) using c-Fos staining in mice subjected to GA induced by continuous intravenous infusion of ciprofol and propofol. This neuronal population, primarily composed of CaMKIIa and Gad1-expressing cells, demonstrated consistent activation in reaction to ciprofol. Optogenetic activation of PMnRCaMKIIa neurons induced a GA state under ciprofol pre-administration, while sole activation of PMnRCaMKIIa neurons induced a motionless state in mice. In addition, conditional inhibition of these neurons resulted in resistance to GA. In summary, we highlight the PMnR as a brain target for ciprofol and propofol. Furthermore, CaMKIIa+ neurons in the PMnR emerge as active promoters of the anesthesia process, shedding light on a previously unrecognized key player in the intricate neural network orchestrating GA.
Collapse
Affiliation(s)
- Xuehan Li
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhixiong Ma
- Chinese Institute for Brain Research, Beijing, China.
| | - Xueliang Liu
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Chen Chen
- National Institute of Biological Sciences, Beijing, China
| | - Ziqing Yu
- National Institute of Biological Sciences, Beijing, China
| | - Di Sang
- National Institute of Biological Sciences, Beijing, China
| | - Tongfei Wang
- Chinese Institute for Brain Research, Beijing, China
| | - Eric Erquan Zhang
- National Institute of Biological Sciences, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Guangyou Duan
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Dapeng Ju
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - He Huang
- Department of Anesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
4
|
Stone ME, Kelz MB, Proekt A, Wasilczuk AZ. A probabilistic model of behavioural emergence from general anaesthesia in mice. Br J Anaesth 2025:S0007-0912(25)00166-7. [PMID: 40287361 DOI: 10.1016/j.bja.2025.02.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/16/2025] [Accepted: 02/17/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Time to emergence from general anaesthesia is highly variable between individuals. This variability has been attributed to individual differences in anaesthetic sensitivity. However, this hypothesis has not been verified experimentally. We explicitly test this hypothesis by quantifying emergence from anaesthesia repeatedly in the same individuals over time. METHODS Genetically identical adult (12-24 weeks old) male (n=40) and female (n=20) C57BL/6J mice were exposed to 2 h of isoflurane (0.90 vol%) on 10 separate occasions. Time to emergence was measured using the return of the righting reflex. Predictions of the standard effect-site pharmacokinetic-pharmacodynamic (PK-PD) model and neuronal dynamics model of stochastic fluctuations between the awake and anaesthetised states were fit to observed emergence times. Repeated steady-state assessments of the righting reflex obtained during the last 2 h of a 4-h exposure to 0.3, 0.4, 0.6, or 0.7 vol% isoflurane (n=20 per concentration) were used to determine individual probabilities of losing the righting reflex, which was defined as an individual's anaesthetic sensitivity. RESULTS Emergence times varied by at least two orders of magnitude after identical anaesthetic exposure. We did not find consistent inter-individual differences in emergence times. Instead, we found that variability in emergence times across trials in each individual was as large as that between two different individuals. Emergence times were not correlated across time. Consistent with previous work, we identified large individual differences in anaesthetic sensitivity which persisted on a time scale of at least 1 week. A standard PK-PD model failed to reproduce inter-trial variability. In contrast, the neuronal dynamics model reproduced both population- and individual-level variability in emergence times. CONCLUSIONS Stochastic state switching contributes to inherent variability in emergence from general anaesthesia. Delayed emergence occurred in a small proportion of anaesthetic exposures in a genetically homogeneous population. The neuronal dynamics model predicts that anaesthetic emergence times will be probabilistically long, which might explain delayed emergence observed in clinical settings.
Collapse
Affiliation(s)
- Martha E Stone
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA; Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Max B Kelz
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA; Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Alex Proekt
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrzej Z Wasilczuk
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Manohara N, Ferrari A, Greenblatt A, Berardino A, Peixoto C, Duarte F, Moyiaeri Z, Robba C, Nascimento FA, Kreuzer M, Vacas S, Lobo FA. Electroencephalogram monitoring during anesthesia and critical care: a guide for the clinician. J Clin Monit Comput 2025; 39:315-348. [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] [MESH Headings] [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 A 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
|
8
|
Li J, Wu Y, Wang Y, Wu Y, Hu R, Long S, Huang W, Nie L, Wang Z. Activation of Glutamatergic Neurons in the Supramammillary Nucleus Promotes the Recovery of Consciousness under Sevoflurane Anesthesia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2406959. [PMID: 40167172 DOI: 10.1002/advs.202406959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 03/04/2025] [Indexed: 04/02/2025]
Abstract
Volatile anesthetics have been widely applied during surgery, but the potential mechanisms by which they influence loss of consciousness (LOC), anesthesia maintenance, and recovery of consciousness (ROC) from anesthesia remain largely unknown. Recent studies have suggested that anesthesia-induced unconsciousness may be due to specific interactions between neural circuits that regulate sleep and wakefulness. Supramammillary (SuM) glutamatergic neurons are essential for sleep-wakefulness regulation. However, whether SuM glutamatergic neurons are involved in the modulation of consciousness under sevoflurane anesthesia is unclear. Here, it is shown that the activity of SuM glutamatergic neurons decreased prior to sevoflurane-induced LOC and gradually increased following ROC. Selective lesioning of SuM glutamatergic neurons promoted the induction of and delayed emergence from sevoflurane anesthesia and increased sevoflurane sensitivity. In addition, optogenetic stimulation of SuM glutamatergic neurons or the SuM-MS projection promoted behavioral arousal and cortical activation under steady-state sevoflurane anesthesia (SSSA) and reduced the depth of anesthesia and caused cortical arousal under sevoflurane-induced burst-suppression conditions. Collectively, these results provide compelling evidence that SuM glutamatergic neurons contribute to regulating states of consciousness under sevoflurane anesthesia.
Collapse
Affiliation(s)
- Jiayan Li
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Yehui Wu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Yihan Wang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Yumin Wu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Rong Hu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Si Long
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Wenqi Huang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Liming Nie
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, China
| | - Zhongxing Wang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| |
Collapse
|
9
|
Yang L, Fang F, Wang WX, Xie Y, Cang J, Li SB. Substantia Innominata Glutamatergic Neurons Modulate Sevoflurane Anesthesia in Male Mice. Anesth Analg 2025; 140:353-365. [PMID: 39008422 DOI: 10.1213/ane.0000000000007092] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
BACKGROUND Accumulated evidence suggests that brain regions that promote wakefulness also facilitate emergence from general anesthesia (GA). Glutamatergic neurons in the substantia innominata (SI) regulate motivation-related aversive, depressive, and aggressive behaviors relying on heightened arousal. Here, we hypothesize that glutamatergic neurons in the SI are also involved in the regulation of the effects of sevoflurane anesthesia. METHODS With a combination of fiber photometry, chemogenetic and optogenetic tools, behavioral tests, and cortical electroencephalogram recordings, we investigated whether and how SI glutamatergic neurons and their projections to the lateral hypothalamus (LH) regulate sevoflurane anesthesia in adult male mice. RESULTS Population activity of glutamatergic neurons in the SI gradually decreased upon sevoflurane-induced loss of consciousness (LOC) and slowly returned as soon as inhalation of sevoflurane discontinued before recovery of consciousness (ROC). Chemogenetic activation of SI glutamatergic neurons dampened the animals' sensitivity to sevoflurane exposure, prolonged induction time (mean ± standard deviation [SD]; 389 ± 67 seconds vs 458 ± 53 seconds; P = .047), and shortened emergence time (305 seconds, 95% confidence interval [CI], 242-369 seconds vs 207 seconds, 95% CI, 135-279 seconds; P = .004), whereas chemogenetic inhibition of these neurons facilitated sevoflurane anesthesia. Furthermore, optogenetic activation of SI glutamatergic neurons and their terminals in LH induced cortical activation and behavioral emergence from different depths of sevoflurane anesthesia. CONCLUSIONS Our study shows that SI glutamatergic neuronal activity facilitates emergence from sevoflurane anesthesia and provides evidence for the involvement of the SI-LH glutamatergic pathway in the regulation of consciousness during GA.
Collapse
Affiliation(s)
- Li Yang
- From the Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fang Fang
- From the Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wen-Xu Wang
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, Frontiers Center for Brain Science of the Ministry of Education (MOE), Fudan University, Shanghai, China
| | - Yunli Xie
- Department of Anesthesiology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Cang
- From the Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shi-Bin Li
- Department of Anesthesiology, Zhongshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, China
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Wang Z, Li L, Li M, Lu Z, Qin L, Naumann RK, Wang H. Chemogenetic Modulation of Preoptic Gabre Neurons Decreases Body Temperature and Heart Rate. Int J Mol Sci 2024; 25:13061. [PMID: 39684772 DOI: 10.3390/ijms252313061] [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: 09/30/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 12/18/2024] Open
Abstract
The preoptic area of the hypothalamus is critical for regulation of brain-body interaction, including circuits that control vital signs such as body temperature and heart rate. The preoptic area contains approximately 70 molecularly distinct cell types. The Gabre gene is expressed in a subset of preoptic area cell types. It encodes the GABA receptor ε-subunit, which is thought to confer resistance to anesthetics at the molecular level, but the function of Gabre cells in the brain remains largely unknown. We generated and have extensively characterized a Gabre-cre knock-in mouse line and used chemogenetic tools to interrogate the function of Gabre cells in the preoptic area. Comparison with macaque GABRE expression revealed the conserved character of Gabre cells in the preoptic area. In awake mice, we found that chemogenetic activation of Gabre neurons in the preoptic area reduced body temperature, whereas chemogenetic inhibition had no effect. Furthermore, chemogenetic inhibition of Gabre neurons in the preoptic area decreased the heart rate, whereas chemogenetic activation had no effect under isoflurane anesthesia. These findings suggest an important role of preoptic Gabre neurons in maintaining vital signs such as body temperature and heart rate during wakefulness and under anesthesia.
Collapse
Affiliation(s)
- Ziyue Wang
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Lanxiang Li
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Miao Li
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Zhonghua Lu
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Lihua Qin
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Robert Konrad Naumann
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Hong Wang
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| |
Collapse
|
13
|
Zheng Z, Liu Y, Mu R, Guo X, Feng Y, Guo C, Yang L, Qiu W, Zhang Q, Yang W, Dong Z, Qiu S, Dong Y, Cui Y. A small population of stress-responsive neurons in the hypothalamus-habenula circuit mediates development of depression-like behavior in mice. Neuron 2024; 112:3924-3939.e5. [PMID: 39389052 DOI: 10.1016/j.neuron.2024.09.012] [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: 12/11/2023] [Revised: 06/25/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
Accumulating evidence has shown that various brain functions are associated with experience-activated neuronal ensembles. However, whether such neuronal ensembles are engaged in the pathogenesis of stress-induced depression remains elusive. Utilizing activity-dependent viral strategies in mice, we identified a small population of stress-responsive neurons, primarily located in the middle part of the lateral hypothalamus (mLH) and the medial part of the lateral habenula (LHbM). These neurons serve as "starter cells" to transmit stress-related information and mediate the development of depression-like behaviors during chronic stress. Starter cells in the mLH and LHbM form dominant connections, which are selectively potentiated by chronic stress. Silencing these connections during chronic stress prevents the development of depression-like behaviors, whereas activating these connections directly elicits depression-like behaviors without stress experience. Collectively, our findings dissect a core functional unit within the LH-LHb circuit that mediates the development of depression-like behaviors in mice.
Collapse
Affiliation(s)
- Zhiwei Zheng
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Yiqin Liu
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Ruiqi Mu
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Xiaonan Guo
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Yirong Feng
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Chen Guo
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Liang Yang
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Wenxi Qiu
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Qi Zhang
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
| | - Wei Yang
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhaoqi Dong
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Shuang Qiu
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China
| | - Yiyan Dong
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China.
| | - Yihui Cui
- Department of Psychiatry of Sir Run Shaw Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Neurology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
14
|
Hu Y, Wang Y, Zhang L, Luo M, Wang Y. Neural Network Mechanisms Underlying General Anesthesia: Cortical and Subcortical Nuclei. Neurosci Bull 2024; 40:1995-2011. [PMID: 39168960 PMCID: PMC11625048 DOI: 10.1007/s12264-024-01286-z] [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: 02/22/2024] [Accepted: 06/10/2024] [Indexed: 08/23/2024] Open
Abstract
General anesthesia plays a significant role in modern medicine. However, the precise mechanism of general anesthesia remains unclear, posing a key scientific challenge in anesthesiology. Advances in neuroscience techniques have enabled targeted manipulation of specific neural circuits and the capture of brain-wide neural activity at high resolution. These advances hold promise for elucidating the intricate mechanisms of action of general anesthetics. This review aims to summarize our current understanding of the role of cortical and subcortical nuclei in modulating general anesthesia, providing new evidence of cortico-cortical and thalamocortical networks in relation to anesthesia and consciousness. These insights contribute to a comprehensive understanding of the neural network mechanisms underlying general anesthesia.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yun Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Lingjing Zhang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Mengqiang Luo
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
15
|
Lu D, Uldry Lavergne CG, Choi S, Park J, Kim J, Zhao S, Desimone Q, Lendaro E, Chen B, Han BX, Wang F, Goldstein N. General anesthesia activates a central anxiolytic center in the BNST. Cell Rep 2024; 43:114909. [PMID: 39460938 PMCID: PMC11645888 DOI: 10.1016/j.celrep.2024.114909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Low doses of general anesthetics like ketamine and dexmedetomidine have anxiolytic properties independent of their sedative effects, but the underlying mechanisms remain unclear. We discovered a population of GABAergic neurons in the oval division of the bed nucleus of the stria terminalis that are activated by multiple anesthetics and the anxiolytic drug diazepam (ovBNSTGA). The majority of ovBNSTGA neurons express neurotensin receptor 1 (Ntsr1) and form circuits with brain regions known to regulate anxiety and stress responses. Optogenetic activation of ovBNSTGA or ovBNSTNtsr1 neurons significantly attenuated anxiety-like behaviors in both naive animals and mice with inflammatory pain, while inhibition of these cells elevated anxiety. Activation of these neurons decreased heart rate and increased heart rate variability, suggesting that they reduce anxiety by modulating autonomic responses. Our study identifies ovBNSTGA/ovBNSTNtsr1 neurons as a common neural substrate mediating the anxiolytic effect of low-dose anesthetics and a potential therapeutic target for treating anxiety-related disorders.
Collapse
Affiliation(s)
- Dongye Lu
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Camille G Uldry Lavergne
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Seonmi Choi
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jaehong Park
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Jiwoo Kim
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Shengli Zhao
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Quinn Desimone
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eva Lendaro
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bin Chen
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bao-Xia Han
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Fan Wang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Nitsan Goldstein
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
16
|
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
|
17
|
Zhang Y, Li J, Li Y, Wang W, Wang D, Ding J, Wang L, Cheng J. Dexmedetomidine Promotes NREM Sleep by Depressing Oxytocin Neurons in the Paraventricular Nucleus in Mice. Neurochem Res 2024; 49:2926-2939. [PMID: 39078522 DOI: 10.1007/s11064-024-04221-w] [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: 04/02/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Dexmedetomidine (DEX) is a highly selective α2-adrenoceptor agonist with sedative effects on sleep homeostasis. Oxytocin-expressing (OXT) neurons in the paraventricular nucleus (PVN) of the hypothalamus (PVNOXT) regulate sexual reproduction, drinking, sleep-wakefulness, and other instinctive behaviors. To investigate the effect of DEX on the activity and signal transmission of PVNOXT in regulating the sleep-wakefulness cycle. Here, we employed OXT-cre mice to selectively target and express the designer receptors exclusively activated by designer drugs (DREADD)-based chemogenetic tool hM3D(Gq) in PVNOXT neurons. Combining chemogenetic methods with electroencephalogram (EEG) /electromyogram (EMG) recordings, we found that cannula injection of DEX in PVN significantly increased the duration of non-rapid eye movement (NREM) sleep in mice. Furthermore, the chemogenetic activation of PVNOXT neurons using i.p. injection of clozapine N-oxide (CNO) after cannula injection of DEX to PVN led to a substantial increase in wakefulness. Electrophysiological results showed that DEX decreased the frequency of action potential (AP) and the spontaneous excitatory postsynaptic current (sEPSC) of PVNOXT neurons through α2-adrenoceptors. Therefore, these results identify that DEX promotes sleep and maintains sleep homeostasis by inhibiting PVNOXT neurons through the α2-adrenoceptor.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Jiaxin Li
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yan Li
- Department of Pharmacy, Linquan People's Hospital, Linquan, 236400, Anhui, China
| | - Wei Wang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Daming Wang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Junli Ding
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Licheng Wang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China.
- College of Stomatology, Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Juan Cheng
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
18
|
Fettiplace MR, Vincent KF, Cho A, Dillon E, Stapley BM, Stewart V, Solt K. Dopaminergic psychostimulants cause arousal from isoflurane-induced sedation without reversing memory impairment in rats. Br J Anaesth 2024; 133:793-803. [PMID: 38965013 PMCID: PMC11443133 DOI: 10.1016/j.bja.2024.05.026] [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: 03/15/2024] [Revised: 04/30/2024] [Accepted: 05/04/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Dopaminergic psychostimulants can restore arousal in anaesthetised animals, and dopaminergic signalling contributes to hippocampal-dependent memory formation. We tested the hypothesis that dopaminergic psychostimulants can antagonise the amnestic effects of isoflurane on visuospatial working memory. METHODS Sixteen adult Sprague-Dawley rats were trained on a trial-unique nonmatching-to-location (TUNL) task which assessed the ability to identify a novel touchscreen location after a fixed delay. Once trained, the effects of low-dose isoflurane (0.3 vol%) on task performance and activity, assessed by infrared beam breaks, were assessed. We attempted to rescue deficits in performance and activity with a dopamine D1 receptor agonist (chloro-APB), a noradrenergic reuptake inhibitor (atomoxetine), and a mixed dopamine/norepinephrine releasing agent (dextroamphetamine). Anaesthetic induction, emergence, and recovery from anaesthesia were also investigated. RESULTS Low-dose isoflurane impaired working memory in a sex-independent and intra-trial delay-independent manner as assessed by task performance, and caused an overall reduction in activity. Administration of chloro-APB, atomoxetine, or dextroamphetamine did not restore visuospatial working memory, but chloro-APB and dextroamphetamine recovered arousal to levels observed in the baseline awake state. Performance did not differ between induction and emergence. Animals recovered to baseline performance within 15 min of discontinuing isoflurane. CONCLUSIONS Low-dose isoflurane impairs visuospatial working memory in a nondurable and delay-independent manner that potentially implicates non-hippocampal structures in isoflurane-induced memory deficits. Dopaminergic psychostimulants counteracted sedation but did not reverse memory impairments, suggesting that isoflurane-induced amnesia and isoflurane-induced sedation have distinct underlying mechanisms that can be antagonised independently.
Collapse
Affiliation(s)
- Michael R Fettiplace
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA.
| | - Kathleen F Vincent
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Angel Cho
- Touro College of Osteopathic Medicine, New York, NY, USA
| | - Emmaline Dillon
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, USA
| | - Brendan M Stapley
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, USA
| | - Victoria Stewart
- University of California Irvine School of Medicine, Irvine, CA, USA
| | - Ken Solt
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Zhao J, Furutani K, McGinnis A, Mathew JP, Wang F, Ji RR. Distinct roles of general anesthesia activated CeA neurons in acute versus late phase of neuropathic pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612553. [PMID: 39314433 PMCID: PMC11418996 DOI: 10.1101/2024.09.11.612553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
A previous study discovered a distinct population of GABAergic neurons in the ce ntral a mygdala (CeA) that can be activated by g eneral a nesthesia (CeA GA ) and exert analgesic functions (Hua et al., 2020). To independently reproduce these prior findings and to investigate the electrophysiological properties of CeA GA neurons, we first used 1.2% isoflurane to induce c-Fos activation in the mouse brain and validated the Fos expression by RNAscope in situ hybridization. Indeed, isoflurane induced robust Fos expression in CeA and these Fos + CeA GA neurons are GABAergic neurons (Vgat + ). We next used Fos-TRAP2 method (different from the CANE method used in the prior study) to label CeA GA neurons (tdTomato + ). Our ex vivo electrophysiological recordings in brain slices revealed that compared to Fos-negative CeA neurons, CeA GA neurons had significantly higher excitability and exhibited distinct patterns of action potentials. Chemogenetic activation of Fos-TRAPed CeA GA neurons was effective at increasing pain thresholds in naïve mice and mice with early-phase neuropathic pain 2 weeks after spared nerve injury (SNI). However, the same chemogenetic activation of CeA GA neurons only had modest analgesia in the late phase of SNI at 8 weeks, although it was highly effective in reducing chronic pain-associated anxiety behaviors at this stage. We found that Fos-negative CeA neurons, but not CeA GA neurons, exhibited increased excitability in the late-phase of SNI, suggesting that chronic pain causes a shift in the relative activity of the CeA microcircuit. Interestingly, Fos-negative neurons exhibited much higher expression of K + -Cl - cotransporter-2 (KCC2), and KCC2 expression was downregulated in the CeA in the late-phase of neuropathic pain. These results support the idea that targeting CeA GA neurons may provide therapeutic benefits for pain relief and chronic pain-associated anxiety. Our findings also suggest distinct roles of CeA GA neurons in regulating physiological pain, acute pain, and chronic pain with a possible involvement of KCC2.
Collapse
|
20
|
Zhao Y, Ou M, Liu J, Jiang J, Zhang D, Ke B, Wu Y, Chen Y, Jiang R, Hemmings HC, Zhu T, Zhou C. Astrocytes Modulate a Specific Paraventricular Thalamus→Prefrontal Cortex Projection to Enhance Consciousness Recovery from Anesthesia. J Neurosci 2024; 44:e1808232024. [PMID: 38926088 PMCID: PMC11340278 DOI: 10.1523/jneurosci.1808-23.2024] [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: 09/23/2023] [Revised: 03/30/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024] Open
Abstract
Current anesthetic theory is mostly based on neurons and/or neuronal circuits. A role for astrocytes also has been shown in promoting recovery from volatile anesthesia, while the exact modulatory mechanism and/or the molecular target in astrocytes is still unknown. In this study by animal models in male mice and electrophysiological recordings in vivo and in vitro, we found that activating astrocytes of the paraventricular thalamus (PVT) and/or knocking down PVT astrocytic Kir4.1 promoted the consciousness recovery from sevoflurane anesthesia. Single-cell RNA sequencing of the PVT reveals two distinct cellular subtypes of glutamatergic neurons: PVT GRM and PVT ChAT neurons. Patch-clamp recording results proved astrocytic Kir4.1-mediated modulation of sevoflurane on the PVT mainly worked on PVT ChAT neurons, which projected mainly to the mPFC. In summary, our findings support the novel conception that there is a specific PVT→prefrontal cortex projection involved in consciousness recovery from sevoflurane anesthesia, which is mediated by the inhibition of sevoflurane on PVT astrocytic Kir4.1 conductance.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
- Research Institution of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Mengchan Ou
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
- Research Institution of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
- Research Institution of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jingyao Jiang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
- Research Institution of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Donghang Zhang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
- Research Institution of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Bowen Ke
- Research Institution of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yujie Wu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
- Research Institution of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yali Chen
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
- Research Institution of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ruotian Jiang
- Research Institution of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Hugh C Hemmings
- Departments of Anesthesiology and Pharmacology, Weill Cornell Medicine, New York, New York 10065
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Cheng Zhou
- Research Institution of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
O’Reilly-Fong J, Simpson NJ, Thirouin ZS, Bastone PA, Zaelzer C, Murtaz A, Bourque CW. Acute and Reversible Hypothalamic Symptoms in a Lateral Head Impact Mouse Model of Mild Traumatic Brain Injury. Neurotrauma Rep 2024; 5:749-759. [PMID: 39184177 PMCID: PMC11342051 DOI: 10.1089/neur.2024.0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024] Open
Abstract
Central autonomic and endocrine dysfunctions following traumatic brain injury (TBI) are believed to involve the hypothalamus; however, underlying mechanisms are unknown. Although chronic deficits might be caused by irreversible tissue damage, various neuroendocrine and autonomic symptoms are only observed transiently, suggesting they might result from a temporary alteration in the activity of hypothalamic neurons. We therefore examined if a mouse model of mild TBI could induce reversible autonomic phenotypes and cause acute changes in c-Fos expression within corresponding regions of the hypothalamus. Adult C57Bl/6 male mice were lightly anesthetized with isoflurane and subjected to TBI by lateral head impact using a Gothenburg impactor. Mice treated the same way, but without the head impact served as controls (shams). We monitored body weight and core body temperature by infrared thermography and performed immunohistochemistry against c-Fos in various regions of the hypothalamus. We determined that a projectile velocity of 9 m/s significantly delayed recovery from the anesthesia without inducing skull fractures and signs of discomfort disappeared within 3 h, as assessed by grimace scale. Compared with shams, TBI mice displayed a rapid decrease in core body temperature which resolved within 48 h. Daily body weight gain was also significantly lower in TBI mice on the day following injury but recovered thereafter. c-Fos analysis revealed a significantly higher density of c-Fos-positive cells in the paraventricular nucleus and a significantly lower density in the median preoptic nucleus and medial preoptic area. We conclude that mild TBI induced by a single lateral head impact in mice at 9 m/s produces acute and reversible symptoms associated with hypothalamic dysfunction accompanied by significant changes in c-Fos expression within relevant areas of the hypothalamus. These findings support the hypothesis that a temporary alteration of neuronal activity may underlie the expression of reversible central autonomic and neuroendocrine symptoms.
Collapse
Affiliation(s)
- Julie O’Reilly-Fong
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Nick J. Simpson
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Zahra S. Thirouin
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Paolo A. Bastone
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Cristian Zaelzer
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Anzala Murtaz
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Charles W. Bourque
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| |
Collapse
|
22
|
Khan S, Huang Y, Timuçin D, Bailey S, Lee S, Lopes J, Gaunce E, Mosberger J, Zhan M, Abdelrahman B, Zeng X, Wiest MC. Microtubule-Stabilizer Epothilone B Delays Anesthetic-Induced Unconsciousness in Rats. eNeuro 2024; 11:ENEURO.0291-24.2024. [PMID: 39147581 PMCID: PMC11363512 DOI: 10.1523/eneuro.0291-24.2024] [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: 07/01/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Volatile anesthetics are currently believed to cause unconsciousness by acting on one or more molecular targets including neural ion channels, receptors, mitochondria, synaptic proteins, and cytoskeletal proteins. Anesthetic gases including isoflurane bind to cytoskeletal microtubules (MTs) and dampen their quantum optical effects, potentially contributing to causing unconsciousness. This possibility is supported by the finding that taxane chemotherapy consisting of MT-stabilizing drugs reduces the effectiveness of anesthesia during surgery in human cancer patients. In order to experimentally assess the contribution of MTs as functionally relevant targets of volatile anesthetics, we measured latencies to loss of righting reflex (LORR) under 4% isoflurane in male rats injected subcutaneously with vehicle or 0.75 mg/kg of the brain-penetrant MT-stabilizing drug epothilone B (epoB). EpoB-treated rats took an average of 69 s longer to become unconscious as measured by latency to LORR. This was a statistically significant difference corresponding to a standardized mean difference (Cohen's d) of 1.9, indicating a "large" normalized effect size. The effect could not be accounted for by tolerance from repeated exposure to isoflurane. Our results suggest that binding of the anesthetic gas isoflurane to MTs causes unconsciousness and loss of purposeful behavior in rats (and presumably humans and other animals). This finding is predicted by models that posit consciousness as a property of a quantum physical state of neural MTs.
Collapse
Affiliation(s)
- Sana Khan
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts 01760
| | - Yixiang Huang
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts 01760
| | - Derin Timuçin
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts 01760
| | - Shantelle Bailey
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts 01760
| | - Sophia Lee
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts 01760
| | - Jessica Lopes
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts 01760
| | - Emeline Gaunce
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts 01760
| | - Jasmine Mosberger
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts 01760
| | - Michelle Zhan
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts 01760
| | | | - Xiran Zeng
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts 01760
| | - Michael C Wiest
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts 01760
| |
Collapse
|
23
|
Onishi T, Hirose K, Sakaba T. Molecular tools to capture active neural circuits. Front Neural Circuits 2024; 18:1449459. [PMID: 39100199 PMCID: PMC11294111 DOI: 10.3389/fncir.2024.1449459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
To understand how neurons and neural circuits function during behaviors, it is essential to record neuronal activity in the brain in vivo. Among the various technologies developed for recording neuronal activity, molecular tools that induce gene expression in an activity-dependent manner have attracted particular attention for their ability to clarify the causal relationships between neuronal activity and behavior. In this review, we summarize recently developed activity-dependent gene expression tools and their potential contributions to the study of neural circuits.
Collapse
Affiliation(s)
- Taichi Onishi
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo City, Bunkyo, Japan
| | - Kenzo Hirose
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo City, Bunkyo, Japan
| | - Takeshi Sakaba
- Graduate School of Brain Science, Doshisha University, Kyotanabe, Kyoto, Japan
| |
Collapse
|
24
|
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.
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
|
25
|
Chen S, Li B, Hu Y, Zhang Y, Dai W, Zhang X, Zhou Y, Su D. Common functional mechanisms underlying dynamic brain network changes across five general anesthetics: A rat fMRI study. CNS Neurosci Ther 2024; 30:e14866. [PMID: 39014472 PMCID: PMC11251872 DOI: 10.1111/cns.14866] [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: 04/26/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Reversible loss of consciousness is the primary therapeutic endpoint of general anesthesia; however, the drug-invariant mechanisms underlying anesthetic-induced unconsciousness are still unclear. This study aimed to investigate the static, dynamic, topological and organizational changes in functional brain network induced by five clinically-used general anesthetics in the rat brain. METHOD Male Sprague-Dawley rats (n = 57) were randomly allocated to received propofol, isoflurane, ketamine, dexmedetomidine, or combined isoflurane plus dexmedetomidine anesthesia. Resting-state functional magnetic resonance images were acquired under general anesthesia and analyzed for changes in dynamic functional brain networks compared to the awake state. RESULTS Different general anesthetics induced distinct patterns of functional connectivity inhibition within brain-wide networks, resulting in multi-level network reorganization primarily by impairing the functional connectivity of cortico-subcortical networks as well as by reducing information transmission capacity, intrinsic connectivity, and network architecture stability of subcortical regions. Conversely, functional connectivity and topological properties were preserved within cortico-cortical networks, albeit with fewer dynamic fluctuations under general anesthesia. CONCLUSIONS Our findings highlighted the effects of different general anesthetics on functional brain network reorganization, which might shed light on the drug-invariant mechanism of anesthetic-induced unconsciousness.
Collapse
Affiliation(s)
- Sifan Chen
- Department of Anesthesiology, Renji HospitalSchool of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of EducationShanghaiChina
- Department of RadiologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Bo Li
- Department of Anesthesiology, Renji HospitalSchool of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of EducationShanghaiChina
- Department of Radiology, Renji HospitalSchool of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Ying Hu
- Department of Radiology, Renji HospitalSchool of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Yizhe Zhang
- Department of Anesthesiology, Renji HospitalSchool of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of EducationShanghaiChina
| | - Wanbing Dai
- Department of Anesthesiology, Renji HospitalSchool of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of EducationShanghaiChina
| | - Xiao Zhang
- Department of Anesthesiology, Renji HospitalSchool of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of EducationShanghaiChina
| | - Yan Zhou
- Department of Radiology, Renji HospitalSchool of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
| | - Diansan Su
- Department of Anesthesiology, Renji HospitalSchool of Medicine, Shanghai Jiao Tong UniversityShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of EducationShanghaiChina
| |
Collapse
|
26
|
Nagayama S, Hasegawa-Ishii S, Kikuta S. Anesthetized animal experiments for neuroscience research. Front Neural Circuits 2024; 18:1426689. [PMID: 38884008 PMCID: PMC11177690 DOI: 10.3389/fncir.2024.1426689] [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: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Brain research has progressed with anesthetized animal experiments for a long time. Recent progress in research techniques allows us to measure neuronal activity in awake animals combined with behavioral tasks. The trends became more prominent in the last decade. This new research style triggers the paradigm shift in the research of brain science, and new insights into brain function have been revealed. It is reasonable to consider that awake animal experiments are more ideal for understanding naturalistic brain function than anesthetized ones. However, the anesthetized animal experiment still has advantages in some experiments. To take advantage of the anesthetized animal experiments, it is important to understand the mechanism of anesthesia and carefully handle the obtained data. In this minireview, we will shortly summarize the molecular mechanism of anesthesia in animal experiments, a recent understanding of the neuronal activities in a sensory system in the anesthetized animal brain, and consider the advantages and disadvantages of the anesthetized and awake animal experiments. This discussion will help us to use both research conditions in the proper manner.
Collapse
Affiliation(s)
- Shin Nagayama
- Department of Neurobiology and Anatomy, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sanae Hasegawa-Ishii
- Pathology Research Team, Faculty of Health Sciences, Kyorin University, Mitaka, Japan
| | - Shu Kikuta
- Department of Otorhinolaryngology, Medical School of Nihon University, Tokyo, Japan
| |
Collapse
|
27
|
Mashour GA. Anesthesia and the neurobiology of consciousness. Neuron 2024; 112:1553-1567. [PMID: 38579714 PMCID: PMC11098701 DOI: 10.1016/j.neuron.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 04/07/2024]
Abstract
In the 19th century, the discovery of general anesthesia revolutionized medical care. In the 21st century, anesthetics have become indispensable tools to study consciousness. Here, I review key aspects of the relationship between anesthesia and the neurobiology of consciousness, including interfaces of sleep and anesthetic mechanisms, anesthesia and primary sensory processing, the effects of anesthetics on large-scale functional brain networks, and mechanisms of arousal from anesthesia. I discuss the implications of the data derived from the anesthetized state for the science of consciousness and then conclude with outstanding questions, reflections, and future directions.
Collapse
Affiliation(s)
- George A Mashour
- Center for Consciousness Science, Department of Anesthesiology, Department of Pharmacology, Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
28
|
Song XJ, Hu JJ. Neurobiological basis of emergence from anesthesia. Trends Neurosci 2024; 47:355-366. [PMID: 38490858 DOI: 10.1016/j.tins.2024.02.006] [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: 10/12/2023] [Revised: 01/25/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024]
Abstract
The suppression of consciousness by anesthetics and the emergence of the brain from anesthesia are complex and elusive processes. Anesthetics may exert their inhibitory effects by binding to specific protein targets or through membrane-mediated targets, disrupting neural activity and the integrity and function of neural circuits responsible for signal transmission and conscious perception/subjective experience. Emergence from anesthesia was generally thought to depend on the elimination of the anesthetic from the body. Recently, studies have suggested that emergence from anesthesia is a dynamic and active process that can be partially controlled and is independent of the specific molecular targets of anesthetics. This article summarizes the fundamentals of anesthetics' actions in the brain and the mechanisms of emergence from anesthesia that have been recently revealed in animal studies.
Collapse
Affiliation(s)
- Xue-Jun Song
- Department of Medical Neuroscience and SUSTech Center for Pain Medicine, Southern University of Science and Technology School of Medicine, Shenzhen, China.
| | - Jiang-Jian Hu
- Department of Medical Neuroscience and SUSTech Center for Pain Medicine, Southern University of Science and Technology School of Medicine, Shenzhen, China
| |
Collapse
|
29
|
Sakurai K. Rethinking c-Fos for understanding drug action in the brain. J Biochem 2024; 175:377-381. [PMID: 38153290 DOI: 10.1093/jb/mvad110] [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: 09/04/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 12/29/2023] Open
Abstract
Understanding the mechanisms of drug action in the brain, from the genetic to the neural circuit level, is crucial for the development of new agents that act upon the central nervous system. Determining the brain regions and neurons affected by a drug is essential for revealing its mechanism of action in the brain. c-Fos, a marker of neuronal activation, has been widely used to detect neurons activated by stimuli with high spatial resolution. In this review, the use of c-Fos for the visualization and manipulation of activated neurons is introduced. I also explain that a higher temporal resolution can be achieved by changing the staining method for visualization of c-Fos. Moreover, a new method that allows labeling and manipulating commonly activated neurons using two different stimuli is proposed.
Collapse
Affiliation(s)
- Katsuyasu Sakurai
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
30
|
Hu Y, Du W, Qi J, Luo H, Zhang Z, Luo M, Wang Y. Comparative brain-wide mapping of ketamine- and isoflurane-activated nuclei and functional networks in the mouse brain. eLife 2024; 12:RP88420. [PMID: 38512722 PMCID: PMC10957177 DOI: 10.7554/elife.88420] [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] [Indexed: 03/23/2024] Open
Abstract
Ketamine (KET) and isoflurane (ISO) are two widely used general anesthetics, yet their distinct and shared neurophysiological mechanisms remain elusive. In this study, we conducted a comparative analysis of the effects of KET and ISO on c-Fos expression across the mouse brain, utilizing hierarchical clustering and c-Fos-based functional network analysis to evaluate the responses of individual brain regions to each anesthetic. Our findings reveal that KET activates a wide range of brain regions, notably in the cortical and subcortical nuclei involved in sensory, motor, emotional, and reward processing, with the temporal association areas (TEa) as a strong hub, suggesting a top-down mechanism affecting consciousness by primarily targeting higher order cortical networks. In contrast, ISO predominantly influences brain regions in the hypothalamus, impacting neuroendocrine control, autonomic function, and homeostasis, with the locus coeruleus (LC) as a connector hub, indicating a bottom-up mechanism in anesthetic-induced unconsciousness. KET and ISO both activate brain areas involved in sensory processing, memory and cognition, reward and motivation, as well as autonomic and homeostatic control, highlighting their shared effects on various neural pathways. In conclusion, our results highlight the distinct but overlapping effects of KET and ISO, enriching our understanding of the mechanisms underlying general anesthesia.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Wenjie Du
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Jiangtao Qi
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Huoqing Luo
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
| | - Zhao Zhang
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Mengqiang Luo
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
31
|
Yang D, Wang Y, Qi T, Zhang X, Shen L, Ma J, Pang Z, Lal NK, McClatchy DB, Seradj SH, Leung VH, Wang K, Xie Y, Polli FS, Maximov A, Gonzalez OC, de Lecea L, Cline HT, Augustine V, Yates JR, Ye L. Phosphorylation of pyruvate dehydrogenase inversely associates with neuronal activity. Neuron 2024; 112:959-971.e8. [PMID: 38266644 PMCID: PMC11021214 DOI: 10.1016/j.neuron.2023.12.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 08/24/2023] [Accepted: 12/21/2023] [Indexed: 01/26/2024]
Abstract
For decades, the expression of immediate early genes (IEGs) such as FOS has been the most widely used molecular marker representing neuronal activation. However, to date, there is no equivalent surrogate available for the decrease of neuronal activity. Here, we developed an optogenetic-based biochemical screen in which population neural activities can be controlled by light with single action potential precision, followed by unbiased phosphoproteomic profiling. We identified that the phosphorylation of pyruvate dehydrogenase (pPDH) inversely correlated with the intensity of action potential firing in primary neurons. In in vivo mouse models, monoclonal antibody-based pPDH immunostaining detected activity decreases across the brain, which were induced by a wide range of factors including general anesthesia, chemogenetic inhibition, sensory experiences, and natural behaviors. Thus, as an inverse activity marker (IAM) in vivo, pPDH can be used together with IEGs or other cell-type markers to profile and identify bi-directional neural dynamics induced by experiences or behaviors.
Collapse
Affiliation(s)
- Dong Yang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yu Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tianbo Qi
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xi Zhang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Leyao Shen
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jingrui Ma
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Neurobiology, University of California San Diego, La Jolla, CA 92093, USA
| | - Zhengyuan Pang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Neeraj K Lal
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Daniel B McClatchy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Saba Heydari Seradj
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Verina H Leung
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kristina Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yi Xie
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Filip S Polli
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Anton Maximov
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Hollis T Cline
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vineet Augustine
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Neurobiology, University of California San Diego, La Jolla, CA 92093, USA
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Li Ye
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
32
|
Jiang-Xie LF, Drieu A, Bhasiin K, Quintero D, Smirnov I, Kipnis J. Neuronal dynamics direct cerebrospinal fluid perfusion and brain clearance. Nature 2024; 627:157-164. [PMID: 38418877 PMCID: PMC12054998 DOI: 10.1038/s41586-024-07108-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024]
Abstract
The accumulation of metabolic waste is a leading cause of numerous neurological disorders, yet we still have only limited knowledge of how the brain performs self-cleansing. Here we demonstrate that neural networks synchronize individual action potentials to create large-amplitude, rhythmic and self-perpetuating ionic waves in the interstitial fluid of the brain. These waves are a plausible mechanism to explain the correlated potentiation of the glymphatic flow1,2 through the brain parenchyma. Chemogenetic flattening of these high-energy ionic waves largely impeded cerebrospinal fluid infiltration into and clearance of molecules from the brain parenchyma. Notably, synthesized waves generated through transcranial optogenetic stimulation substantially potentiated cerebrospinal fluid-to-interstitial fluid perfusion. Our study demonstrates that neurons serve as master organizers for brain clearance. This fundamental principle introduces a new theoretical framework for the functioning of macroscopic brain waves.
Collapse
Affiliation(s)
- Li-Feng Jiang-Xie
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA.
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
| | - Antoine Drieu
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Kesshni Bhasiin
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Daniel Quintero
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Igor Smirnov
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Jonathan Kipnis
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA.
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
| |
Collapse
|
33
|
McKinstry-Wu AR, Kelz MB. One node among many: sevoflurane-induced hypnosis and the challenge of an integrative network-level view of anaesthetic action. Br J Anaesth 2024; 132:220-223. [PMID: 38000931 DOI: 10.1016/j.bja.2023.11.002] [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: 09/21/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
Building on their known ability to influence sleep and arousal, Li and colleagues show that modulating the activity of glutamatergic pedunculopontine tegmental neurones also alters sevoflurane-induced hypnosis. This finding adds support for the shared sleep-anaesthesia circuit hypothesis. However, the expanding recognition of many neuronal clusters capable of modulating anaesthetic hypnosis raises the question of how disparate and anatomically distant sites ultimately interact to coordinate global changes in the state of the brain. Understanding how these individual sites work in concert to disrupt cognition and behaviour is the next challenge for anaesthetic mechanisms research.
Collapse
Affiliation(s)
- Andrew R McKinstry-Wu
- Department of Anaesthesiology and Critical Care, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA; Center for Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Max B Kelz
- Department of Anaesthesiology and Critical Care, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA; Center for Neuroscience of Unconsciousness and Reanimation Research Alliance, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA; Mahoney Institute of Neuroscience, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
34
|
Bárez‐López S, Bishop P, Searby D, Murphy D, Greenwood MP. Male rat hypothalamic extraretinal photoreceptor Opsin3 is sensitive to osmotic stimuli and light. J Neuroendocrinol 2024; 36:e13363. [PMID: 38192267 PMCID: PMC11475585 DOI: 10.1111/jne.13363] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/13/2023] [Accepted: 12/16/2023] [Indexed: 01/10/2024]
Abstract
The light-sensitive protein Opsin 3 (Opn3) is present throughout the mammalian brain; however, the role of Opn3 in this organ remains unknown. Since Opn3 encoded mRNA is modulated in the supraoptic and paraventricular nucleus of the hypothalamus in response to osmotic stimuli, we have explored by in situ hybridization the expression of Opn3 in these nuclei. We have demonstrated that Opn3 is present in the male rat magnocellular neurones expressing either the arginine vasopressin or oxytocin neuropeptides and that Opn3 increases in both neuronal types in response to osmotic stimuli, suggesting that Opn3 functions in both cell types and that it might be involved in regulating water balance. Using rat hypothalamic organotypic cultures, we have demonstrated that the hypothalamus is sensitive to light and that the observed light sensitivity is mediated, at least in part, by Opn3. The data suggests that hypothalamic Opn3 can mediate a light-sensitive role to regulate circadian homeostatic processes.
Collapse
Affiliation(s)
- Soledad Bárez‐López
- Translational Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
- Present address:
Instituto de Investigaciones BiomédicasConsejo Superior de Investigaciones Científicas (CSIC)—Universidad Autónoma de Madrid (UAM)MadridSpain
| | - Paul Bishop
- Translational Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
| | - Daniel Searby
- Translational Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
| | - David Murphy
- Translational Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
| | - Michael P. Greenwood
- Translational Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
| |
Collapse
|
35
|
Li J, Hu R, Tan W, Li J, Huang W, Wang Z. Activation of glutamatergic neurones in the pedunculopontine tegmental nucleus promotes cortical activation and behavioural emergence from sevoflurane-induced unconsciousness in mice. Br J Anaesth 2024; 132:320-333. [PMID: 37953203 DOI: 10.1016/j.bja.2023.08.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 07/30/2023] [Accepted: 08/26/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND The neural mechanisms underlying sevoflurane-induced loss of consciousness and recovery of consciousness after anaesthesia remain unknown. We investigated whether glutamatergic pedunculopontine tegmental nucleus (PPT) neurones are involved in the regulation of states of consciousness under sevoflurane anaesthesia. METHODS In vivo fibre photometry combined with electroencephalography (EEG)/electromyography recording was used to record changes in the activity of glutamatergic PPT neurones under sevoflurane anaesthesia. Chemogenetic and cortical EEG recordings were used to explore their roles in the induction of and emergence from sevoflurane anaesthesia. Optogenetic methods combined with EEG recordings were used to explore the roles of glutamatergic PPT neurones and of the PPT-ventral tegmental area pathway in maintenance of anaesthesia. RESULTS The population activity of glutamatergic PPT neurones was reduced before sevoflurane-induced loss of righting reflex and gradually recovered after return of righting reflex. Chemogenetic inhibition of glutamatergic PPT neurones accelerated induction of anaesthesia (hM4Di-CNO vs mCherry-CNO, 76 [17] vs 121 [27] s, P<0.0001) and delayed emergence from sevoflurane anaesthesia (278 [98] vs 145 [53] s, P<0.0001) but increased sevoflurane sensitivity. Optogenetic stimulation of glutamatergic PPT neurons or of the PPT-ventral tegmental area pathway promoted cortical activation and behavioural emergence during steady-state sevoflurane anaesthesia, reduced the depth of anaesthesia, and caused cortical arousal during sevoflurane-induced EEG burst suppression. CONCLUSIONS Glutamatergic PPT neurones regulate induction and emergence of sevoflurane anaesthesia.
Collapse
Affiliation(s)
- Jiayan Li
- Department of Anaesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rong Hu
- Department of Anaesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wulin Tan
- Department of Anaesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Li
- Department of Anaesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenqi Huang
- Department of Anaesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Zhongxing Wang
- Department of Anaesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
36
|
Wasilczuk AZ, Rinehart C, Aggarwal A, Stone ME, Mashour GA, Avidan MS, Kelz MB, Proekt A, ReCCognition Study Group. Hormonal basis of sex differences in anesthetic sensitivity. Proc Natl Acad Sci U S A 2024; 121:e2312913120. [PMID: 38190526 PMCID: PMC10801881 DOI: 10.1073/pnas.2312913120] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/20/2023] [Indexed: 01/10/2024] Open
Abstract
General anesthesia-a pharmacologically induced reversible state of unconsciousness-enables millions of life-saving procedures. Anesthetics induce unconsciousness in part by impinging upon sexually dimorphic and hormonally sensitive hypothalamic circuits regulating sleep and wakefulness. Thus, we hypothesized that anesthetic sensitivity should be sex-dependent and modulated by sex hormones. Using distinct behavioral measures, we show that at identical brain anesthetic concentrations, female mice are more resistant to volatile anesthetics than males. Anesthetic sensitivity is bidirectionally modulated by testosterone. Castration increases anesthetic resistance. Conversely, testosterone administration acutely increases anesthetic sensitivity. Conversion of testosterone to estradiol by aromatase is partially responsible for this effect. In contrast, oophorectomy has no effect. To identify the neuronal circuits underlying sex differences, we performed whole brain c-Fos activity mapping under anesthesia in male and female mice. Consistent with a key role of the hypothalamus, we found fewer active neurons in the ventral hypothalamic sleep-promoting regions in females than in males. In humans, we demonstrate that females regain consciousness and recover cognition faster than males after identical anesthetic exposures. Remarkably, while behavioral and neurocognitive measures in mice and humans point to increased anesthetic resistance in females, cortical activity fails to show sex differences under anesthesia in either species. Cumulatively, we demonstrate that sex differences in anesthetic sensitivity are evolutionarily conserved and not reflected in conventional electroencephalographic-based measures of anesthetic depth. This covert resistance to anesthesia may explain the higher incidence of unintended awareness under general anesthesia in females.
Collapse
Affiliation(s)
- Andrzej Z. Wasilczuk
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Cole Rinehart
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
| | - Adeeti Aggarwal
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA19104
| | - Martha E. Stone
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA19104
| | - George A. Mashour
- Center for Consciousness Science, Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI48105
| | - Michael S. Avidan
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO63110
| | - Max B. Kelz
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA19104
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Alex Proekt
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Neuroscience of Unconsciousness and Reanimation Research Alliance, Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA19104
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - ReCCognition Study Group
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA19104
- Center for Consciousness Science, Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI48105
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO63110
| |
Collapse
|
37
|
Huang Y, Xiao Y, Li L, Feng X, Ding W, Cai F. Propofol-induced anesthesia involves the direct inhibition of glutamatergic neurons in the lateral hypothalamus. Front Neurosci 2024; 18:1327293. [PMID: 38282977 PMCID: PMC10811086 DOI: 10.3389/fnins.2024.1327293] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Propofol is the most widely used intravenous general anesthetic; however, the neuronal circuits that mediate its anesthetic effects are still poorly understood. Glutamatergic neurons in the lateral hypothalamus have been reported to be involved in maintenance of arousal and consciousness. Using Vglut2-Cre transgenic mice, we recorded this group of cells specifically and found that propofol can directly inhibit the glutamatergic neurons, and enhance inhibitory synaptic inputs on these cells, thereby reducing neuronal excitability. Through chemogenetic interventions, we found that inhibition of these neurons increased the duration of propofol-induced anesthesia and reduced movement in the animals after the recovery of right reflex. In contrast, activating this group of cells reduced the duration of propofol anesthesia and increased the animals' locomotor activity after the recovery of right reflex. These results suggest that propofol-induced anesthesia involves the inhibition of glutamatergic neurons in the lateral hypothalamus.
Collapse
Affiliation(s)
- Yan Huang
- Department of Anesthesiology, Nanchong Central Hospital, Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Yong Xiao
- Emergency Department of the General Hospital of the Tibet Military Region, Lhasa, China
| | - Linji Li
- Department of Anesthesiology, Nanchong Central Hospital, Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Xinglong Feng
- Department of Anesthesiology, Nanchong Central Hospital, Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Weixing Ding
- Qujing Secend Peopie’s Hospital, Department of Pain, Qujing, Yunnan, China
| | - Feng Cai
- Department of Urologyand Neurocardiothoracic Surgery, 927 Hospital of the Joint Logistics Support Force of the Chinese People’s LiberationArmy, Puer, China
| |
Collapse
|
38
|
Lu H, He F, Huang Y, Wei Z. Different Doses of Dexmedetomidine Reduce Postoperative Sleep Disturbance Incidence in Patients under General Anesthesia by Elevating Serum Neurotransmitter Levels. Crit Rev Immunol 2024; 44:63-73. [PMID: 38848294 DOI: 10.1615/critrevimmunol.2024051294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Postoperative sleep disturbance is a common issue that affects recovery in patients undergoing general anesthesia. Dexmedetomidine (Dex) has a potential role in improving postoperative sleep quality. We evaluated the effects of different doses of Dex on postoperative sleep disturbance and serum neurotransmitters in patients undergoing radical gastrectomy under general anesthesia. Patients were assigned to the control, NS, and Dex (Dex-L/M/H) groups based on different treatment doses [0.2, 0.4, and 0.6 μg/(kg · h)]. The Athens Insomnia Scale (AIS) and ELISA kits were used to assess sleep disturbance and serum neurotransmitter (GABA, 5-HT, NE) levels before surgery and on postoperative days one, four, and seven. The effects of different doses on postoperative sleep disturbance incidence and serum neurotransmitter levels were analyzed by the Fisher exact test and one-way and repeated-measures ANOVA. Patients had no differences in gender, age, body mass index, operation time, and bleeding volume. Different Dex doses reduced the postoperative AIS score of patients under general anesthesia, improved their sleep, and increased serum levels of 5-HT, NE, and GABA. Furthermore, the effects were dose-dependent within the range of safe clinical use. Specifically, Dex at doses of 0.2, 0.4, and 0.6 μg/(kg · h) reduced postoperative AIS score, elevated serum neurotransmitter levels, and reduced postoperative sleep disturbance incidence. Collectively, Dex has a potential preventive effect on postoperative sleep disturbance in patients undergoing general anesthesia for radical gastrectomy. The optimal dose of Dex is between 0.2 and 0.6 μg/(kg · h), which significantly reduces the incidence of postoperative sleep disturbance and increases serum neurotransmitter levels.
Collapse
Affiliation(s)
- Huifei Lu
- Department of Medical Records Room, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise City, Guangxi, 533000, China
| | - Fei He
- Department of Anesthesia, Guangxi Baise Pingguo Aluminum Hospital, Baise City, Guangxi, 533000, China
| | - Ying Huang
- Department of Anesthesia, Medical Center of Matou Town, Pingguo City, Baise, Guangxi, 533000, China
| | - Zhongliang Wei
- Affiliated Hospital of Youjiang Medical University for Nationalities
| |
Collapse
|
39
|
Xia JM, Fan BQ, Yi XW, Ni WW, Zhou Y, Chen DD, Yi WJ, Feng LL, Xia Y, Li SS, Qu WM, Han Y, Huang ZL, Li WX. Medial Septal Glutamatergic Neurons Modulate States of Consciousness during Sevoflurane Anesthesia in Mice. Anesthesiology 2024; 140:102-115. [PMID: 37812765 DOI: 10.1097/aln.0000000000004798] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
BACKGROUND Multiple neural structures involved in maintaining wakefulness have been found to promote arousal from general anesthesia. The medial septum is a critical region that modulates arousal behavior. This study hypothesized that glutamatergic neurons in the medial septum play a crucial role in regulating states of consciousness during sevoflurane general anesthesia. METHODS Adult male mice were used in this study. The effects of sevoflurane anesthesia on neuronal activity were determined by fiber photometry. Lesions and chemogenetic manipulations were used to study the effects of the altered activity of medial septal glutamatergic neurons on anesthesia induction, emergence, and sensitivity to sevoflurane. Optogenetic stimulation was used to observe the role of acute activation of medial septal glutamatergic neurons on cortical activity and behavioral changes during sevoflurane-induced continuous steady state of general anesthesia and burst suppression state. RESULTS The authors found that medial septal glutamatergic neuronal activity decreased during sevoflurane anesthesia induction and recovered in the early period of emergence. Chemogenetic activation of medial septal glutamatergic neurons prolonged the induction time (mean ± SD, hM3Dq-clozapine N-oxide vs. hM3Dq-saline, 297.5 ± 60.1 s vs. 229.4 ± 29.9 s, P < 0.001, n = 11) and decreased the emergence time (53.2 ± 11.8 s vs. 77.5 ± 33.5 s, P = 0.025, n = 11). Lesions or chemogenetic inhibition of these neurons produced the opposite effects. During steady state of general anesthesia and deep anesthesia-induced burst suppression state, acute optogenetic activation of medial septal glutamatergic neurons induced cortical activation and behavioral emergence. CONCLUSIONS The study findings reveal that activation of medial septal glutamatergic neurons has arousal-promoting effects during sevoflurane anesthesia in male mice. The activation of these neurons prolongs the induction and accelerates the emergence of anesthesia. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Jun-Ming Xia
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Bing-Qian Fan
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China; Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiu-Wen Yi
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Wen-Wen Ni
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Yu Zhou
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Dan-Dan Chen
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Wen-Jing Yi
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Li-Li Feng
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Ying Xia
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Shuang-Shuang Li
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yuan Han
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wen-Xian Li
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| |
Collapse
|
40
|
He Y, Liu T, He Q, Ke W, Li X, Du J, Deng S, Shu Z, Wu J, Yang B, Wang Y, Mao Y, Rao Y, Shu Y, Peng B. Microglia facilitate and stabilize the response to general anesthesia via modulating the neuronal network in a brain region-specific manner. eLife 2023; 12:RP92252. [PMID: 38131301 PMCID: PMC10746144 DOI: 10.7554/elife.92252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
General anesthesia leads to a loss of consciousness and an unrousable state in patients. Although general anesthetics are widely used in clinical practice, their underlying mechanisms remain elusive. The potential involvement of nonneuronal cells is unknown. Microglia are important immune cells in the central nervous system (CNS) that play critical roles in CNS function and dysfunction. We unintentionally observed delayed anesthesia induction and early anesthesia emergence in microglia-depleted mice. We found that microglial depletion differentially regulates neuronal activities by suppressing the neuronal network of anesthesia-activated brain regions and activating emergence-activated brain regions. Thus, microglia facilitate and stabilize the anesthesia status. This influence is not mediated by dendritic spine plasticity. Instead, it relies on the activation of microglial P2Y12 and subsequent calcium influx, which facilitates the general anesthesia response. Together, we elucidate the regulatory role of microglia in general anesthesia, extending our knowledge of how nonneuronal cells modulate neuronal activities.
Collapse
Affiliation(s)
- Yang He
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Taohui Liu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Quansheng He
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Wei Ke
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Xiaoyu Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Jinjin Du
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Suixin Deng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Zhenfeng Shu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Jialin Wu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Baozhi Yang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Yuqing Wang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Yanxia Rao
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan UniversityShanghaiChina
| | - Yousheng Shu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- Co-Innovation Center of Neurodegeneration, Nantong UniversityNantongChina
| |
Collapse
|
41
|
Lu D, Choi S, Park J, Kim J, Zhao S, Uldry Lavergne CG, Desimone Q, Chen B, Han BX, Wang F, Goldstein N. General Anesthesia Activates a Central Anxiolytic Center in the BNST. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572586. [PMID: 38187782 PMCID: PMC10769264 DOI: 10.1101/2023.12.20.572586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Low doses of general anesthetics like ketamine and dexmedetomidine have anxiolytic properties independent of their sedative effects. How these different drugs exert these anxiolytic effects is not well understood. We discovered a population of GABAergic neurons in the oval division of the bed nucleus of the stria terminalis that is activated by multiple anesthetics and the anxiolytic drug diazepam (ovBNST GA ). A majority of ovBNST GA neurons express neurotensin receptor 1 (Ntsr1) and innervate brain regions known to regulate anxiety and stress responses. Optogenetic activation ovBNST GA or ovBNST Ntsr1 neurons significantly attenuated anxiety-like behaviors in both naïve animals and mice with inflammatory pain, while inhibition of these cells increased anxiety. Notably, activation of these neurons decreased heart rate and increased heart rate variability, suggesting that they reduce anxiety through modulation of the autonomic nervous system. Our study identifies ovBNST GA /ovBNST Ntsr1 neurons as one of the brain's endogenous anxiolytic centers and a potential therapeutic target for treating anxiety-related disorders. HIGHLIGHTS General anesthetics and anxiolytics activate a population of neurons in the ovBNSTAnesthesia-activated ovBNST neurons bidirectionally modulate anxiety-like behaviorMost anesthesia-activated ovBNST neurons express neurotensin receptor 1 ovBNST Ntsr1 neuron activation shifts autonomic responses to an anxiolytic state.
Collapse
|
42
|
He J, Zhu Y, Wu C, Wu J, Chen Y, Yuan M, Cheng Z, Zeng L, Ji X. Transcranial ultrasound neuromodulation facilitates isoflurane-induced general anesthesia recovery and improves cognition in mice. ULTRASONICS 2023; 135:107132. [PMID: 37604030 DOI: 10.1016/j.ultras.2023.107132] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/13/2023] [Accepted: 08/05/2023] [Indexed: 08/23/2023]
Abstract
Delayed arousal and cognitive dysfunction are common, especially in older patients after general anesthesia (GA). Elevating central nervous system serotonin (5-HT) levels can promote recovery from GA and increase synaptic plasticity to improve cognition. Ultrasound neuromodulation has become a noninvasive physical intervention therapy with high spatial resolution and penetration depth, which can modulate neuronal excitability to treat psychiatric and neurodegenerative diseases. This study aims to use ultrasound to noninvasively modulate the brain 5-HT levels of mice to promote recovery from GA and improve cognition in mice. The dorsal raphe nucleus (DRN) of mice during GA was stimulated by the 1.1 MHz ultrasound with a negative pressure of 356 kPa, and the liquid chromatography coupled tandem mass spectrometry (LC-MS/MS) method was used to measure the DRN 5-HT concentrations. The mice's recovery time from GA was assessed, and the cognition was evaluated through spontaneous alternation Y-maze and novel object recognition (NOR) tests. After ultrasound stimulation, the mice's DRN 5-HT levels were significantly increased (control: 554.0 ± 103.2 ng/g, anesthesia + US: 664.2 ± 84.1 ng/g, *p = 0.0389); the GA recovery time (return of the righting reflex (RORR) emergence latency time) of mice was significantly reduced (anesthesia: 331.6 ± 70 s, anesthesia + US: 223.2 ± 67.7 s, *p = 0.0215); the spontaneous rotation behavior score of mice was significantly increased (anesthesia: 59.46 ± 5.26 %, anesthesia + US: 68.55 ± 5.24 %; *p = 0.0126); the recognition index was significantly increased (anesthesia: 55.02 ± 6.23 %, anesthesia + US: 78.52 ± 12.21 %; ***p = 0.0009). This study indicates that ultrasound stimulation of DRN increases serotonin levels, accelerates recovery from anesthesia, and improves cognition, which could be an important strategy for treating delayed arousal, postoperative delirium, or even lasting cognitive dysfunction after GA.
Collapse
Affiliation(s)
- Jiaru He
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Yiyue Zhu
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Canwen Wu
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Junwei Wu
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Yan Chen
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Maodan Yuan
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhongwen Cheng
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Lvming Zeng
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Xuanrong Ji
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
43
|
Zhang T, Song N, Li S, Yu L, Xie Y, Yue Z, Zhang R, Wang L, Tan H. S-Ketamine Improves Slow Wave Sleep and the Associated Changes in Serum Protein Among Gynecological Abdominal Surgery Patients: A Randomized Controlled Trial. Nat Sci Sleep 2023; 15:903-913. [PMID: 37954026 PMCID: PMC10637210 DOI: 10.2147/nss.s430453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/31/2023] [Indexed: 11/14/2023] Open
Abstract
Purpose This study aims to evaluate the effect of S-ketamine on slow wave sleep (SWS) and the related changes in serum protein in gynecological patients after open abdomen surgery. Methods This was a randomized controlled trial. One hundred gynecological patients undergoing open abdomen surgery were randomized into an S-ketamine group (group S) or placebo group (0.9% saline; group C). During operation, patients in group S received adjuvant S-ketamine infusion (0.2 mg·kg-1·h-1) while those in group C received 0.9% saline. All patients were connected to patient-controlled intravenous analgesia (PCIA) pump in the end of the surgery and the patients in group S with an additional S-ketamine in PCIA pump. Polysomnogram (PSG) was monitored during the next night after surgery with PCIA pump. Blood samples were collected for proteomic analysis at 6:00 AM after PSG monitoring. The primary outcome was the percentage of SWS (also known as stage 3 non-rapid eye movement sleep, stage N3) on the next night after surgery, and the secondary outcome was subjective sleep quality, pain scores, and the changes in serum proteomics. Results Complete polysomnogram recordings were obtained from 64 study participants (31 in group C and 33 in group S). The administration of S-ketamine infusion resulted in a significant increase in the percentage of SWS/N3 compared to the control group (group C, median (IQR [range]), 8.9 (6.3, 12.5); group S, median (IQR [range]), 15.6 (12.4, 18.8), P<0.001). However, subjective evaluations of sleep quality revealed no significant variances between the two groups. The protein affected by S-ketamine was primarily associated with posttranslational modification, protein turnover, carbohydrate transport, and metabolism. Conclusion In patients undergoing open gynecological surgery, S-ketamine enhanced the percentage of objective sleep of SWS during the next night after surgery. Additionally, there were differences observed in serum protein levels between the two groups. Trial Registration ChiCTR2200055180. Registered on 02/01/2022.
Collapse
Affiliation(s)
- Tianzhuo Zhang
- Department of Gastroenterology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, 100045, People’s Republic of China
- Department of Anesthesiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, People’s Republic of China
| | - Nan Song
- Department of Gynecology, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, People’s Republic of China
| | - Shuo Li
- Department of Anesthesiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, People’s Republic of China
| | - Ling Yu
- Department of Anesthesiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, People’s Republic of China
| | - Yining Xie
- Department of Anesthesiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, People’s Republic of China
| | - Zhijie Yue
- Department of Anesthesiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, People’s Republic of China
| | - Rui Zhang
- Philips (China) Investment Co., Ltd., Beijing, 100600, People’s Republic of China
| | - Lijie Wang
- Philips (China) Investment Co., Ltd., Beijing, 100600, People’s Republic of China
| | - Hongyu Tan
- Department of Anesthesiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, People’s Republic of China
| |
Collapse
|
44
|
Bai F, Huang L, Deng J, Long Z, Hao X, Chen P, Wu G, Wen H, Deng Q, Bao X, Huang J, Yang M, Li D, Ren Y, Zhang M, Xiong Y, Li H. Prelimbic area to lateral hypothalamus circuit drives social aggression. iScience 2023; 26:107718. [PMID: 37810230 PMCID: PMC10551839 DOI: 10.1016/j.isci.2023.107718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/06/2023] [Accepted: 08/22/2023] [Indexed: 10/10/2023] Open
Abstract
Controlling aggression is a vital skill in social species such as rodents and humans and has been associated with the medial prefrontal cortex (mPFC). In this study, we showed that during aggressive behavior, the activity of GABAergic neurons in the prelimbic area (PL) of the mPFC was significantly suppressed. Specific activation of GABAergic PL neurons significantly curbed male-to-male aggression and inhibited conditioned place preference (CPP) for aggression-paired contexts, whereas specific inhibition of GABAergic PL neurons brought about the opposite effect. Moreover, GABAergic projections from PL neurons to the lateral hypothalamus (LH) orexinergic neurons mediated aggressive behavior. Finally, directly modulated LH-orexinergic neurons influence aggressive behavior. These results suggest that GABAergic PL-orexinergic LH projection is an important control circuit for intermale aggressive behavior, both of which could be targets for curbing aggression.
Collapse
Affiliation(s)
- Fuhai Bai
- Department of Anesthesiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Lu Huang
- Department of Anesthesiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Jiao Deng
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi 710032, China
| | - Zonghong Long
- Department of Anesthesiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Xianglin Hao
- Department of Pathology, Xinqiao Hospital, Army Medical University, Chongqing 400037, P.R. China
| | - Penghui Chen
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Guangyan Wu
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Huizhong Wen
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Qiangting Deng
- Editorial Office of Journal of Army Medical University, Chongqing 400038, China
| | - Xiaohang Bao
- Department of Anesthesiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Jing Huang
- Department of Anesthesiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Ming Yang
- Department of Anesthesiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Defeng Li
- Clinical Medical Research Center, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Yukun Ren
- Department of Anesthesiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Min Zhang
- Department of Anesthesiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Ying Xiong
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Hong Li
- Department of Anesthesiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| |
Collapse
|
45
|
Vincent KF, Solt K. Modulating anesthetic emergence with pathway-selective dopamine signaling. Curr Opin Anaesthesiol 2023; 36:468-475. [PMID: 37552017 PMCID: PMC10528732 DOI: 10.1097/aco.0000000000001293] [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] [Indexed: 08/09/2023]
Abstract
PURPOSE OF REVIEW To summarize the recent preclinical findings investigating dopaminergic circuits for their involvement in reversing anesthetic-induced unconsciousness. RECENT FINDINGS The release of dopamine from the ventral tegmental area onto dopamine D1 receptor-expressing neurons in the nucleus accumbens promotes emergence following general anesthesia. Two relevant targets of dopamine D1 receptor-expressing neurons in the nucleus accumbens include the lateral hypothalamus and ventral pallidum. Activating mesocortical dopaminergic projections from the ventral tegmental area to the prelimbic cortex has also been shown to hasten emergence from general anesthesia. In contrast, the nigrostriatal dopamine pathway is not involved in regulating anesthetic emergence. The role of the tuberoinfundibular endocrine dopamine pathway remains to be tested; however, recent studies have identified an important function of neuroendocrine signaling on modulating general anesthesia. SUMMARY Potential avenues for accelerating anesthetic emergence may be found through targeting specific arousal-promoting pathways in the brain. Accumulating evidence from rodent studies manipulating cell type- and circuit-specific signaling pathways have identified dopamine as a potent modulator of general anesthesia. Specifically, dopamine signaling along the mesolimbic and mesocortical pathways plays a fundamental role in regulating consciousness.
Collapse
Affiliation(s)
- Kathleen F. Vincent
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Ken Solt
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Yan J, Hang BN, Ma LH, Lin JT, Zhou Y, Jiao XH, Yuan YX, Shao KJ, Zhang LM, Xue Q, Li ZY, Zhang HX, Cao JL, Li S, Zheng H, Wu YQ. GABAergic Neurons in the Nucleus Accumbens are Involved in the General Anesthesia Effect of Propofol. Mol Neurobiol 2023; 60:5789-5804. [PMID: 37349621 DOI: 10.1007/s12035-023-03445-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/03/2023] [Indexed: 06/24/2023]
Abstract
The mechanism underlying the hypnosis effect of propofol is still not fully understood. In essence, the nucleus accumbens (NAc) is crucial for regulating wakefulness and may be directly engaged in the principle of general anesthesia. However, the role of NAc in the process of propofol-induced anesthesia is still unknown. We used immunofluorescence, western blotting, and patch-clamp to access the activities of NAc GABAergic neurons during propofol anesthesia, and then we utilized chemogenetic and optogenetic methods to explore the role of NAc GABAergic neurons in regulating propofol-induced general anesthesia states. Moreover, we also conducted behavioral tests to analyze anesthetic induction and emergence. We found out that c-Fos expression was considerably dropped in NAc GABAergic neurons after propofol injection. Meanwhile, patch-clamp recording of brain slices showed that firing frequency induced by step currents in NAc GABAergic neurons significantly decreased after propofol perfusion. Notably, chemically selective stimulation of NAc GABAergic neurons during propofol anesthesia lowered propofol sensitivity, prolonged the induction of propofol anesthesia, and facilitated recovery; the inhibition of NAc GABAergic neurons exerted opposite effects. Furthermore, optogenetic activation of NAc GABAergic neurons promoted emergence whereas the result of optogenetic inhibition was the opposite. Our results demonstrate that NAc GABAergic neurons modulate propofol anesthesia induction and emergence.
Collapse
Affiliation(s)
- Jing Yan
- Jiangsu Province Key Laboratory of Anesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Bei-Ning Hang
- Jiangsu Province Key Laboratory of Anesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Lin-Hui Ma
- Jiangsu Province Key Laboratory of Anesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Jia-Tao Lin
- Jiangsu Province Key Laboratory of Anesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yue Zhou
- Jiangsu Province Key Laboratory of Anesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, 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, 221004, China
| | - Ying-Xuan Yuan
- Jiangsu Province Key Laboratory of Anesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, 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, 221004, China
| | - Le-Meng Zhang
- Jiangsu Province Key Laboratory of Anesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Qi Xue
- Jiangsu Province Key Laboratory of Anesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, 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, 221004, China
| | - Hong-Xing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Shuai Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - 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, 221004, China.
| |
Collapse
|
47
|
Li Q, Mathena RP, Li F, Dong X, Guan Y, Mintz CD. Effects of Early Exposure to Isoflurane on Susceptibility to Chronic Pain Are Mediated by Increased Neural Activity Due to Actions of the Mammalian Target of the Rapamycin Pathway. Int J Mol Sci 2023; 24:13760. [PMID: 37762067 PMCID: PMC10530853 DOI: 10.3390/ijms241813760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Patients who have undergone surgery in early life may be at elevated risk for suffering neuropathic pain in later life. The risk factors for this susceptibility are not fully understood. Here, we used a mouse chronic pain model to test the hypothesis that early exposure to the general anesthetic (GA) Isoflurane causes cellular and molecular alterations in dorsal spinal cord (DSC) and dorsal root ganglion (DRG) that produces a predisposition to neuropathic pain via an upregulation of the mammalian target of the rapamycin (mTOR) signaling pathway. Mice were exposed to isoflurane at postnatal day 7 (P7) and underwent spared nerve injury at P28 which causes chronic pain. Selected groups were treated with rapamycin, an mTOR inhibitor, for eight weeks. Behavioral tests showed that early isoflurane exposure enhanced susceptibility to chronic pain, and rapamycin treatment improved outcomes. Immunohistochemistry, Western blotting, and q-PCR indicated that isoflurane upregulated mTOR expression and neural activity in DSC and DRG. Accompanying upregulation of mTOR and rapamycin-reversible changes in chronic pain-associated markers, including N-cadherin, cAMP response element-binding protein (CREB), purinergic P2Y12 receptor, glial fibrillary acidic protein (GFAP) in DSC; and connexin 43, phospho-extracellular signal-regulated kinase (p-ERK), GFAP, Iba1 in DRG, were observed. We concluded that early GA exposure, at least with isoflurane, alters the development of pain circuits such that mice are subsequently more vulnerable to chronic neuropathic pain states.
Collapse
Affiliation(s)
- Qun Li
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.P.M.); (F.L.); (Y.G.)
| | - Reilley Paige Mathena
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.P.M.); (F.L.); (Y.G.)
| | - Fengying Li
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.P.M.); (F.L.); (Y.G.)
| | - Xinzhong Dong
- Solomon H. Snyder Department of Neuroscience and Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.P.M.); (F.L.); (Y.G.)
| | - Cyrus David Mintz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.P.M.); (F.L.); (Y.G.)
| |
Collapse
|
48
|
Sun H, Li Z, Qiu Z, Shen Y, Guo Q, Hu SW, Ding HL, An S, Cao JL. A common neuronal ensemble in nucleus accumbens regulates pain-like behaviour and sleep. Nat Commun 2023; 14:4700. [PMID: 37543693 PMCID: PMC10404280 DOI: 10.1038/s41467-023-40450-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 07/28/2023] [Indexed: 08/07/2023] Open
Abstract
A comorbidity of chronic pain is sleep disturbance. Here, we identify a dual-functional ensemble that regulates both pain-like behaviour induced by chronic constrictive injury or complete Freund's adjuvant, and sleep wakefulness, in the nucleus accumbens (NAc) in mice. Specifically, a select population of NAc neurons exhibits increased activity either upon nociceptive stimulation or during wakefulness. Experimental activation of the ensemble neurons exacerbates pain-like (nociceptive) responses and reduces NREM sleep, while inactivation of these neurons produces the opposite effects. Furthermore, NAc ensemble primarily consists of D1 neurons and projects divergently to the ventral tegmental area (VTA) and preoptic area (POA). Silencing an ensemble innervating VTA neurons selectively increases nociceptive responses without affecting sleep, whereas inhibiting ensemble-innervating POA neurons decreases NREM sleep without affecting nociception. These results suggest a common NAc ensemble that encodes chronic pain and controls sleep, and achieves the modality specificity through its divergent downstream circuit targets.
Collapse
Affiliation(s)
- Haiyan Sun
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
- Department of Pediatrics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China
| | - Zhilin Li
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Zhentong Qiu
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yu Shen
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Qingchen Guo
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Su-Wan Hu
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Shuming An
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China.
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China.
| |
Collapse
|
49
|
Bao W, Ding J, Jiang S, Yao Z, Qu W, Li W, Huang Z, Han Y. Selective Activation of NAc D1R-VP/LH Circuits Promotes Reanimation From Sevoflurane Anesthesia in Mice. Anesth Analg 2023; 137:87-97. [PMID: 36944111 DOI: 10.1213/ane.0000000000006436] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
BACKGROUND Emerging evidence has uncovered a vital role of nucleus accumbens (NAc) neurons that express the dopamine D1 receptor (D1R) and its upstream neural circuit in general anesthesia (GA) regulation. However, the underlying downstream neural basis of the modulation of GA emergence by NAc D1R neurons remains unknown. In the present study, we explored the downstream neural mechanism of NAc D1R neurons in the modulation of emergence from sevoflurane GA. METHODS We traced the axonal projections of NAc D1R neurons using a cell type-specific anterograde tracing method and immunohistochemical techniques in D1R-Cre mice. Optogenetic stimulations combined with electroencephalogram/electromyogram recordings and behavioral tests were used to determine the effects of optogenetic activation of the axonal terminals of NAc D1R neurons on sevoflurane emergence during sevoflurane-induced continuous, steady-state general anesthesia (CSSGA) or burst-suppression oscillations. RESULTS Labeled efferent fibers of NAc D1R neurons were highly distributed in the ventral pallidum (VP), lateral hypothalamus (LH), and substantia nigra pars compacta. Optogenetic activation of the NAc D1R -VP circuit during CSSGA with sevoflurane induced cortical activation (mean ± standard deviation [SD]; delta power: prestimulation versus during stimulation, 48.7% ± 5.7% vs 35.1% ± 3.3%, P < .0001; beta power: 7.1% ± 2.7% vs 14.2% ± 3.3%, P = .0264) and behavioral emergence, and restored the righting reflex in 66.7% of ChR2 mice. Optogenetic stimulation of the NAc D1R -LH circuit also produced cortical activation (delta power: prestimulation versus during stimulation, 45.0% ± 6.5% vs 36.1% ± 4.6%, P = .0016) and behavioral emergence, and restored the righting reflex in 100% of the ChR2 mice during CSSGA with sevoflurane. Under a sevoflurane-induced burst-suppression state, NAc D1R -VP/LH circuit activation produced evidence of cortical activation (burst-suppression ratio [BSR]: NAc D1R -VP circuit, prestimulation versus during stimulation, 42.4% ± 4.0% vs 26.3% ± 6.0%, P = .0120; prestimulation versus poststimulation, 42.4% ± 4.0% vs 5.9% ± 5.6%, P = .0002; BSR: NAc D1R -LH circuit, prestimulation versus during stimulation, 33.3% ± 13.4% vs 5.1% ± 4.9%, P = .0177; prestimulation vs poststimulation, 33.3% ± 13.4% vs 3.2% ± 4.0%, P = .0105) and behavioral emergence. CONCLUSIONS Both NAc D1R -VP and NAc D1R -LH circuits are sufficient to promote reanimation from sevoflurane GA by simultaneously inducing cortical and behavioral emergence.
Collapse
Affiliation(s)
- Weiwei Bao
- From the Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiahui Ding
- From the Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Shan Jiang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen Yao
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weimin Qu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenxian Li
- From the Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Zhili Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuan Han
- From the Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, China
| |
Collapse
|
50
|
Bárez-López S, Gadd GJ, Pauža AG, Murphy D, Greenwood MP. Isoflurane Rapidly Modifies Synaptic and Cytoskeletal Phosphoproteomes of the Supraoptic Nucleus of the Hypothalamus and the Cortex. Neuroendocrinology 2023; 113:1008-1023. [PMID: 37271138 DOI: 10.1159/000531352] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/24/2023] [Indexed: 06/06/2023]
Abstract
INTRODUCTION Despite the widespread use of general anaesthetics, the mechanisms mediating their effects are still not understood. Although suppressed in most parts of the brain, neuronal activity, as measured by FOS activation, is increased in the hypothalamic supraoptic nucleus (SON) by numerous general anaesthetics, and evidence points to this brain region being involved in the induction of general anaesthesia (GA) and natural sleep. Posttranslational modifications of proteins, including changes in phosphorylation, enable fast modulation of protein function which could be underlying the rapid effects of GA. In order to identify potential phosphorylation events in the brain-mediating GA effects, we have explored the phosphoproteome responses in the rat SON and compared these to cingulate cortex (CC) which displays no FOS activation in response to general anaesthetics. METHODS Adult Sprague-Dawley rats were treated with isoflurane for 15 min. Proteins from the CC and SON were extracted and processed for nano-LC mass spectrometry (LC-MS/MS). Phosphoproteomic determinations were performed by LC-MS/MS. RESULTS We found many changes in the phosphoproteomes of both the CC and SON in response to 15 min of isoflurane exposure. Pathway analysis indicated that proteins undergoing phosphorylation adaptations are involved in cytoskeleton remodelling and synaptic signalling events. Importantly, changes in protein phosphorylation appeared to be brain region specific suggesting that differential phosphorylation adaptations might underlie the different neuronal activity responses to GA between the CC and SON. CONCLUSION In summary, these data suggest that rapid posttranslational modifications in proteins involved in cytoskeleton remodelling and synaptic signalling events might mediate the central mechanisms mediating GA.
Collapse
Affiliation(s)
- Soledad Bárez-López
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - George J Gadd
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| | - Audrys G Pauža
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
- Translational Cardio-Respiratory Research Group, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| | - Michael P Greenwood
- Molecular Neuroendocrinology Research Group, Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|