1
|
Zhuang Z, Li X, Luo Y, Li Y, Ahmed Isse S, Zhang Z, Luo Q, Chen X. Developmental neurotoxicity of anesthetic etomidate in zebrafish larvae: Alterations in motor function, neurotransmitter signaling, and lipid metabolism. JOURNAL OF HAZARDOUS MATERIALS 2025; 494:138598. [PMID: 40373404 DOI: 10.1016/j.jhazmat.2025.138598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/16/2025] [Accepted: 05/11/2025] [Indexed: 05/17/2025]
Abstract
Etomidate (ETO), a widely used anesthetic, has emerged as a concerning environmental contaminant due to its increasing misuse and demonstrated neurotoxicity in aquatic organisms. This study employed an integrated multi-omics strategy to investigate the developmental neurotoxic effects of ETO in zebrafish (Danio rerio). ETO exposure induced dose-dependent toxicity in zebrafish embryos, characterized by decreased hatching rates (10-20 %), elevated mortality (up to 30 %), and morphological abnormalities such as scoliosis and pericardial edema. Behavioral assays revealed marked locomotor suppression (40-65 % reduction) and disrupted circadian rhythmicity. Neurochemical profiling indicated a 2.1-fold increase in dopamine levels, accompanied by significant reductions in GABAergic (38 %) and serotonergic (42 %) signaling, consistent with transcriptomic downregulation of related pathway genes. Metabolomic analysis revealed dysregulated lipid metabolism, including a 3.2-fold increase in eicosapentaenoic acid (EPA), and perturbations in phenylalanine metabolism. Transgenic zebrafish models (Tg(hb9:eGFP), Tg(coro1a:DsRed), Tg(elavl3:GCaMP6f)) further demonstrated motor neuron damage, inflammatory cell infiltration in the brain, and disrupted Ca2 + dynamics, indicating blood-brain barrier disruption and neuroinflammation responses. Molecular docking analysis confirmed ETO's binding affinity for GABA-A receptors, aligning with observed neurotransmitter imbalances. These findings elucidate ETO's neurotoxic mechanisms, involving neurotransmitter imbalance, metabolic disruption, and neuroinflammatory. The results underscore the dual threat of ETO as both an emerging aquatic pollutant and a developmental neurotoxicant, highlighting the urgent need for stricter environmental monitoring and a reevaluation of its safety profile, particularly during critical developmental windows.
Collapse
Affiliation(s)
- Zile Zhuang
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Xuewei Li
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Yuxuan Luo
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Yihan Li
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Said Ahmed Isse
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Zheng Zhang
- Institute of Forensic Science XiangTan City Public Security Bureau, Xiangtan, PR China
| | - Qizhi Luo
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China.
| | - Xuncai Chen
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
2
|
Li X, Lin X, Zhang Z, Zhuang Z, Li Y, Luo Y, Pan Y, Luo Q, Chen X. Neurotoxicity and aggressive behavior induced by anesthetic etomidate exposure in zebrafish: Insights from multi-omics and machine learning. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 282:107321. [PMID: 40068374 DOI: 10.1016/j.aquatox.2025.107321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 04/05/2025]
Abstract
Etomidate (ETO), widely employed as a surgical anesthetic and more recently recognized as a drug of abuse, has been frequently detected in aquatic environment. However, the toxicity assessment of ETO is insufficient. Adult zebrafish were used to investigate toxicological effects of ETO. Four weeks ETO exposure could induced abnormal behaviors, including reduced anxiety, memory impairment, and heightened aggression. The increased aggression was quantitatively characterized using machine learning, which revealed significantly elevated instantaneous velocity and drastic changes in angular velocity. ETO was predominantly accumulated in the zebrafish brain, where it binds to GABA-A receptors, leading to a significant increase in GABA content. Furthermore, fluorescent staining of reactive oxygen species (ROS) in the brain revealed that ETO exposure significantly increased the oxidative stress level. This oxidative stress resulted in mitochondrial swelling, rupture, and damage to myelinated nerve fibers, ultimately causing cerebral injury in zebrafish. Multi-omics analysis further elucidated that ETO exposure down-regulated the MAPK signaling pathway, hyperactivated motor proteins, and induced metabolic disorders of lipids and amino acids. In summary, this study demonstrates that ETO induces neurotoxicity and behavioral alterations in zebrafish. These findings provide a critical insight into the mechanisms underlying ETO's neurotoxic effects and contribute to a more comprehensive understanding of its environmental and health risks.
Collapse
Affiliation(s)
- Xuewei Li
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xuhui Lin
- The Bartlett School of Sustainable Construction, University College London, London, UK
| | - Zheng Zhang
- Institute of Forensic Science XiangTan City Public Security Bureau, Xiangtan, China
| | - Zile Zhuang
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yihan Li
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yuxuan Luo
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yupeng Pan
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qizhi Luo
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China; Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Xuncai Chen
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China; Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
3
|
Yang X, Zhu S, Xia M, Sun L, Li S, Xiang P, Li F, Deng Q, Chen L, Zhang W, Wang Y, Li Q, Lyu Z, Du X, Du J, Yang Q, Luo Y. The Serotonergic Dorsal Raphe Promotes Emergence from Propofol Anesthesia in Zebrafish. J Neurosci 2025; 45:e2125232025. [PMID: 39947921 PMCID: PMC11984078 DOI: 10.1523/jneurosci.2125-23.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/30/2024] [Accepted: 01/27/2025] [Indexed: 04/11/2025] Open
Abstract
The mechanisms through which general anesthetics induce loss of consciousness remain unclear. Previous studies have suggested that dorsal raphe nucleus serotonergic (DRN5-HT) neurons are involved in inhalational anesthesia, but the underlying neuronal and synaptic mechanisms are not well understood. In this study, we investigated the role of DRN5-HT neurons in propofol-induced anesthesia in larval zebrafish (sex undetermined at this developmental stage) using a combination of in vivo single-cell calcium imaging, two-photon laser ablation, optogenetic activation, in vivo glutamate imaging, and in vivo whole-cell recording. We found that calcium activity of DRN5-HT neurons reversibly decreased during propofol perfusion. Ablation of DRN5-HT neurons prolonged emergence from 30 µM propofol anesthesia, while induction times were not affected under concentrations of 1, 3, and 30 µM. Additionally, optogenetic activation of DRN5-HT neurons strongly promoted emergence from propofol anesthesia. Propofol application to DRN5-HT neurons suppressed both spontaneous and current injection-evoked spike firing, abolished spontaneous excitatory postsynaptic currents, and decreased membrane input resistance. Presynaptic glutamate release events in DRN5-HT neurons were also abolished by propofol. Furthermore, the hyperpolarization of DRN5-HT neurons caused by propofol was abolished by picrotoxin, a GABAA receptor antagonist, which shortened emergence time from propofol anesthesia when locally applied to the DRN. Our results reveal that DRN5-HT neurons in zebrafish are involved in the emergence from propofol anesthesia by inhibiting presynaptic excitatory glutamate inputs and inducing GABAA receptor-mediated hyperpolarization.
Collapse
Affiliation(s)
- Xiaoxuan Yang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shan Zhu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Miaoyun Xia
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Le Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sha Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Peishan Xiang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Funing Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiusui Deng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lijun Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ying Wang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qiang Li
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhuochen Lyu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xufei Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qianzi Yang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
4
|
Zhou K, Hou ZJ, Jiang XL, Xiao YJ, Zhang LC, Xu W, Xiong B, Qu WM, Huang YG, Huang ZL, Wang L. Striatal neurones expressing D1 dopamine receptors modulate consciousness in sevoflurane but not propofol anaesthesia in mice. Br J Anaesth 2025; 134:1105-1121. [PMID: 39915158 PMCID: PMC11947605 DOI: 10.1016/j.bja.2024.10.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 09/09/2024] [Accepted: 10/02/2024] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Sevoflurane and propofol are the most widely used inhaled and i.v. general anaesthetics, respectively. The mechanisms by which sevoflurane and propofol induce loss of consciousness (LOC) remain unclear. Recent studies implicate the brain dopaminergic circuit in anaesthetic-induced LOC and the cortical-striatal-thalamic-cortical loop in decoding consciousness. We investigated the contribution of the dorsal striatum, which is a critical interface between the dopaminergic circuit and the cortical-striatal-thalamic-cortical loop, in sevoflurane and propofol anaesthesia. METHODS Electroencephalography and electromyography recordings and righting reflex tests were used to determine LOC and recovery of consciousness (ROC). The activity of D1 dopamine receptor (D1R)-expressing neurones in the dorsal striatum was monitored using fibre photometry, and regulated using optogenetic and chemogenetic methods in D1R-Cre mice. RESULTS Population activities of striatal D1R neurones began to decrease before LOC and gradually returned after ROC. During sevoflurane anaesthesia, optogenetic activation of striatal D1R neurones induced ROC at cortical and behavioural levels in steady-state anaesthesia and promoted cortical activation in deep burst suppression anaesthesia. Chemogenetic inhibition of striatal D1R neurones accelerated induction (from 242.0 [46.1] to 194.0 [26.9] s; P=0.010) and delayed emergence (from 93.5 [21.2] to 133.5 [33.9] s; P=0.005), whereas chemogenetic activation of these neurones accelerated emergence (from 107 [23.7] to 81.3 [16.1] s; P=0.011). However, neither optogenetic nor chemogenetic manipulation of striatal D1R neurones had any effects on propofol anaesthesia. CONCLUSIONS Striatal D1R neurones modulate the state of consciousness in sevoflurane anaesthesia, but not in propofol anaesthesia.
Collapse
Affiliation(s)
- Kang Zhou
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, Fudan University, Shanghai, China; Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zi-Jun Hou
- Department of Anesthesiology, Yijishan Hospital, Wannan Medical College, Wuhu, China
| | - Xu-Liang Jiang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Jie Xiao
- Department of Anesthesiology, Central South University, Changsha, Hunan, China
| | - Lin-Chen Zhang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, Fudan University, Shanghai, China
| | - Wei Xu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, Fudan University, Shanghai, China
| | - Bo Xiong
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, Fudan University, Shanghai, China
| | - Yu-Guang Huang
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, Fudan University, Shanghai, China; Department of Anesthesiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Lu Wang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Joint International Research Laboratory of Sleep, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
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
|
6
|
Locskai LF, Gill T, Tan SAW, Burton AH, Alyenbaawi H, Burton EA, Allison WT. A larval zebrafish model of traumatic brain injury: optimizing the dose of neurotrauma for discovery of treatments and aetiology. Biol Open 2025; 14:bio060601. [PMID: 39936823 PMCID: PMC11849975 DOI: 10.1242/bio.060601] [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: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 02/13/2025] Open
Abstract
Traumatic brain injuries (TBI) are diverse with heterogeneous injury pathologies, which creates challenges for the clinical treatment and prevention of secondary pathologies such as post-traumatic epilepsy and subsequent dementias. To develop pharmacological strategies that treat TBI and prevent complications, animal models must capture the spectrum of TBI severity to better understand pathophysiological events that occur during and after injury. To address such issues, we improved upon our recent larval zebrafish TBI paradigm emphasizing titrating to different injury levels. We observed coordination between an increase in injury level and clinically relevant injury phenotypes including post-traumatic seizures (PTS) and tau aggregation. This preclinical TBI model is simple to implement, allows dosing of injury levels to model diverse pathologies, and can be scaled to medium- or high-throughput screening.
Collapse
Affiliation(s)
- Laszlo F. Locskai
- Department of Biological Sciences, University of Alberta, Edmonton AB, T6G 2E9, Canada
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton AB, T6G 2M8, Canada
| | - Taylor Gill
- Department of Biological Sciences, University of Alberta, Edmonton AB, T6G 2E9, Canada
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton AB, T6G 2M8, Canada
| | - Samantha A. W. Tan
- Department of Biological Sciences, University of Alberta, Edmonton AB, T6G 2E9, Canada
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton AB, T6G 2M8, Canada
| | - Alexander H. Burton
- Departments of Chemical and Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hadeel Alyenbaawi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
| | - Edward A. Burton
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Geriatric Research, Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA 15213, USA
| | - W. Ted Allison
- Department of Biological Sciences, University of Alberta, Edmonton AB, T6G 2E9, Canada
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton AB, T6G 2M8, Canada
- Department of Medical Genetics, University of Alberta, Edmonton AB, T6G 2H7, Canada
| |
Collapse
|
7
|
Crowder CM, Forman SA. Systematized Serendipity: Fishing Expeditions for Anesthetic Drugs and Targets. Anesthesiology 2024; 141:997-1006. [PMID: 39240535 PMCID: PMC11461116 DOI: 10.1097/aln.0000000000005153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Most of science involves making observations, forming hypotheses, and testing those hypotheses, to form valid conclusions. However, a distinct, longstanding, and very productive scientific approach does not follow this paradigm; rather, it begins with a screen through a random collection of drugs or genetic variations for a particular effect or phenotype. Subsequently, the identity of the drug or gene is determined, and only then are hypotheses formed and the more standard scientific method employed. This alternative approach is called forward screening and includes methods such as genetic mutant screens, small molecule screens, metabolomics, proteomics, and transcriptomics. This review explains the rational for forward screening approaches and uses examples of screens for mutants with altered anesthetic sensitivities and for novel anesthetics to illustrate the methods and impact of the approach. Forward screening approaches are becoming even more powerful with advances in bioinformatics aided by artificial intelligence.
Collapse
Affiliation(s)
- C. Michael Crowder
- Department of Anesthesiology and Pain Medicine, Department of Genome Sciences, Mitochondrial and Metabolism Center, University of Washington, Seattle, WA 98109
| | - Stuart A. Forman
- Department of Anesthesia Critical Care & Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts and Harvard Medical School, Boston Massachusetts
| |
Collapse
|
8
|
Li L, Rana AN, Li EM, Travis MO, Bruchas MR. Noradrenergic tuning of arousal is coupled to coordinated movements. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599619. [PMID: 38948871 PMCID: PMC11212988 DOI: 10.1101/2024.06.18.599619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Matching arousal level to the motor activity of an animal is important for efficiently allocating cognitive resources and metabolic supply in response to behavioral demands, but how the brain coordinates changes in arousal and wakefulness in response to motor activity remains an unclear phenomenon. We hypothesized that the locus coeruleus (LC), as the primary source of cortical norepinephrine (NE) and promoter of cortical and sympathetic arousal, is well-positioned to mediate movement-arousal coupling. Here, using a combination of physiological recordings, fiber photometry, optogenetics, and behavioral tracking, we show that the LCNE activation is tightly coupled to the return of organized movements during waking from an anesthetized state. Moreover, in an awake animal, movement initiations are coupled to LCNE activation, while movement arrests, to LCNE deactivation. We also report that LCNE activity covaries with the depth of anesthesia and that LCNE photoactivation leads to sympathetic activation, consistent with its role in mediating increased arousal. Together, these studies reveal a more nuanced, modulatory role that LCNE plays in coordinating movement and arousal.
Collapse
Affiliation(s)
- Li Li
- Departments of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
- Seattle Children's Research Institute" Seattle, WA 98101, USA
| | - Akshay N Rana
- Departments of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Esther M Li
- Departments of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
- Department of Psychology, University of Washington, Seattle, WA 98105, USA
| | - Myesa O Travis
- Seattle Children's Research Institute" Seattle, WA 98101, USA
| | - Michael R Bruchas
- Departments of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
9
|
Benoit E, Lyons DG, Rihel J. Noradrenergic tone is not required for neuronal activity-induced rebound sleep in zebrafish. J Comp Physiol B 2024; 194:279-298. [PMID: 37480493 PMCID: PMC11233345 DOI: 10.1007/s00360-023-01504-6] [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: 03/20/2023] [Accepted: 07/03/2023] [Indexed: 07/24/2023]
Abstract
Sleep pressure builds during wakefulness, but the mechanisms underlying this homeostatic process are poorly understood. One zebrafish model suggests that sleep pressure increases as a function of global neuronal activity, such as during sleep deprivation or acute exposure to drugs that induce widespread brain activation. Given that the arousal-promoting noradrenergic system is important for maintaining heightened neuronal activity during wakefulness, we hypothesised that genetic and pharmacological reduction of noradrenergic tone during drug-induced neuronal activation would dampen subsequent rebound sleep in zebrafish larvae. During stimulant drug treatment, dampening noradrenergic tone with the α2-adrenoceptor agonist clonidine unexpectedly enhanced subsequent rebound sleep, whereas enhancing noradrenergic signalling with a cocktail of α1- and β-adrenoceptor agonists did not enhance rebound sleep. Similarly, CRISPR/Cas9-mediated elimination of the dopamine β-hydroxylase (dbh) gene, which encodes an enzyme required for noradrenalin synthesis, enhanced baseline sleep in larvae but did not prevent additional rebound sleep following acute induction of neuronal activity. Across all drug conditions, c-fos expression immediately after drug exposure correlated strongly with the amount of induced rebound sleep, but was inversely related to the strength of noradrenergic modulatory tone. These results are consistent with a model in which increases in neuronal activity, as reflected by brain-wide levels of c-fos induction, drive a sleep pressure signal that promotes rebound sleep independently of noradrenergic tone.
Collapse
Affiliation(s)
- Eleanor Benoit
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Declan G Lyons
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
10
|
Gao H, Wang J, Zhang R, Luo T. Recent advances in neural mechanism of general anesthesia induced unconsciousness: insights from optogenetics and chemogenetics. Front Pharmacol 2024; 15:1360864. [PMID: 38655183 PMCID: PMC11035785 DOI: 10.3389/fphar.2024.1360864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 03/20/2024] [Indexed: 04/26/2024] Open
Abstract
For over 170 years, general anesthesia has played a crucial role in clinical practice, yet a comprehensive understanding of the neural mechanisms underlying the induction of unconsciousness by general anesthetics remains elusive. Ongoing research into these mechanisms primarily centers around the brain nuclei and neural circuits associated with sleep-wake. In this context, two sophisticated methodologies, optogenetics and chemogenetics, have emerged as vital tools for recording and modulating the activity of specific neuronal populations or circuits within distinct brain regions. Recent advancements have successfully employed these techniques to investigate the impact of general anesthesia on various brain nuclei and neural pathways. This paper provides an in-depth examination of the use of optogenetic and chemogenetic methodologies in studying the effects of general anesthesia on specific brain nuclei and pathways. Additionally, it discusses in depth the advantages and limitations of these two methodologies, as well as the issues that must be considered for scientific research applications. By shedding light on these facets, this paper serves as a valuable reference for furthering the accurate exploration of the neural mechanisms underlying general anesthesia. It aids researchers and clinicians in effectively evaluating the applicability of these techniques in advancing scientific research and clinical practice.
Collapse
Affiliation(s)
- Hui Gao
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jingyi Wang
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Rui Zhang
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Tao Luo
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
11
|
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
|
12
|
Zhong T, Lin Y, Zhuge R, Lin Y, Huang B, Zeng R. Reviewing the mechanism of propofol addiction. ALL LIFE 2023. [DOI: 10.1080/26895293.2023.2174708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Affiliation(s)
- Tianhao Zhong
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Yuyan Lin
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Ruohuai Zhuge
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Yujie Lin
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Bingwu Huang
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, People’s Republic of China
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Ruifeng Zeng
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, People’s Republic of China
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, People’s Republic of China
| |
Collapse
|
13
|
Bao WW, Jiang S, Qu WM, Li WX, Miao CH, Huang ZL. Understanding the Neural Mechanisms of General Anesthesia from Interaction with Sleep-Wake State: A Decade of Discovery. Pharmacol Rev 2023; 75:532-553. [PMID: 36627210 DOI: 10.1124/pharmrev.122.000717] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/10/2022] [Accepted: 11/16/2022] [Indexed: 01/11/2023] Open
Abstract
The development of cutting-edge techniques to study specific brain regions and neural circuits that regulate sleep-wake brain states and general anesthesia (GA), has increased our understanding of these states that exhibit similar neurophysiologic traits. This review summarizes current knowledge focusing on cell subtypes and neural circuits that control wakefulness, rapid eye movement (REM) sleep, non-REM sleep, and GA. We also review novel insights into their interactions and raise unresolved questions and challenges in this field. Comparisons of the overlapping neural substrates of sleep-wake and GA regulation will help us to understand sleep-wake transitions and how anesthetics cause reversible loss of consciousness. SIGNIFICANCE STATEMENT: General anesthesia (GA), sharing numerous neurophysiologic traits with the process of natural sleep, is administered to millions of surgical patients annually. In the past decade, studies exploring the neural mechanisms underlying sleep-wake and GA have advanced our understanding of their interactions and how anesthetics cause reversible loss of consciousness. Pharmacotherapies targeting the neural substrates associated with sleep-wake and GA regulations have significance for clinical practice in GA and sleep medicine.
Collapse
Affiliation(s)
- Wei-Wei Bao
- Department of Anesthesiology, Zhongshan Hospital; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College (W.W.B., C.H.M., Z.L.H.); Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College (W.W.B., S.J., W.M.Q., Z.L.H.), and Department of Anesthesiology, Eye and Ear, Nose and Throat Hospital (W.X.L.), Fudan University, Shanghai, China
| | - Shan Jiang
- Department of Anesthesiology, Zhongshan Hospital; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College (W.W.B., C.H.M., Z.L.H.); Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College (W.W.B., S.J., W.M.Q., Z.L.H.), and Department of Anesthesiology, Eye and Ear, Nose and Throat Hospital (W.X.L.), Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Anesthesiology, Zhongshan Hospital; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College (W.W.B., C.H.M., Z.L.H.); Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College (W.W.B., S.J., W.M.Q., Z.L.H.), and Department of Anesthesiology, Eye and Ear, Nose and Throat Hospital (W.X.L.), Fudan University, Shanghai, China
| | - Wen-Xian Li
- Department of Anesthesiology, Zhongshan Hospital; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College (W.W.B., C.H.M., Z.L.H.); Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College (W.W.B., S.J., W.M.Q., Z.L.H.), and Department of Anesthesiology, Eye and Ear, Nose and Throat Hospital (W.X.L.), Fudan University, Shanghai, China
| | - Chang-Hong Miao
- Department of Anesthesiology, Zhongshan Hospital; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College (W.W.B., C.H.M., Z.L.H.); Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College (W.W.B., S.J., W.M.Q., Z.L.H.), and Department of Anesthesiology, Eye and Ear, Nose and Throat Hospital (W.X.L.), Fudan University, Shanghai, China
| | - Zhi-Li Huang
- Department of Anesthesiology, Zhongshan Hospital; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College (W.W.B., C.H.M., Z.L.H.); Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College (W.W.B., S.J., W.M.Q., Z.L.H.), and Department of Anesthesiology, Eye and Ear, Nose and Throat Hospital (W.X.L.), Fudan University, Shanghai, China
| |
Collapse
|
14
|
Lawn T, Martins D, O'Daly O, Williams S, Howard M, Dipasquale O. The effects of propofol anaesthesia on molecular-enriched networks during resting-state and naturalistic listening. Neuroimage 2023; 271:120018. [PMID: 36935083 PMCID: PMC10410200 DOI: 10.1016/j.neuroimage.2023.120018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/09/2023] [Indexed: 03/19/2023] Open
Abstract
Placing a patient in a state of anaesthesia is crucial for modern surgical practice. However, the mechanisms by which anaesthetic drugs, such as propofol, impart their effects on consciousness remain poorly understood. Propofol potentiates GABAergic transmission, which purportedly has direct actions on cortex as well as indirect actions via ascending neuromodulatory systems. Functional imaging studies to date have been limited in their ability to unravel how these effects on neurotransmission impact the system-level dynamics of the brain. Here, we leveraged advances in multi-modal imaging, Receptor-Enriched Analysis of functional Connectivity by Targets (REACT), to investigate how different levels of propofol-induced sedation alter neurotransmission-related functional connectivity (FC), both at rest and when individuals are exposed to naturalistic auditory stimulation. Propofol increased GABA-A- and noradrenaline transporter-enriched FC within occipital and somatosensory regions respectively. Additionally, during auditory stimulation, the network related to the dopamine transporter showed reduced FC within bilateral regions of temporal and mid/posterior cingulate cortices, with the right temporal cluster showing an interaction between auditory stimulation and level of consciousness. In bringing together these micro- and macro-scale systems, we provide support for both direct GABAergic and indirect noradrenergic and dopaminergic-related network changes under propofol sedation. Further, we delineate a cognition-related reconfiguration of the dopaminergic network, highlighting the utility of REACT to explore the molecular substrates of consciousness and cognition.
Collapse
Affiliation(s)
- Timothy Lawn
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's college London, London, UK.
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's college London, London, UK
| | - Owen O'Daly
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's college London, London, UK
| | - Steve Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's college London, London, UK
| | - Matthew Howard
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's college London, London, UK
| | - Ottavia Dipasquale
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's college London, London, UK
| |
Collapse
|
15
|
Yin J, Qin J, Lin Z, Li A, Liu D, Jiang Y, Zhao Q, Chen L, Liu C. Glutamatergic neurons in the paraventricular hypothalamic nucleus regulate isoflurane anesthesia in mice. FASEB J 2023; 37:e22762. [PMID: 36719765 DOI: 10.1096/fj.202200974rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 12/11/2022] [Accepted: 12/27/2022] [Indexed: 02/01/2023]
Abstract
The glutamatergic-mediated excitatory system in the brain is vital for the regulation of sleep-wake and general anesthesia. Specifically, the paraventricular hypothalamic nucleus (PVH), which contains mainly glutamatergic neurons, has been shown to play a critical role in sleep-wake. Here, we sought to explore whether the PVH glutamatergic neurons have an important effect on the process of general anesthesia. We used c-fos staining and in vivo calcium signal recording to observe the activity changes of the PVH glutamatergic neurons during isoflurane anesthesia and found that both c-fos expression in the PVH and the calcium activity of PVH glutamatergic neurons decreased in isoflurane anesthesia and significantly increased during the recovery process. Chemogenetic activation of PVH glutamatergic neurons prolonged induction time and shortened emergence time from anesthesia by decreasing the depth of anesthesia. Using chemogenetic inhibition of PVH glutamatergic neurons under isoflurane anesthesia, we found that inhibition of PVH glutamatergic neurons facilitated the induction process and delayed the emergence accompanied by deepening the depth of anesthesia. Together, these results identify a crucial role for PVH glutamatergic neurons in modulating isoflurane anesthesia.
Collapse
Affiliation(s)
- Jianyin Yin
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital (Hunan Institute of Reproductive Medicine), Changsha, China
| | - Jie Qin
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China.,Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zhaojing Lin
- Department of Anesthesiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Aiyuan Li
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital (Hunan Institute of Reproductive Medicine), Changsha, China
| | - Damin Liu
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital (Hunan Institute of Reproductive Medicine), Changsha, China
| | - Yurong Jiang
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital (Hunan Institute of Reproductive Medicine), Changsha, China
| | - Qiuni Zhao
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital (Hunan Institute of Reproductive Medicine), Changsha, China
| | - Liang Chen
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital (Hunan Institute of Reproductive Medicine), Changsha, China
| | - Chengxi Liu
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, China.,Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
16
|
Zhai X, Yuan Y, Xu L, Jun J, Li Y, Yan Y, Zhang L. Cerebrospinal fluid contacting nucleus and its 5-HT: A new insight into the regulation mechanism of general intravenous anesthesia. Brain Res 2022; 1798:148168. [DOI: 10.1016/j.brainres.2022.148168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
|
17
|
Jung J, Kim T. General anesthesia and sleep: like and unlike. Anesth Pain Med (Seoul) 2022; 17:343-351. [DOI: 10.17085/apm.22227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
General anesthesia and sleep have long been discussed in the neurobiological context owingto their commonalities, such as unconsciousness, immobility, non-responsiveness to externalstimuli, and lack of memory upon returning to consciousness. Sleep is regulated bycomplex interactions between wake-promoting and sleep-promoting neural circuits. Anestheticsexert their effects partly by inhibiting wake-promoting neurons or activating sleep-promotingneurons. Unconscious but arousable sedation is more related to sleep-wake circuitries,whereas unconscious and unarousable anesthesia is independent of them. Generalanesthesia is notable for its ability to decrease sleep propensity. Conversely, increasedsleep propensity due to insufficient sleep potentiates anesthetic effects. Taken together, it isplausible that sleep and anesthesia are closely related phenomena but not the same ones.Further investigations on the relationship between sleep and anesthesia are warranted.
Collapse
|
18
|
Wagle M, Zarei M, Lovett-Barron M, Poston KT, Xu J, Ramey V, Pollard KS, Prober DA, Schulkin J, Deisseroth K, Guo S. Brain-wide perception of the emotional valence of light is regulated by distinct hypothalamic neurons. Mol Psychiatry 2022; 27:3777-3793. [PMID: 35484242 PMCID: PMC9613822 DOI: 10.1038/s41380-022-01567-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/25/2022] [Accepted: 04/06/2022] [Indexed: 02/08/2023]
Abstract
Salient sensory stimuli are perceived by the brain, which guides both the timing and outcome of behaviors in a context-dependent manner. Light is such a stimulus, which is used in treating mood disorders often associated with a dysregulated hypothalamic-pituitary-adrenal stress axis. Relationships between the emotional valence of light and the hypothalamus, and how they interact to exert brain-wide impacts remain unclear. Employing larval zebrafish with analogous hypothalamic systems to mammals, we show in free-swimming animals that hypothalamic corticotropin releasing factor (CRFHy) neurons promote dark avoidance, and such role is not shared by other hypothalamic peptidergic neurons. Single-neuron projection analyses uncover processes extended by individual CRFHy neurons to multiple targets including sensorimotor and decision-making areas. In vivo calcium imaging uncovers a complex and heterogeneous response of individual CRFHy neurons to the light or dark stimulus, with a reduced overall sum of CRF neuronal activity in the presence of light. Brain-wide calcium imaging under alternating light/dark stimuli further identifies distinct and distributed photic response neuronal types. CRFHy neuronal ablation increases an overall representation of light in the brain and broadly enhances the functional connectivity associated with an exploratory brain state. These findings delineate brain-wide photic perception, uncover a previously unknown role of CRFHy neurons in regulating the perception and emotional valence of light, and suggest that light therapy may alleviate mood disorders through reducing an overall sum of CRF neuronal activity.
Collapse
Affiliation(s)
- Mahendra Wagle
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-2811, USA
| | - Mahdi Zarei
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-2811, USA
| | - Matthew Lovett-Barron
- Department of Bioengineering, Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Kristina Tyler Poston
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-2811, USA
| | - Jin Xu
- Tianqiao and Chrissy Chen Institute for Neuroscience, Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Vince Ramey
- Biophysics Graduate Group, University of California, Berkeley, CA, USA
- Invitae Inc., San Francisco, CA, USA
| | - Katherine S Pollard
- Gladstone Institute of Data Science & Biotechnology, San Francisco, CA, USA
- Department of Epidemiology & Biostatistics, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - David A Prober
- Tianqiao and Chrissy Chen Institute for Neuroscience, Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Jay Schulkin
- Department of Obstetrics & Gynecology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Karl Deisseroth
- Department of Bioengineering, Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Su Guo
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-2811, USA.
- Programs in Human Genetics and Biological Sciences, Kavli Institute of Fundamental Neuroscience, The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Bakar Aging Research Institute, University of California, San Francisco, CA, 94143-2811, USA.
| |
Collapse
|
19
|
Zhang K, Pan J, Yu Y. Regulation of Neural Circuitry under General Anesthesia: New Methods and Findings. Biomolecules 2022; 12:biom12070898. [PMID: 35883456 PMCID: PMC9312763 DOI: 10.3390/biom12070898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 02/01/2023] Open
Abstract
General anesthesia has been widely utilized since the 1840s, but its underlying neural circuits remain to be completely understood. Since both general anesthesia and sleep are reversible losses of consciousness, studies on the neural-circuit mechanisms affected by general anesthesia have mainly focused on the neural nuclei or the pathways known to regulate sleep. Three advanced technologies commonly used in neuroscience, in vivo calcium imaging, chemogenetics, and optogenetics, are used to record and modulate the activity of specific neurons or neural circuits in the brain areas of interest. Recently, they have successfully been used to study the neural nuclei and pathways of general anesthesia. This article reviews these three techniques and their applications in the brain nuclei or pathways affected by general anesthesia, to serve as a reference for further and more accurate exploration of other neural circuits under general anesthesia and to contribute to other research fields in the future.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; (K.Z.); (J.P.)
- Tianjin Institute of Anesthesiology, Tianjin 300052, China
| | - Jiacheng Pan
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; (K.Z.); (J.P.)
- Tianjin Institute of Anesthesiology, Tianjin 300052, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; (K.Z.); (J.P.)
- Tianjin Institute of Anesthesiology, Tianjin 300052, China
- Correspondence:
| |
Collapse
|
20
|
Heshmati M, Bruchas MR. Historical and Modern Evidence for the Role of Reward Circuitry in Emergence. Anesthesiology 2022; 136:997-1014. [PMID: 35362070 PMCID: PMC9467375 DOI: 10.1097/aln.0000000000004148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Increasing evidence supports a role for brain reward circuitry in modulating arousal along with emergence from anesthesia. Emergence remains an important frontier for investigation, since no drug exists in clinical practice to initiate rapid and smooth emergence. This review discusses clinical and preclinical evidence indicating a role for two brain regions classically considered integral components of the mesolimbic brain reward circuitry, the ventral tegmental area and the nucleus accumbens, in emergence from propofol and volatile anesthesia. Then there is a description of modern systems neuroscience approaches to neural circuit investigations that will help span the large gap between preclinical and clinical investigation with the shared aim of developing therapies to promote rapid emergence without agitation or delirium. This article proposes that neuroscientists include models of whole-brain network activity in future studies to inform the translational value of preclinical investigations and foster productive dialogues with clinician anesthesiologists.
Collapse
Affiliation(s)
- Mitra Heshmati
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, and Department of Biological Structure, University of Washington, Seattle, Washington
| | - Michael R Bruchas
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, and Department of Pharmacology, University of Washington, Seattle, Washington
| |
Collapse
|
21
|
Locus Coeruleus in Non-Mammalian Vertebrates. Brain Sci 2022; 12:brainsci12020134. [PMID: 35203898 PMCID: PMC8870555 DOI: 10.3390/brainsci12020134] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/08/2022] [Accepted: 01/15/2022] [Indexed: 11/30/2022] Open
Abstract
The locus coeruleus (LC) is a vertebrate-specific nucleus and the primary source of norepinephrine (NE) in the brain. This nucleus has conserved properties across species: highly homogeneous cell types, a small number of cells but extensive axonal projections, and potent influence on brain states. Comparative studies on LC benefit greatly from its homogeneity in cell types and modularity in projection patterns, and thoroughly understanding the LC-NE system could shed new light on the organization principles of other more complex modulatory systems. Although studies on LC are mainly focused on mammals, many of the fundamental properties and functions of LC are readily observable in other vertebrate models and could inform mammalian studies. Here, we summarize anatomical and functional studies of LC in non-mammalian vertebrate classes, fish, amphibians, reptiles, and birds, on topics including axonal projections, gene expressions, homeostatic control, and modulation of sensorimotor transformation. Thus, this review complements mammalian studies on the role of LC in the brain.
Collapse
|
22
|
Yang Q, Zhou F, Li A, Dong H. Neural Substrates for Regulation of Sleep and General Anesthesia. Curr Neuropharmacol 2021; 20:72-84. [PMID: 34906058 PMCID: PMC9199549 DOI: 10.2174/1570159x19666211214144639] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/09/2021] [Accepted: 12/10/2021] [Indexed: 11/30/2022] Open
Abstract
General anesthesia has been successfully used in clinics for over 170 years, but its mechanisms of effect remain unclear. Behaviorally, general anesthesia is similar to sleep as it produces a reversible transition between wakefulness and the state of being unaware of one’s surroundings. A discussion regarding the common circuits of sleep and general anesthesia has been ongoing as an increasing number of sleep-arousal regulatory nuclei are reported to participate in the consciousness shift occurring during general anesthesia. Recently, with progress in research technology, both positive and negative evidence for overlapping neural circuits between sleep and general anesthesia has emerged. This article provides a review of the latest evidence on the neural substrates for sleep and general anesthesia regulation by comparing the roles of pivotal nuclei in sleep and anesthesia.
Collapse
Affiliation(s)
- Qianzi Yang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an. China
| | - Fang Zhou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an. China
| | - Ao Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an. China
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an. China
| |
Collapse
|
23
|
Wang Y, Xu L, Liu MZ, Hu DD, Fang F, Xu DJ, Zhang R, Hua XX, Li JB, Zhang L, Huang LN, Mu D. Norepinephrine modulates wakefulness via α1 adrenoceptors in paraventricular thalamic nucleus. iScience 2021; 24:103015. [PMID: 34522858 PMCID: PMC8426266 DOI: 10.1016/j.isci.2021.103015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/10/2021] [Accepted: 08/18/2021] [Indexed: 11/26/2022] Open
Abstract
Norepinephrine (NE) neurons in the locus coeruleus (LC) play key roles in modulating sleep and wakefulness. Recent studies have revealed that the paraventricular thalamic nucleus (PVT) is a critical wakefulness-controlling nucleus in mice. However, the effects of NE on PVT neurons remain largely unknown. Here, we investigated the mechanisms of NE modulating wakefulness in the PVT by using viral tracing, behavioral tests, slice electrophysiology, and optogenetics techniques. We found that the PVT-projecting LC neurons had few collateral projections to other brain nuclei. Behavioral tests showed that specific activation of the LC-PVT projections or microinjection of NE into the PVT accelerated emergence from general anesthesia and enhanced locomotion activity. Moreover, brain slice recording results indicated that NE increased the activity of the PVT neurons mainly by increasing the frequency of spontaneous excitatory postsynaptic currents via α1 adrenoceptors. Thus, our results demonstrate that NE modulates wakefulness via α1 adrenoceptors in the PVT.
Collapse
Affiliation(s)
- Yan Wang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 Xin Song Jiang Road, Shanghai 201620, China
| | - Ling Xu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 Xin Song Jiang Road, Shanghai 201620, China
| | - Ming-Zhe Liu
- Department of Respiratory, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Dan-Dan Hu
- Tongji University School of Medicine, Shanghai, China
| | - Fang Fang
- Department of Endocrinology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Dao-Jie Xu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Rui Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 Xin Song Jiang Road, Shanghai 201620, China
| | - Xiao-Xiao Hua
- Tongji University School of Medicine, Shanghai, China
| | - Jin-Bao Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 Xin Song Jiang Road, Shanghai 201620, China
| | - Ling Zhang
- Tongji University School of Medicine, Shanghai, China
| | - Li-Na Huang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 Xin Song Jiang Road, Shanghai 201620, China
| | - Di Mu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 Xin Song Jiang Road, Shanghai 201620, China
| |
Collapse
|
24
|
Könemann S, Meyer S, Betz A, Županič A, Vom Berg C. Sub-Lethal Peak Exposure to Insecticides Triggers Olfaction-Mediated Avoidance in Zebrafish Larvae. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:11835-11847. [PMID: 34398619 DOI: 10.1021/acs.est.1c01792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In agricultural areas, insecticides inevitably reach water bodies via leaching or run-off. While designed to be neurotoxic to insects, insecticides have adverse effects on a multitude of organisms due to the high conservation of the nervous system among phyla. To estimate the ecological effects of insecticides, it is important to investigate their impact on non-target organisms such as fish. Using zebrafish as the model, we investigated how different classes of insecticides influence fish behavior and uncovered neuronal underpinnings of the associated behavioral changes, providing an unprecedented insight into the perception of these chemicals by fish. We observed that zebrafish larvae avoid diazinon and imidacloprid while showing no response to other insecticides with the same mode of action. Moreover, ablation of olfaction abolished the aversive responses, indicating that fish smelled the insecticides. Assessment of neuronal activity in 289 brain regions showed that hypothalamic areas involved in stress response were among the regions with the largest changes, indicating that the observed behavioral response resembles reactions to stimuli that threaten homeostasis, such as changes in water chemistry. Our results contribute to the understanding of the environmental impact of insecticide exposure and can help refine acute toxicity assessment.
Collapse
Affiliation(s)
- Sarah Könemann
- Department of Environmental Toxicology, Eawag, Überlandstrasse 133, 8600 Dübendorf, Switzerland
- École Polytechnique Fédérale de Lausanne, EPFL, Route Cantonale, 1015 Lausanne, Switzerland
| | - Stéphanie Meyer
- École Polytechnique Fédérale de Lausanne, EPFL, Route Cantonale, 1015 Lausanne, Switzerland
| | - Alexander Betz
- Department of Environmental Toxicology, Eawag, Überlandstrasse 133, 8600 Dübendorf, Switzerland
| | - Anže Županič
- Department of Environmental Toxicology, Eawag, Überlandstrasse 133, 8600 Dübendorf, Switzerland
| | - Colette Vom Berg
- Department of Environmental Toxicology, Eawag, Überlandstrasse 133, 8600 Dübendorf, Switzerland
| |
Collapse
|
25
|
Zhang Y, Gui H, Duan Z, Yu T, Zhang J, Liang X, Liu C. Dopamine D1 Receptor in the Nucleus Accumbens Modulates the Emergence from Propofol Anesthesia in Rat. Neurochem Res 2021; 46:1435-1446. [PMID: 33683630 DOI: 10.1007/s11064-021-03284-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 01/26/2021] [Accepted: 02/23/2021] [Indexed: 01/04/2023]
Abstract
It has been reported that systemic activation of D1 receptors promotes emergence from isoflurane-induced unconsciousness, suggesting that the central dopaminergic system is involved in the process of recovering from general anesthesia. The nucleus accumbens (NAc) contains abundant GABAergic medium spiny neurons (MSNs) expressing the D1 receptor (D1R), which plays a key role in sleep-wake behavior. However, the role of NAc D1 receptors in the process of emergence from general anesthesia has not been identified. Here, using real-time in vivo fiber photometry, we found that neuronal activity in the NAc was markedly disinhibited during recovery from propofol anesthesia. Subsequently, microinjection of a D1R selective agonist (chloro-APB hydrobromide) into the NAc notably reduced the time to emerge from propofol anesthesia with a decrease in δ-band power and an increase in β-band power evident in the cortical electroencephalogram. These effects were prevented by pretreatment with a D1R antagonist (SCH-23390). Whole-cell patch clamp recordings were performed to further explore the cellular mechanism underlying the modulation of D1 receptors on MSNs under propofol anesthesia. Our data primarily demonstrated that propofol increased the frequency and prolonged the decay time of spontaneous inhibitory postsynaptic currents (sIPSCs) and miniature IPSCs (mIPSCs) of MSNs expressing D1 receptors. A D1R agonist attenuated the effect of propofol on the frequency of sIPSCs and mIPSCs, and the effects of the agonist were eliminated by preapplication of SCH-23390. Collectively, these results indicate that modulation of the D1 receptor on the activity of NAc MSNs is vital for emergence from propofol-induced unconsciousness.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Huan Gui
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zikun Duan
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tian Yu
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jie Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaoli Liang
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chengxi Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China.
- Guizhou Key Laboratory of Brain Science, Guizhou Key Laboratory of Anesthesia and Organ Protection, The Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
26
|
Liu PF, Wang Y, Zhang R, Xu L, Li JB, Mu D. Propofol modulates inhibitory inputs in paraventricular thalamic nucleus of mice. Neurosci Lett 2021; 756:135950. [PMID: 33979698 DOI: 10.1016/j.neulet.2021.135950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/11/2021] [Accepted: 05/07/2021] [Indexed: 11/19/2022]
Abstract
The mechanisms of general anaesthetics such as propofol have drawn substantial attention. The effects of propofol on inhibitory postsynaptic currents are not exactly the same in different brain nuclei. Recent studies revealed that the paraventricular thalamic nucleus (PVT) is a critical nucleus modulating wakefulness. However, the effects of propofol on PVT neurons and the mechanisms underlying such effects remain unknown. Here, we performed the whole-cell recording of the PVT neurons in acute brain slices and bath application of propofol. We found that propofol hyperpolarized the membrane potentials of the PVT neurons and suppressed the action potentials induced by step-current injection. Propofol did not affect the spontaneous inhibitory postsynaptic currents (sIPSCs) amplitude or frequency, but prolonged the sIPSCs half-width. Besides, propofol increased miniature inhibitory synaptic currents (mIPSCs) frequency and half-width. Furthermore, propofol could induce GABAA receptors-mediated tonic inhibitory currents dose-dependently. Thus, our results demonstrate that propofol hyperpolarizes PVT neurons by modulating inhibitory currents via GABAA receptors in mice.
Collapse
Affiliation(s)
- Peng-Fei Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Wang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Xu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin-Bao Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Di Mu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
27
|
Wei Y, Zhang D, Liu J, Ou M, Liang P, Zuo Y, Zhou C. Effects of sevoflurane anesthesia and abdominal surgery on the systemic metabolome: a prospective observational study. BMC Anesthesiol 2021; 21:80. [PMID: 33731015 PMCID: PMC7968205 DOI: 10.1186/s12871-021-01301-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 03/08/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Metabolic status can be impacted by general anesthesia and surgery. However, the exact effects of general anesthesia and surgery on systemic metabolome remain unclear, which might contribute to postoperative outcomes. METHODS Five hundred patients who underwent abdominal surgery were included. General anesthesia was mainly maintained with sevoflurane. The end-tidal sevoflurane concentration (ETsevo) was adjusted to maintain BIS (Bispectral index) value between 40 and 60. The mean ETsevo from 20 min after endotracheal intubation to 2 h after the beginning of surgery was calculated for each patient. The patients were further divided into low ETsevo group (mean - SD) and high ETsevo group (mean + SD) to investigate the possible metabolic changes relevant to the amount of sevoflurane exposure. RESULTS The mean ETsevo of the 500 patients was 1.60% ± 0.34%. Patients with low ETsevo (n = 55) and high ETsevo (n = 59) were selected for metabolomic analysis (1.06% ± 0.13% vs. 2.17% ± 0.16%, P < 0.001). Sevoflurane and abdominal surgery disturbed the tricarboxylic acid cycle as identified by increased citrate and cis-aconitate levels and impacted glycometabolism as identified by increased sucrose and D-glucose levels in these 114 patients. Glutamate metabolism was also impacted by sevoflurane and abdominal surgery in all the patients. In the patients with high ETsevo, levels of L-glutamine, pyroglutamic acid, sphinganine and L-selenocysteine after sevoflurane anesthesia and abdominal surgery were significantly higher than those of the patients with low ETsevo, suggesting that these metabolic changes might be relevant to the amount of sevoflurane exposure. CONCLUSIONS Sevoflurane anesthesia and abdominal surgery can impact principal metabolic pathways in clinical patients including tricarboxylic acid cycle, glycometabolism and glutamate metabolism. This study may provide a resource data for future studies about metabolism relevant to general anaesthesia and surgeries. TRIAL REGISTRATION www.chictr.org.cn . identifier: ChiCTR1800014327 .
Collapse
Affiliation(s)
- Yiyong Wei
- Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, 37# Guoxue Xiang, Chengdu, 610041, Sichuan, China
- Department of Anesthesiology, West China Hospital of Sichuan University, 37# Guoxue Xiang, Chengdu, 610041, Sichuan, China
| | - Donghang Zhang
- Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, 37# Guoxue Xiang, Chengdu, 610041, Sichuan, China
- Department of Anesthesiology, West China Hospital of Sichuan University, 37# Guoxue Xiang, Chengdu, 610041, Sichuan, China
| | - Jin Liu
- Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, 37# Guoxue Xiang, Chengdu, 610041, Sichuan, China.
- Department of Anesthesiology, West China Hospital of Sichuan University, 37# Guoxue Xiang, Chengdu, 610041, Sichuan, China.
| | - Mengchan Ou
- Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, 37# Guoxue Xiang, Chengdu, 610041, Sichuan, China
- Department of Anesthesiology, West China Hospital of Sichuan University, 37# Guoxue Xiang, Chengdu, 610041, Sichuan, China
| | - Peng Liang
- Department of Anesthesiology, West China Hospital of Sichuan University, 37# Guoxue Xiang, Chengdu, 610041, Sichuan, China
| | - Yunxia Zuo
- Department of Anesthesiology, West China Hospital of Sichuan University, 37# Guoxue Xiang, Chengdu, 610041, Sichuan, China.
| | - Cheng Zhou
- Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, 37# Guoxue Xiang, Chengdu, 610041, Sichuan, China
| |
Collapse
|
28
|
Venincasa MJ, Randlett O, Sumathipala SH, Bindernagel R, Stark MJ, Yan Q, Sloan SA, Buglo E, Meng QC, Engert F, Züchner S, Kelz MB, Syed S, Dallman JE. Elevated preoptic brain activity in zebrafish glial glycine transporter mutants is linked to lethargy-like behaviors and delayed emergence from anesthesia. Sci Rep 2021; 11:3148. [PMID: 33542258 PMCID: PMC7862283 DOI: 10.1038/s41598-021-82342-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 01/19/2021] [Indexed: 11/17/2022] Open
Abstract
Delayed emergence from anesthesia was previously reported in a case study of a child with Glycine Encephalopathy. To investigate the neural basis of this delayed emergence, we developed a zebrafish glial glycine transporter (glyt1 - / -) mutant model. We compared locomotor behaviors; dose-response curves for tricaine, ketamine, and 2,6-diisopropylphenol (propofol); time to emergence from these anesthetics; and time to emergence from propofol after craniotomy in glyt1-/- mutants and their siblings. To identify differentially active brain regions in glyt1-/- mutants, we used pERK immunohistochemistry as a proxy for brain-wide neuronal activity. We show that glyt1-/- mutants initiated normal bouts of movement less frequently indicating lethargy-like behaviors. Despite similar anesthesia dose-response curves, glyt1-/- mutants took over twice as long as their siblings to emerge from ketamine or propofol, mimicking findings from the human case study. Reducing glycine levels rescued timely emergence in glyt1-/- mutants, pointing to a causal role for elevated glycine. Brain-wide pERK staining showed elevated activity in hypnotic brain regions in glyt1-/- mutants under baseline conditions and a delay in sensorimotor integration during emergence from anesthesia. Our study links elevated activity in preoptic brain regions and reduced sensorimotor integration to lethargy-like behaviors and delayed emergence from propofol in glyt1-/- mutants.
Collapse
Affiliation(s)
- Michael J Venincasa
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Owen Randlett
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, 69008, Lyon, France
| | - Sureni H Sumathipala
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Richard Bindernagel
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Matthew J Stark
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Qing Yan
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Steven A Sloan
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Elena Buglo
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, 33101, USA
- Dr. John T. MacDonald Foundation Department of Human Genetics, University of Miami, Miami, FL, 33136, USA
| | - Qing Cheng Meng
- Departments of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Florian Engert
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Stephan Züchner
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, 33101, USA
- Dr. John T. MacDonald Foundation Department of Human Genetics, University of Miami, Miami, FL, 33136, USA
| | - Max B Kelz
- Departments of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sheyum Syed
- Department of Physics, University of Miami, Coral Gables, FL, 33146, USA
| | - Julia E Dallman
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA.
| |
Collapse
|
29
|
Cao S, Fisher DW, Rodriguez G, Yu T, Dong H. Comparisons of neuroinflammation, microglial activation, and degeneration of the locus coeruleus-norepinephrine system in APP/PS1 and aging mice. J Neuroinflammation 2021; 18:10. [PMID: 33407625 PMCID: PMC7789762 DOI: 10.1186/s12974-020-02054-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/09/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The role of microglia in Alzheimer's disease (AD) pathogenesis is becoming increasingly important, as activation of these cell types likely contributes to both pathological and protective processes associated with all phases of the disease. During early AD pathogenesis, one of the first areas of degeneration is the locus coeruleus (LC), which provides broad innervation of the central nervous system and facilitates norepinephrine (NE) transmission. Though the LC-NE is likely to influence microglial dynamics, it is unclear how these systems change with AD compared to otherwise healthy aging. METHODS In this study, we evaluated the dynamic changes of neuroinflammation and neurodegeneration in the LC-NE system in the brain and spinal cord of APP/PS1 mice and aged WT mice using immunofluorescence and ELISA. RESULTS Our results demonstrated increased expression of inflammatory cytokines and microglial activation observed in the cortex, hippocampus, and spinal cord of APP/PS1 compared to WT mice. LC-NE neuron and fiber loss as well as reduced norepinephrine transporter (NET) expression was more evident in APP/PS1 mice, although NE levels were similar between 12-month-old APP/PS1 and WT mice. Notably, the degree of microglial activation, LC-NE nerve fiber loss, and NET reduction in the brain and spinal cord were more severe in 12-month-old APP/PS1 compared to 12- and 24-month-old WT mice. CONCLUSION These results suggest that elevated neuroinflammation and microglial activation in the brain and spinal cord of APP/PS1 mice correlate with significant degeneration of the LC-NE system.
Collapse
Affiliation(s)
- Song Cao
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, China
- Guizhou Key Lab of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, 6 West Xuefu Street, Zunyi, 563002, Guizhou, China
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL, 60611, USA
| | - Daniel W Fisher
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL, 60611, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington Medical Center, 1959 NE Pacific St, Seattle, WA, 98195, USA
| | - Guadalupe Rodriguez
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL, 60611, USA
| | - Tian Yu
- Guizhou Key Lab of Anesthesia and Organ Protection, Affiliated Hospital of Zunyi Medical University, 6 West Xuefu Street, Zunyi, 563002, Guizhou, China
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL, 60611, USA.
| |
Collapse
|
30
|
Zhao G, Han H, Wang W, Jia K. Propofol rather than Isoflurane Accelerates the Interstitial Fluid Drainage in the Deep Rat Brain. Int J Med Sci 2021; 18:652-659. [PMID: 33437200 PMCID: PMC7797541 DOI: 10.7150/ijms.54320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 11/23/2020] [Indexed: 11/15/2022] Open
Abstract
Objective: Different anesthetics have distinct effects on the interstitial fluid (ISF) drainage in the extracellular space (ECS) of the superficial rat brain, while their effects on ISF drainage in the ECS of the deep rat brain still remain unknown. Herein, we attempt to investigate and compare the effects of propofol and isoflurane on ECS structure and ISF drainage in the caudate-putamen (CPu) and thalamus (Tha) of the deep rat brain. Methods: Adult Sprague-Dawley rats were anesthetized with propofol or isoflurane, respectively. Twenty-four anesthetized rats were randomly divided into the propofol-CPu, isoflurane-CPu, propofol-Tha, and isoflurane-Tha groups. Tracer-based magnetic resonance imaging (MRI) and fluorescent-labeled tracer assay were utilized to quantify ISF drainage in the deep brain. Results: The half-life of ISF in the propofol-CPu and propofol-Tha groups was shorter than that in the isoflurane-CPu and isoflurane-Tha groups, respectively. The ECS volume fraction in the propofol-CPu and propofol-Tha groups was much higher than that in the isoflurane-CPu and isoflurane-Tha groups, respectively. However, the ECS tortuosity in the propofol-CPu and propofol-Tha groups was much smaller than that in isoflurane-CPu and isoflurane-Tha groups, respectively. Conclusions: Our results demonstrate that propofol rather than isoflurane accelerates the ISF drainage in the deep rat brain, which provides novel insights into the selective control of ISF drainage and guides selection of anesthetic agents in different clinical settings, and unravels the mechanism of how general anesthetics function.
Collapse
Affiliation(s)
- Guomei Zhao
- Department of Geriatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Magnetic Resonance Imaging Technology, Beijing 100191, China.,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Wei Wang
- Research Institute for Translation Medicine on Molecular Function and Artificial Intelligence Imaging, Department of Radiology, The First People's Hospital of FoShan, Foshan 52800, China
| | - Kaiying Jia
- Department of Geriatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| |
Collapse
|
31
|
Kuner R, Kuner T. Cellular Circuits in the Brain and Their Modulation in Acute and Chronic Pain. Physiol Rev 2020; 101:213-258. [PMID: 32525759 DOI: 10.1152/physrev.00040.2019] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic, pathological pain remains a global health problem and a challenge to basic and clinical sciences. A major obstacle to preventing, treating, or reverting chronic pain has been that the nature of neural circuits underlying the diverse components of the complex, multidimensional experience of pain is not well understood. Moreover, chronic pain involves diverse maladaptive plasticity processes, which have not been decoded mechanistically in terms of involvement of specific circuits and cause-effect relationships. This review aims to discuss recent advances in our understanding of circuit connectivity in the mammalian brain at the level of regional contributions and specific cell types in acute and chronic pain. A major focus is placed on functional dissection of sub-neocortical brain circuits using optogenetics, chemogenetics, and imaging technological tools in rodent models with a view towards decoding sensory, affective, and motivational-cognitive dimensions of pain. The review summarizes recent breakthroughs and insights on structure-function properties in nociceptive circuits and higher order sub-neocortical modulatory circuits involved in aversion, learning, reward, and mood and their modulation by endogenous GABAergic inhibition, noradrenergic, cholinergic, dopaminergic, serotonergic, and peptidergic pathways. The knowledge of neural circuits and their dynamic regulation via functional and structural plasticity will be beneficial towards designing and improving targeted therapies.
Collapse
Affiliation(s)
- Rohini Kuner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; and Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Thomas Kuner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; and Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
32
|
Song SY, Li Y, Zhai XM, Li YH, Bao CY, Shan CJ, Hong J, Cao JL, Zhang LC. Connection Input Mapping and 3D Reconstruction of the Brainstem and Spinal Cord Projections to the CSF-Contacting Nucleus. Front Neural Circuits 2020; 14:11. [PMID: 32296310 PMCID: PMC7136615 DOI: 10.3389/fncir.2020.00011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/10/2020] [Indexed: 01/04/2023] Open
Abstract
Objective To investigate whether the CSF-contacting nucleus receives brainstem and spinal cord projections and to understand the functional significance of these connections. Methods The retrograde tracer cholera toxin B subunit (CB) was injected into the CSF-contacting nucleus in Sprague-Dawley rats according the previously reported stereotaxic coordinates. After 7–10 days, these rats were perfused and their brainstem and spinal cord were sliced (thickness, 40 μm) using a freezing microtome. All the sections were subjected to CB immunofluorescence staining. The distribution of CB-positive neuron in different brainstem and spinal cord areas was observed under fluorescence microscope. Results The retrograde labeled CB-positive neurons were found in the midbrain, pons, medulla oblongata, and spinal cord. Four functional areas including one hundred and twelve sub-regions have projections to the CSF-contacting nucleus. However, the density of CB-positive neuron distribution ranged from sparse to dense. Conclusion Based on the connectivity patterns of the CSF-contacting nucleus receives anatomical inputs from the brainstem and spinal cord, we preliminarily conclude and summarize that the CSF-contacting nucleus participates in pain, visceral activity, sleep and arousal, emotion, and drug addiction. The present study firstly illustrates the broad projections of the CSF-contacting nucleus from the brainstem and spinal cord, which implies the complicated functions of the nucleus especially for the unique roles of coordination in neural and body fluids regulation.
Collapse
Affiliation(s)
- Si-Yuan Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Ying Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Xiao-Meng Zhai
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Yue-Hao Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Cheng-Yi Bao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Cheng-Jing Shan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Jia Hong
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Li-Cai Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
33
|
Drug-selective Anesthetic Insensitivity of Zebrafish Lacking γ-Aminobutyric Acid Type A Receptor β3 Subunits. Anesthesiology 2020; 131:1276-1291. [PMID: 31567362 DOI: 10.1097/aln.0000000000002963] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Transgenic mouse studies suggest that γ-aminobutyric acid type A (GABAA) receptors containing β3 subunits mediate important effects of etomidate, propofol, and pentobarbital. Zebrafish, recently introduced for rapid discovery and characterization of sedative-hypnotics, could also accelerate pharmacogenetic studies if their transgenic phenotypes reflect those of mammals. The authors hypothesized that, relative to wild-type, GABAA-β3 functional knock-out (β3) zebrafish would show anesthetic sensitivity changes similar to those of β3 mice. METHODS Clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 mutagenesis was used to create a β3 zebrafish line. Wild-type and β3 zebrafish were compared for fertility, growth, and craniofacial development. Sedative and hypnotic effects of etomidate, propofol, pentobarbital, alphaxalone, ketamine, tricaine, dexmedetomidine, butanol, and ethanol, along with overall activity and thigmotaxis were quantified in 7-day postfertilization larvae using video motion analysis of up to 96 animals simultaneously. RESULTS Xenopus oocyte electrophysiology showed that the wild-type zebrafish β3 gene encodes ion channels activated by propofol and etomidate, while the β3 zebrafish transgene does not. Compared to wild-type, β3 zebrafish showed similar morphology and growth, but more rapid swimming. Hypnotic EC50s (mean [95% CI]) were significantly higher for β3 versus wild-type larvae with etomidate (1.3 [1.0 to 1.6] vs. 0.6 [0.5 to 0.7] µM; P < 0.0001), propofol (1.1 [1.0 to 1.4] vs. 0.7 [0.6 to 0.8] µM; P = 0.0005), and pentobarbital (220 [190 to 240] vs. 130 [94 to 179] μM; P = 0.0009), but lower with ethanol (150 [106 to 213] vs. 380 [340 to 420] mM; P < 0.0001) and equivalent with other tested drugs. Comparing β3 versus wild-type sedative EC50s revealed a pattern similar to hypnosis. CONCLUSIONS Global β3 zebrafish are selectively insensitive to the same few sedative-hypnotics previously reported in β3 transgenic mice, indicating phylogenetic conservation of β3-containing GABAA receptors as anesthetic targets. Transgenic zebrafish are potentially valuable models for sedative-hypnotic mechanisms research.
Collapse
|
34
|
Fast Localization and Sectioning of Mouse Locus Coeruleus. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4860735. [PMID: 32190666 PMCID: PMC7071797 DOI: 10.1155/2020/4860735] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 02/15/2020] [Indexed: 01/29/2023]
Abstract
The locus nucleus (LC) is a multifunctional nucleus which is also the source of norepinephrine in the brain. To date, there is no simple and easy method to locate the small LC in brain sectioning. Here we report a fast, accurate, and easy-to-follow protocol for the localization of mice LC in frozen sectioning. After fixation and dehydration, the intact brains of adult mice were placed on a horizontal surface and vertically cut along the posterior margin of the bilateral cerebral cortex. In the coronal cutting plane, the aqueduct of midbrain can be seen easily with the naked eyes. After embedding the cerebellum part with optimal cutting temperature (OCT) compound, coronal brain slices were cut from the cutting plane, within 1 mm, the aqueduct of midbrain disappeared and the fourth ventricle appeared, then the brain slices contained LC and were collected. From the first collection, at ~200 μm, the noradrenergic neurons' most enriched brain slices can be collected. The tyrosine hydroxylase immunofluorescence staining confirmed that the localization of LC with this method is accurate and the noradrenergic neuron most abundant slices can be determined with this method.
Collapse
|
35
|
Espino-Saldaña AE, Rodríguez-Ortiz R, Pereida-Jaramillo E, Martínez-Torres A. Modeling Neuronal Diseases in Zebrafish in the Era of CRISPR. Curr Neuropharmacol 2020; 18:136-152. [PMID: 31573887 PMCID: PMC7324878 DOI: 10.2174/1570159x17666191001145550] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/04/2019] [Accepted: 09/29/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Danio rerio is a powerful experimental model for studies in genetics and development. Recently, CRISPR technology has been applied in this species to mimic various human diseases, including those affecting the nervous system. Zebrafish offer multiple experimental advantages: external embryogenesis, rapid development, transparent embryos, short life cycle, and basic neurobiological processes shared with humans. This animal model, together with the CRISPR system, emerging imaging technologies, and novel behavioral approaches, lay the basis for a prominent future in neuropathology and will undoubtedly accelerate our understanding of brain function and its disorders. OBJECTIVE Gather relevant findings from studies that have used CRISPR technologies in zebrafish to explore basic neuronal function and model human diseases. METHODS We systematically reviewed the most recent literature about CRISPR technology applications for understanding brain function and neurological disorders in D. rerio. We highlighted the key role of CRISPR in driving forward our understanding of particular topics in neuroscience. RESULTS We show specific advances in neurobiology when the CRISPR system has been applied in zebrafish and describe how CRISPR is accelerating our understanding of brain organization. CONCLUSION Today, CRISPR is the preferred method to modify genomes of practically any living organism. Despite the rapid development of CRISPR technologies to generate disease models in zebrafish, more efforts are needed to efficiently combine different disciplines to find the etiology and treatments for many brain diseases.
Collapse
Affiliation(s)
- Angeles Edith Espino-Saldaña
- Departamento de Neurobiología Celular y Molecular, Laboratorio de Neurobiología Molecular y Celular, Instituto de Neurobiología, Campus UNAM Juriquilla, Querétaro, Qro CP76230, México
- Universidad Autónoma de Querétaro, Facultad de Ciencias Naturales, Av. de las Ciencias S/N, Querétaro, Mexico
| | - Roberto Rodríguez-Ortiz
- CONACYT - Instituto de Neurobiología, Universidad Nacional Autónoma de México. Querétaro, Qro., México
| | - Elizabeth Pereida-Jaramillo
- Departamento de Neurobiología Celular y Molecular, Laboratorio de Neurobiología Molecular y Celular, Instituto de Neurobiología, Campus UNAM Juriquilla, Querétaro, Qro CP76230, México
| | - Ataúlfo Martínez-Torres
- Departamento de Neurobiología Celular y Molecular, Laboratorio de Neurobiología Molecular y Celular, Instituto de Neurobiología, Campus UNAM Juriquilla, Querétaro, Qro CP76230, México
| |
Collapse
|
36
|
Félix L, Coimbra AM, Valentim AM, Antunes L. Review on the use of zebrafish embryos to study the effects of anesthetics during early development. Crit Rev Toxicol 2019; 49:357-370. [PMID: 31314655 DOI: 10.1080/10408444.2019.1617236] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Over the years, the potential toxicity of anesthetics has raised serious concerns about its safe use during pregnancy. As evidence emerged from research in animal models, showing that some anesthetic drugs are potential teratogenic, the determination of the risk of exposures to anesthetic drugs at early life stages became mandatory. However, due to inaccessibility and ethical constrains related to experimental conditions, the use of early life stages in mammalian models is limited. In this regard, some animal and nonanimal models have been suggested to surpass mammalian use in experimentation. Among them, the zebrafish embryo test has been recognized as a promising alternative in toxicology research, as well as an inexpensive and practical test. Substantial information collected from developmental research following compounds exposure, has contributed to the application of zebrafish assays in research, although only a few studies have focused on the use of early life stages of zebrafish to evaluate the developmental effects of anesthetics. Based on the recent advances of science and technology, there is a clear potential for zebrafish early life stages to provide new insights into anesthetics teratogenicity. This review provides an overview of recent anesthesia research using zebrafish embryos, demonstrating its usefulness to the anesthesia field, discussing the recent findings on various aspects related to the effects of anesthetics during early life development and the strengths and limitations of this model system.
Collapse
Affiliation(s)
- Luís Félix
- Institute for Research and Innovation in Health, Laboratory Animal Science, Institute of Molecular and Cell Biology, University of Porto , Porto , Portugal.,Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro , Vila Real , Portugal
| | - Ana Maria Coimbra
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro , Vila Real , Portugal
| | - Ana Maria Valentim
- Institute for Research and Innovation in Health, Laboratory Animal Science, Institute of Molecular and Cell Biology, University of Porto , Porto , Portugal.,Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro , Vila Real , Portugal
| | - Luís Antunes
- Institute for Research and Innovation in Health, Laboratory Animal Science, Institute of Molecular and Cell Biology, University of Porto , Porto , Portugal.,Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes and Alto Douro , Vila Real , Portugal
| |
Collapse
|