1
|
Chen H, Liu C, Liu J, Yuan C, He H, Zhang Y, Yu S, Luo T, Shen W, Yu T. Zona Incerta GABAergic Neurons Facilitate Emergence from Isoflurane Anesthesia in Mice. Neurochem Res 2024; 49:3297-3307. [PMID: 39312079 PMCID: PMC11502554 DOI: 10.1007/s11064-024-04230-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 10/25/2024]
Abstract
The zona incerta (ZI) predominantly consists of gamma-aminobutyric acid (GABAergic) neurons, located adjacent to the lateral hypothalamus. GABA, acting on GABAA receptors, serves as a crucial neuromodulator in the initiation and maintenance of general anesthesia. In this study, we aimed to investigate the involvement of ZI GABAergic neurons in the general anesthesia process. Utilizing in-vivo calcium signal optical fiber recording, we observed a decrease in the activity of ZI GABAergic neurons during isoflurane anesthesia, followed by a significant increase during the recovery phase. Subsequently, we selectively ablated ZI GABAergic neurons to explore their role in general anesthesia, revealing no impact on the induction of isoflurane anesthesia but a prolonged recovery time, accompanied by a reduction in delta-band power in mice under isoflurane anesthesia. Finally, through optogenetic activation/inhibition of ZI GABAergic neurons during isoflurane anesthesia, we discovered that activation of these neurons facilitated emergence without affecting the induction process, while inhibition delayed emergence, leading to fluctuations in delta band activity. In summary, these findings highlight the involvement of ZI GABAergic neurons in modulating the emergence of isoflurane anesthesia.
Collapse
Affiliation(s)
- Hong Chen
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Department of Anesthesiology, KweiChow Moutai Hospital, Renhuai, Guizhou, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Chengxi Liu
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Junxiao Liu
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Department of Anesthesiology, Zunyi Hospital of Traditional Chinese Medicine, Zunyi, Guizhou, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Chengdong Yuan
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Department of Anesthesiology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Haifeng He
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Yu Zhang
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
| | - Shouyang Yu
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Tianyuan Luo
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China
- Guizhou Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, China
| | - Wei Shen
- School of Life Science and Technology and Shanghai Institute of Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Tian Yu
- Key Laboratory of Anesthesia and Organ Protestion of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China.
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
2
|
Li J, Wei Y, Xiang J, Zhang D. Role of the ventral tegmental area in general anesthesia. Eur J Pharmacol 2024:177145. [PMID: 39566814 DOI: 10.1016/j.ejphar.2024.177145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/22/2024]
Abstract
The ventral tegmental area (VTA), located in the midbrain, plays a pivotal role in the regulation of many important behaviors, such as reward, addiction, aversion, memory, learning, and sleep-wakefulness cycles. The majority of VTA neurons are dopaminergic neurons, although there is a significant proportion of GABAergic neurons and few glutamatergic neurons. These neuronal types project to different brain regions, thus mediating various biological functions. Therefore, the diverse roles of the VTA might depend on its heterogeneous neuronal types and projecting circuits. General anesthesia and sleep-wakefulness cycles share the feature of reversible loss of consciousness, and several common neural mechanisms underlie these two conditions. In addition to the well-known regulatory role of VTA in sleep-wakefulness, emerging evidence has demonstrated that VTA activity is also associated with promoting emergence from general anesthesia. Herein, we reviewed the literature and summarized the evidence regarding the modulation of the VTA by general anesthesia in rodents, which will improve the understanding of the modulatory mechanism of the VTA in general anesthesia.
Collapse
Affiliation(s)
- Jia Li
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430048, China.
| | - Yiyong Wei
- Department of Anesthesiology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518100, China
| | - Jiaxin Xiang
- Department of Anesthesiology, Weill Cornell Medicine, New York, 10065, USA
| | - Donghang Zhang
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430048, China; Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
3
|
Sharma R, Parikh M, Chischolm A, Kempuraj D, Thakkar M. Dopamine D2 receptors in the accumbal core region mediates the effects of fentanyl on sleep-wakefulness. Neuroscience 2024; 560:11-19. [PMID: 39276843 DOI: 10.1016/j.neuroscience.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024]
Abstract
Fentanyl, a potent analgesic and addictive substance, significantly impacts sleep-wakefulness (S-W). Acutely, it promotes wake, whereas chronic abuse leads to severe sleep disruptions, including insomnia, which contributes to opioid use disorders (OUD), a chronic brain disease characterized by compulsive opioid use and harmful consequences. Although the critical association between sleep disruptions and fentanyl addiction is acknowledged, the precise mechanisms through which fentanyl influences sleep remain elusive. Recent studies highlight the role of the dopaminergic system of the nucleus accumbens (NAc) in S-W regulation, but its specific involvement in mediating fentanyl's effects on S-W remains unexplored. We hypothesized that dopamine D2 receptors mediate fentanyl-induced effects on S-W. To test this hypothesis, male C57BL/6J mice, instrumented with sleep recording electrodes and bilateral guide cannulas above the accumbal core region (NAcC), were utilized in this study. At dark onset, animals were bilaterally administered sulpiride (D2 receptors antagonist; 250 ng/side) in the NAcC followed by an intraperitoneal injection of fentanyl (1.2 mg/Kg). S-W was examined for the next 12 h. We found that systemic administration of fentanyl significantly increased wakefulness during the first 6 h of the dark which was followed by a significant increase in NREM and REM sleep during the second 6 h of the dark period. D2-receptor blockade significantly reduced this effect as evidenced by a significant reduction in fentanyl-induced wakefulness during first 6 h of dark period and sleep rebound during the second 6 h. Our findings suggest that D2 receptors in the NAcC plays a vital role in mediating the fentanyl-induced changes in S-W.
Collapse
Affiliation(s)
- Rishi Sharma
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO 65201, United States.
| | - Meet Parikh
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO 65201, United States
| | - Abigail Chischolm
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO 65201, United States
| | - Deepak Kempuraj
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO 65201, United States
| | - Mahesh Thakkar
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO 65201, United States
| |
Collapse
|
4
|
Jiang S, Chen L, Qu WM, Huang ZL, Chen CR. Hypothalamic corticotropin-releasing hormone neurons modulate sevoflurane anesthesia and the post-anesthesia stress responses. eLife 2024; 12:RP90191. [PMID: 39526880 PMCID: PMC11554309 DOI: 10.7554/elife.90191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
General anesthesia (GA) is an indispensable procedure necessary for safely and compassionately administering a significant number of surgical procedures and invasive diagnostic tests. However, the undesired stress response associated with GA causes delayed recovery and even increased morbidity in the clinic. Here, a core hypothalamic ensemble, corticotropin-releasing hormone neurons in the paraventricular nucleus of the hypothalamus (PVHCRH neurons), is discovered to play a role in regulating sevoflurane GA. Chemogenetic activation of these neurons delay the induction of and accelerated emergence from sevoflurane GA, whereas chemogenetic inhibition of PVHCRH neurons accelerates induction and delays awakening. Moreover, optogenetic stimulation of PVHCRH neurons induce rapid cortical activation during both the steady and deep sevoflurane GA state with burst-suppression oscillations. Interestingly, chemogenetic inhibition of PVHCRH neurons relieve the sevoflurane GA-elicited stress response (e.g., excessive self-grooming and elevated corticosterone level). These findings identify PVHCRH neurons modulate states of anesthesia in sevoflurane GA, being a part of anesthesia regulatory network of sevoflurane.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Lu Chen
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| | - Chang-Rui Chen
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan UniversityShanghaiChina
| |
Collapse
|
5
|
Zhang Y, Li J, Li Y, Wang W, Wang D, Ding J, Wang L, Cheng J. Dexmedetomidine Promotes NREM Sleep by Depressing Oxytocin Neurons in the Paraventricular Nucleus in Mice. Neurochem Res 2024; 49:2926-2939. [PMID: 39078522 DOI: 10.1007/s11064-024-04221-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Dexmedetomidine (DEX) is a highly selective α2-adrenoceptor agonist with sedative effects on sleep homeostasis. Oxytocin-expressing (OXT) neurons in the paraventricular nucleus (PVN) of the hypothalamus (PVNOXT) regulate sexual reproduction, drinking, sleep-wakefulness, and other instinctive behaviors. To investigate the effect of DEX on the activity and signal transmission of PVNOXT in regulating the sleep-wakefulness cycle. Here, we employed OXT-cre mice to selectively target and express the designer receptors exclusively activated by designer drugs (DREADD)-based chemogenetic tool hM3D(Gq) in PVNOXT neurons. Combining chemogenetic methods with electroencephalogram (EEG) /electromyogram (EMG) recordings, we found that cannula injection of DEX in PVN significantly increased the duration of non-rapid eye movement (NREM) sleep in mice. Furthermore, the chemogenetic activation of PVNOXT neurons using i.p. injection of clozapine N-oxide (CNO) after cannula injection of DEX to PVN led to a substantial increase in wakefulness. Electrophysiological results showed that DEX decreased the frequency of action potential (AP) and the spontaneous excitatory postsynaptic current (sEPSC) of PVNOXT neurons through α2-adrenoceptors. Therefore, these results identify that DEX promotes sleep and maintains sleep homeostasis by inhibiting PVNOXT neurons through the α2-adrenoceptor.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Jiaxin Li
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yan Li
- Department of Pharmacy, Linquan People's Hospital, Linquan, 236400, Anhui, China
| | - Wei Wang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Daming Wang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Junli Ding
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Licheng Wang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China.
- College of Stomatology, Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Juan Cheng
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
6
|
Pardo-Valencia J, Moreno-Gomez M, Mercado N, Pro B, Ammann C, Humanes-Valera D, Foffani G. Local wakefulness-like activity of layer 5 cortex under general anaesthesia. J Physiol 2024; 602:5289-5307. [PMID: 39316039 DOI: 10.1113/jp286417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 08/08/2024] [Indexed: 09/25/2024] Open
Abstract
Consciousness, defined as being aware of and responsive to one's surroundings, is characteristic of normal waking life and typically is lost during sleep and general anaesthesia. The traditional view of consciousness as a global brain state has evolved toward a more sophisticated interplay between global and local states, with the presence of local sleep in the awake brain and local wakefulness in the sleeping brain. However, this interplay is not clear for general anaesthesia, where loss of consciousness was recently suggested to be associated with a global state of brain-wide synchrony that selectively involves layer 5 cortical pyramidal neurons across sensory, motor and associative areas. According to this global view, local wakefulness of layer 5 cortex should be incompatible with deep anaesthesia, a hypothesis that deserves to be scrutinised with causal manipulations. Here, we show that unilateral chemogenetic activation of layer 5 pyramidal neurons in the sensorimotor cortex of isoflurane-anaesthetised mice induces a local state transition from slow-wave activity to tonic firing in the transfected hemisphere. This wakefulness-like activity dramatically disrupts layer 5 interhemispheric synchrony with mirror-image locations in the contralateral hemisphere, but does not reduce the level of unconsciousness under deep anaesthesia, nor in the transitions to/from anaesthesia. Global layer 5 synchrony may thus be a sufficient condition for anaesthesia-induced unconsciousness, but is not a necessary one, at least under isoflurane anaesthesia. Local wakefulness-like activity of layer 5 cortex can be induced and maintained under deep anaesthesia, encouraging further investigation into the local vs. global aspects of anaesthesia-induced unconsciousness. KEY POINTS: The neural correlates of consciousness have evolved from global brain states to a nuanced interplay between global and local states, evident in terms of local sleep in awake brains and local wakefulness in sleeping brains. The concept of local wakefulness remains unclear for general anaesthesia, where the loss of consciousness has been recently suggested to involve brain-wide synchrony of layer 5 cortical neurons. We found that local wakefulness-like activity of layer 5 cortical can be chemogenetically induced in anaesthetised mice without affecting the depth of anaesthesia or the transitions to and from unconsciousness. Global layer 5 synchrony may thus be a sufficient but not necessary feature for the unconsciousness induced by general anaesthesia. Local wakefulness-like activity of layer 5 neurons is compatible with general anaesthesia, thus promoting further investigation into the local vs. global aspects of anaesthesia-induced unconsciousness.
Collapse
Affiliation(s)
- Jesús Pardo-Valencia
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Escuela Técnica Superior de Ingenieros de Telecomunicación, Universidad Politécnica de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
| | - Miryam Moreno-Gomez
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
- PhD Program in Neuroscience, Universidad Autonoma de Madrid-Cajal Institute, Madrid, Spain
| | - Noelia Mercado
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
| | - Beatriz Pro
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
| | - Claudia Ammann
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain
| | - Desire Humanes-Valera
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
| | - Guglielmo Foffani
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Spain
- Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
7
|
Feng CH, Du XN, Wang Z, Wu T, Zhang LN. The activity of cholinergic neurons in the basal forebrain interferes with anesthesia-arousal process of propofol. Neuropeptides 2024; 107:102449. [PMID: 38908356 DOI: 10.1016/j.npep.2024.102449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
Previous research has demonstrated that basal forebrain (BF) regulates arousal during propofol anesthesia. However, as the BF comprises cholinergic neurons alongside two other types of neurons, the specific role of cholinergic neurons has not been definitively elucidated. In our study, calcium signal imaging was utilized to monitor the real-time activities of cholinergic neurons in the BF during propofol anesthesia. Additionally, we selectively stimulated these neurons to investigate EEG and behavioral responses during propofol anesthesia. Furthermore, we specifically lesioned cholinergic neurons in the BF to investigate the sensitivity to propofol and the induction time. The results revealed that propofol suppressed calcium signals of cholinergic neurons within the BF following intraperitoneal injection. Notably, upon recovery of the righting reflex, the calcium signals partially recovered. Spectral analysis of the EEG elucidated that optical stimulation of cholinergic neurons led to a decrease in δ power underlie propofol anesthesia. Conversely, depletion of cholinergic neurons in the BF enhanced sensitivity to propofol and shortened the induction time. These findings clarify the role of cholinergic neurons in the anesthesia-arousal process, as well as the depth and the sensitivity of propofol anesthesia.
Collapse
Affiliation(s)
- Cai-Hua Feng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Xiao-Nan Du
- Department of Anesthesiology, Central Hospital of Wuhan Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430014, China
| | - Zhi Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Ting Wu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Li-Na Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
8
|
Fettiplace MR, Vincent KF, Cho A, Dillon E, Stapley BM, Stewart V, Solt K. Dopaminergic psychostimulants cause arousal from isoflurane-induced sedation without reversing memory impairment in rats. Br J Anaesth 2024; 133:793-803. [PMID: 38965013 PMCID: PMC11443133 DOI: 10.1016/j.bja.2024.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/30/2024] [Accepted: 05/04/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Dopaminergic psychostimulants can restore arousal in anaesthetised animals, and dopaminergic signalling contributes to hippocampal-dependent memory formation. We tested the hypothesis that dopaminergic psychostimulants can antagonise the amnestic effects of isoflurane on visuospatial working memory. METHODS Sixteen adult Sprague-Dawley rats were trained on a trial-unique nonmatching-to-location (TUNL) task which assessed the ability to identify a novel touchscreen location after a fixed delay. Once trained, the effects of low-dose isoflurane (0.3 vol%) on task performance and activity, assessed by infrared beam breaks, were assessed. We attempted to rescue deficits in performance and activity with a dopamine D1 receptor agonist (chloro-APB), a noradrenergic reuptake inhibitor (atomoxetine), and a mixed dopamine/norepinephrine releasing agent (dextroamphetamine). Anaesthetic induction, emergence, and recovery from anaesthesia were also investigated. RESULTS Low-dose isoflurane impaired working memory in a sex-independent and intra-trial delay-independent manner as assessed by task performance, and caused an overall reduction in activity. Administration of chloro-APB, atomoxetine, or dextroamphetamine did not restore visuospatial working memory, but chloro-APB and dextroamphetamine recovered arousal to levels observed in the baseline awake state. Performance did not differ between induction and emergence. Animals recovered to baseline performance within 15 min of discontinuing isoflurane. CONCLUSIONS Low-dose isoflurane impairs visuospatial working memory in a nondurable and delay-independent manner that potentially implicates non-hippocampal structures in isoflurane-induced memory deficits. Dopaminergic psychostimulants counteracted sedation but did not reverse memory impairments, suggesting that isoflurane-induced amnesia and isoflurane-induced sedation have distinct underlying mechanisms that can be antagonised independently.
Collapse
Affiliation(s)
- Michael R Fettiplace
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA.
| | - Kathleen F Vincent
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Angel Cho
- Touro College of Osteopathic Medicine, New York, NY, USA
| | - Emmaline Dillon
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, USA
| | - Brendan M Stapley
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, USA
| | - Victoria Stewart
- University of California Irvine School of Medicine, Irvine, CA, USA
| | - Ken Solt
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Galindo-Charles L, Reyes-Legorreta C, Garduño J, Galarraga E, Tapia D, Hernández-López S. The activation of D2-like dopamine receptors increases NMDA currents in the dorsal raphe serotonergic neurons. Neurosci Lett 2024; 839:137933. [PMID: 39128818 DOI: 10.1016/j.neulet.2024.137933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The dorsal raphe nucleus (DRN) receives dopaminergic inputs from the ventral tegmental area (VTA). Also, the DRN contains a small population of cells that express dopamine (DRNDA neurons). However, the physiological role of dopamine (DA) in the DRN and its interaction with serotonergic (5-HT) neurons is poorly understood. Several works have reported moderate levels of D1, D2, and D3 DA receptors in the DRN. Furthermore, it was found that the activation of D2 receptors increased the firing of putative 5-HT neurons. Other studies have reported that D1 and D2 dopamine receptors can interact with glutamate NMDA receptors, modulating the excitability of different cell types. In the present work, we used immunocytochemical techniques to determine the kind of DA receptors in the DRN. Additionally, we performed electrophysiological experiments in brainstem slices to study the effect of DA agonists on NMDA-elicited currents recorded from identified 5-HT DRN neurons. We found that D2 and D3 but not D1 receptors are present in this nucleus. Also, we demonstrated that the activation of D2-like receptors increases NMDA-elicited currents in 5-HT neurons through a mechanism involving phospholipase C (PLC) and protein kinase C (PKC) enzymes. Possible physiological implications related to the sleep-wake cycle are discussed.
Collapse
Affiliation(s)
- L Galindo-Charles
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - C Reyes-Legorreta
- Laboratorio de Neuroprotección, Instituto Nacional de Rehabilitación-LGII, Ciudad de México 14389, Mexico
| | - J Garduño
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - E Galarraga
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - D Tapia
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - S Hernández-López
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico.
| |
Collapse
|
10
|
Wang D, Bao C, Wu H, Li J, Zhang X, Wang S, Zhou F, Li H, Dong H. A hypothalamus-lateral periaqueductal gray GABAergic neural projection facilitates arousal following sevoflurane anesthesia in mice. CNS Neurosci Ther 2024; 30:e70047. [PMID: 39317457 PMCID: PMC11421888 DOI: 10.1111/cns.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/01/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND The lateral hypothalamus (LHA) is an evolutionarily conserved structure that regulates basic functions of an organism, particularly wakefulness. To clarify the function of LHAGABA neurons and their projections on regulating general anesthesia is crucial for understanding the excitatory and inhibitory effects of anesthetics on the brain. The aim of the present study is to investigate whether LHAGABA neurons play either an inhibitory or a facilitatory role in sevoflurane-induced anesthetic arousal regulation. METHODS We used fiber photometry and immunofluorescence staining to monitor changes in neuronal activity during sevoflurane anesthesia. Opto-/chemogenetic modulations were employed to study the effect of neurocircuit modulations during the anesthesia. Anterograde tracing was used to identify a GABAergic projection from the LHA to a periaqueductal gray (PAG) subregion. RESULTS c-Fos staining showed that LHAGABA activity was inhibited by induction of sevoflurane anesthesia. Anterograde tracing revealed that LHAGABA neurons project to multiple arousal-associated brain areas, with the lateral periaqueductal gray (LPAG) being one of the dense projection areas. Optogenetic experiments showed that activation of LHAGABA neurons and their downstream target LPAG reduced the burst suppression ratio (BSR) during continuous sevoflurane anesthesia. Chemogenetic experiments showed that activation of LHAGABA and its projection to LPAG neurons prolonged the anesthetic induction time and promoted wakefulness. CONCLUSIONS In summary, we show that an inhibitory projection from LHAGABA to LPAGGABA neurons promotes arousal from sevoflurane-induced loss of consciousness, suggesting a complex control of wakefulness through intimate interactions between long-range connections.
Collapse
Affiliation(s)
- Dan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Chang Bao
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Huimin Wu
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Jiannan Li
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Xinxin Zhang
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Sa Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Fang Zhou
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Huiming Li
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing HospitalThe Fourth Military Medical UniversityXi'anShaanxiChina
- Key Laboratory of Anesthesiology (The Fourth Military Medical University)Ministry of Education of ChinaXi'anChina
| |
Collapse
|
11
|
Hu Y, Wang Y, Zhang L, Luo M, Wang Y. Neural Network Mechanisms Underlying General Anesthesia: Cortical and Subcortical Nuclei. Neurosci Bull 2024:10.1007/s12264-024-01286-z. [PMID: 39168960 DOI: 10.1007/s12264-024-01286-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/10/2024] [Indexed: 08/23/2024] Open
Abstract
General anesthesia plays a significant role in modern medicine. However, the precise mechanism of general anesthesia remains unclear, posing a key scientific challenge in anesthesiology. Advances in neuroscience techniques have enabled targeted manipulation of specific neural circuits and the capture of brain-wide neural activity at high resolution. These advances hold promise for elucidating the intricate mechanisms of action of general anesthetics. This review aims to summarize our current understanding of the role of cortical and subcortical nuclei in modulating general anesthesia, providing new evidence of cortico-cortical and thalamocortical networks in relation to anesthesia and consciousness. These insights contribute to a comprehensive understanding of the neural network mechanisms underlying general anesthesia.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yun Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Lingjing Zhang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Mengqiang Luo
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
12
|
Zhong CC, Xu Z, Gan J, Yu YM, Tang HM, Zhu Y, Yang JX, Ding HL, Cao JL. Acute Ongoing Nociception Delays Recovery of Consciousness from Sevoflurane Anesthesia via a Midbrain Circuit. J Neurosci 2024; 44:e0740242024. [PMID: 39019613 PMCID: PMC11340287 DOI: 10.1523/jneurosci.0740-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
Although anesthesia provides favorable conditions for surgical procedures, recent studies have revealed that the brain remains active in processing noxious signals even during anesthesia. However, whether and how these responses affect the anesthesia effect remains unclear. The ventrolateral periaqueductal gray (vlPAG), a crucial hub for pain regulation, also plays an essential role in controlling general anesthesia. Hence, it was hypothesized that the vlPAG may be involved in the regulation of general anesthesia by noxious stimuli. Here, we found that acute noxious stimuli, including capsaicin-induced inflammatory pain, acetic acid-induced visceral pain, and incision-induced surgical pain, significantly delayed recovery from sevoflurane anesthesia in male mice, whereas this effect was absent in the spared nerve injury-induced chronic pain. Pretreatment with peripheral analgesics could prevent the delayed recovery induced by acute nociception. Furthermore, we found that acute noxious stimuli, induced by the injection of capsaicin under sevoflurane anesthesia, increased c-Fos expression and activity in the GABAergic neurons of the ventrolateral periaqueductal gray. Specific reactivation of capsaicin-activated vlPAGGABA neurons mimicked the effect of capsaicin and its chemogenetic inhibition prevented the delayed recovery from anesthesia induced by capsaicin. Finally, we revealed that the vlPAGGABA neurons regulated the recovery from anesthesia through the inhibition of ventral tegmental area dopaminergic neuronal activity, thus decreasing dopamine (DA) release and activation of DA D1-like receptors in the brain. These findings reveal a novel, cell- and circuit-based mechanism for regulating anesthesia recovery by nociception, and it is important to provide new insights for guiding the management of the anesthesia recovery period.
Collapse
Affiliation(s)
- Chao-Chao Zhong
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Zheng Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Jun Gan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Yu-Mei Yu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Hui-Mei Tang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Yangzi Zhu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Jun-Xia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China
| |
Collapse
|
13
|
Vincent KF, Park GH, Stapley BM, Dillon EJ, Solt K. Methylphenidate Reversal of Dexmedetomidine-Induced Versus Ketamine-Induced Sedation in Rats. Anesth Analg 2024:00000539-990000000-00894. [PMID: 39110627 DOI: 10.1213/ane.0000000000007085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024]
Abstract
BACKGROUND Dexmedetomidine and ketamine have long elimination half-lives in humans and have no clinically approved reversal agents. Methylphenidate enhances dopaminergic and noradrenergic neurotransmission by inhibiting reuptake transporters for these arousal-promoting neurotransmitters. Previous studies in rats demonstrated that intravenous methylphenidate induces emergence from isoflurane and propofol general anesthesia. These 2 anesthetics are thought to act primarily through enhancement of inhibitory Gamma-aminobutyric acid type A (GABAA) receptors. In this study, we tested the behavioral and neurophysiological effects of methylphenidate in rats after low and high doses of dexmedetomidine (an alpha-2 adrenergic receptor agonist) and ketamine (an N-methyl-D-aspartate [NMDA] receptor antagonist) that induce sedation and unconsciousness, respectively. METHODS All experiments used adult male and female Sprague-Dawley rats (n = 32 total) and all drugs were administered intravenously in a crossover, blinded experimental design. Locomotion after sedating doses of dexmedetomidine (10 µg/kg) or ketamine (10 mg/kg) with and without methylphenidate (5 mg/kg) was tested using the open field test (n = 16). Recovery of righting reflex after either high-dose dexmedetomidine (50 µg/kg) or high-dose ketamine (50 mg/kg) with and without methylphenidate (1-5 mg/kg) was assessed in a second cohort of rats (n = 8). Finally, in a third cohort of rats (n = 8), frontal electroencephalography (EEG) was recorded for spectral analysis under both low and high doses of dexmedetomidine and ketamine with and without methylphenidate. RESULTS Low-dose dexmedetomidine reduced locomotion by 94% in rats. Methylphenidate restored locomotion after low-dose dexmedetomidine (rank difference = 88.5, 95% confidence interval [CI], 70.8-106) and the effect was blocked by coadministration with a dopamine D1 receptor antagonist (rank difference = 86.2, 95% CI, 68.6-104). Low-dose ketamine transiently attenuated mobility by 58% and was not improved with methylphenidate. Methylphenidate did not affect the return of righting reflex latency in rats after high-dose dexmedetomidine nor ketamine. Frontal EEG analysis revealed that methylphenidate reversed spectral changes induced by low-dose dexmedetomidine (F [8,87] = 3.27, P = .003) but produced only transient changes after high-dose dexmedetomidine. Methylphenidate did not induce spectral changes in the EEG after low- or high-dose ketamine. CONCLUSIONS Methylphenidate reversed behavioral and neurophysiological correlates of sedation, but not unconsciousness, induced by dexmedetomidine. In contrast, methylphenidate did not affect sedation, unconsciousness, nor EEG signatures in rats after ketamine. These findings suggest that methylphenidate may be efficacious to reverse dexmedetomidine sedation in humans.
Collapse
Affiliation(s)
- Kathleen F Vincent
- From the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| | - Gwi H Park
- From the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| | - Brendan M Stapley
- From the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Emmaline J Dillon
- From the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Ken Solt
- From the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
14
|
Zhang S, Zhang X, Li H, Wang D, Wang S, Wang Y, Zhao G, Dong H, Li J. Ventral Tegmental Area Glutamatergic Neurons Facilitated Emergence From Isoflurane Anesthesia Involves Excitation of Lateral Septum GABA-ergic Neurons in Mice. Anesth Analg 2024; 139:397-410. [PMID: 38048607 DOI: 10.1213/ane.0000000000006739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
BACKGROUND Ventral tegmental area (VTA) glutamatergic neurons promote wakefulness in the sleep-wake cycle; however, their roles and neural circuit mechanisms during isoflurane (ISO) anesthesia remain unclear. METHODS Fiber photometry and in vivo electrophysiology were used to observe the changes in neuronal or terminal activity during ISO anesthesia and arousal processes. Optogenetic and anesthesia behaviors were used to investigate the effects of VTA glutamatergic neurons and their projections to the lateral septum (LS) during ISO anesthesia and arousal. Anterograde and retrograde tracings were performed to identify the connections between VTA glutamatergic neurons and the LS. RESULTS Population activity and firing rates of VTA glutamatergic neurons decreased during ISO anesthesia (ISO: 95% confidence interval [CI], 0.83-2.06 Spikes.s -1 vs wake: 95% CI, 3.53-7.83 Spikes.s -1 ; P =.0001; n = 34 from 4 mice). Optogenetic activation of VTA glutamatergic neurons reduced the burst-suppression ratio in electroencephalography (laser: 95% CI, 13.09%-28.76% vs pre: 95% CI, 52.85%-71.59%; P =.0009; n = 6) and facilitated emergence (ChR2: 95% CI, 343.3-388.0 seconds vs mCherry: 95% CI, 447.6-509.8 seconds; P < .0001; n = 11/12) from ISO anesthesia. VTA glutamatergic neurons monosynaptically innervated LS γ-aminobutyric acid (GABA)-ergic neurons. The activity of VTA glutamatergic terminals in the LS decreased during ISO anesthesia, and optogenetic activation of the VTA glutamatergic terminals in the LS facilitated emergence from ISO anesthesia. Furthermore, optogenetic activation of VTA glutamatergic terminals increased the firing rates of LS γ-aminobutyric acid-ergic (GABAergic) neurons (laser: 95% CI, 0.85-4.03 Spikes.s -1 vs pre: 95% CI, 0.24-0.78 Spikes.s -1 ; P =.008; n = 23 from 4 mice) during ISO anesthesia. CONCLUSIONS VTA glutamatergic neurons facilitated emergence from ISO anesthesia involving excitation of LS GABAergic neurons.
Collapse
Affiliation(s)
- Simin Zhang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Xinxin Zhang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huiming Li
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dan Wang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Sa Wang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuhao Wang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Guangchao Zhao
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hailong Dong
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiannan Li
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
15
|
Jia L, Yin J, Liu T, Qi W, Du T, Li Q, Ma K, Si J, Yin J, Li Y. Activation of Ventral Tegmental Area Dopaminergic Neurons Projecting to the Parabrachial Nucleus Promotes Emergence from Propofol Anesthesia in Male Rats. Neurochem Res 2024; 49:2060-2074. [PMID: 38814359 DOI: 10.1007/s11064-024-04169-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/22/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
Since the clinical introduction of general anesthesia, its underlying mechanisms have not been fully elucidated. The ventral tegmental area (VTA) and parabrachial nucleus (PBN) play pivotal roles in the mechanisms underlying general anesthesia. However, whether dopaminergic (DA) projections from the VTA to the PBN play a role in mediating the effects of general anesthesia is unclear. We microinjected 6-hydroxydopamine into the PBN to damage tyrosine hydroxylase positive (TH+) neurons and found a prolonged recovery time from propofol anesthesia. We used calcium fiber photometry recording to explore the activity of TH + neurons in the PBN. Then, we used chemogenetic and optogenetic approaches either activate the VTADA-PBN pathway, shortening the propofol anesthesia emergence time, or inhibit this pathway, prolonging the emergence time. These data indicate the crucial involvement of TH + neurons in the PBN in regulating emergence from propofol anesthesia, while the activation of the VTADA-PBN pathway facilitates the emergence of propofol anesthesia.
Collapse
Affiliation(s)
- Lei Jia
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jieting Yin
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Tielong Liu
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Wenqiang Qi
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Tongyu Du
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Quntao Li
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Ketao Ma
- Department of Physiology, School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, China
| | - Junqiang Si
- Department of Physiology, School of Medicine, Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi, China
| | - Jiangwen Yin
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.
| | - Yan Li
- Department of Anesthesiology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.
| |
Collapse
|
16
|
Wang X, Yi R, Liang X, Zhang N, Zhong F, Lu Y, Chen W, Yu T, Zhang L, Wang H, Zhou L. Myelin modulates the process of isoflurane anesthesia through the regulation of neural activity. CNS Neurosci Ther 2024; 30:e14922. [PMID: 39138640 PMCID: PMC11322027 DOI: 10.1111/cns.14922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/01/2024] [Accepted: 08/04/2024] [Indexed: 08/15/2024] Open
Abstract
AIMS The mechanism underlying the reversible unconsciousness induced by general anesthetics (GA) remains unclear. Recent studies revealed the critical roles of myelin and oligodendrocytes (OLs) in higher functions of the brain. However, it is unknown whether myelin actively participates in the regulation of GA. The aim of this study is to investigate the roles and possible mechanisms of myelin in the regulation of consciousness alterations induced by isoflurane anesthesia. METHODS First, demyelination models for the entire brain and specific neural nuclei were established to investigate the potential role of myelination in the regulation of GA, as well as its possible regional specificity. c-Fos staining was then performed on the demyelinated nuclei to verify the impact of myelin loss on neuronal activity. Finally, the activity of neurons during isoflurane anesthesia in demyelinated mice was recorded by optical fiber photometric calcium signal. The related behavioral indicators and EEG were recorded and analyzed. RESULTS A prolonged emergence time was observed from isoflurane anesthesia in demyelinated mice, which suggested the involvement of myelin in regulating GA. The demyelination in distinct nuclei by LPC further clarified the region-specific roles of isoflurane anesthesia regulation by myelin. The effect of demyelination on isoflurane anesthesia in the certain nucleus was consistent with that in neurons towards isoflurane anesthesia. Finally, we found that the mechanism of myelin in the modulation of isoflurane anesthesia is possibly through the regulation of neuronal activity. CONCLUSIONS In brief, myelin in the distinct neural nucleus plays an essential role in regulating the process of isoflurane anesthesia. The possible mechanism of myelin in the regulation of isoflurane anesthesia is neuronal activity modification by myelin integrity during GA. Our findings enhanced the comprehension of myelin function, and offered a fresh perspective for investigating the neural mechanisms of GA.
Collapse
Affiliation(s)
- Xu Wang
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
| | - Rulan Yi
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Xiaoling Liang
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
| | - Ning Zhang
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
| | - Fuwang Zhong
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
| | - Yali Lu
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
| | - Wenjia Chen
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
| | - Tian Yu
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Linyong Zhang
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Haiying Wang
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Liang Zhou
- Key Laboratory of Anesthesia and Organ Protection (Zunyi Medical University), Ministry of EducationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
- Guizhou Key Laboratory of Anesthesia and Organ ProtectionZunyi Medical UniversityZunyiChina
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| |
Collapse
|
17
|
Szabadi E. Three paradoxes related to the mode of action of pramipexole: The path from D2/D3 dopamine receptor stimulation to modification of dopamine-modulated functions. J Psychopharmacol 2024; 38:581-596. [PMID: 39041250 DOI: 10.1177/02698811241261022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Pramipexole, a D2/D3 dopamine receptor agonist, is used to treat the motor symptoms of Parkinson's disease, caused by degeneration of the dopaminergic nigrostriatal pathway. There are three paradoxes associated with its mode of action. Firstly, stimulation of D2/D3 receptors leads to neuronal inhibition, although pramipexole does not inhibit but promotes some dopamine-modulated functions, such as locomotion and reinforcement. Secondly, another dopamine-modulated function, arousal, is not promoted but inhibited by pramipexole, leading to sedation. Thirdly, pramipexole-evoked sedation is associated with an increase in pupil diameter, although sedation is expected to cause pupil constriction. To resolve these paradoxes, the path from stimulation of D2/D3 receptors to the modification of dopamine-modulated functions has been tracked. The functions considered are modulated by midbrain dopaminergic nuclei: locomotion - substantia nigra pars compacta (SNc), reinforcement/motivation - ventral tegmental area (VTA), sympathetic activity (as reflected in pupil function) - VTA; arousal - ventral periaqueductal grey (vPAG), with contributions from VTA and SNc. The application of genetics-based molecular techniques (optogenetics and chemogenetics) has enabled tracing the chains of neurones from the dopaminergic nuclei to their final targets executing the functions. The functional neuronal circuits linked to the D2/D3 receptors in the dorsal and ventral striata, stimulated by inputs from SNc and VTA, respectively, may explain how neuronal inhibition induced by pramipexole is translated into the promotion of locomotion, reinforcement/motivation and sympathetic activity. As the vPAG may increase arousal mainly by stimulating cortical D1 dopamine receptors, pramipexole would stimulate only presynaptic D2/D3 receptors on vPAG neurones, curtailing their activity and leading to sedation.
Collapse
Affiliation(s)
- Elemer Szabadi
- Developmental Psychiatry, University of Nottingham, Nottingham, UK
| |
Collapse
|
18
|
Yu Q, Wang Y, Gu L, Shao W, Gu J, Liu L, Lian X, Xu Q, Zhang Y, Yang Y, Zhang Z, Wu Y, Ma H, Shen Y, Ye W, Wu Y, Yang H, Chen L, Nagayasu K, Zhang H. Dorsal raphe nucleus to basolateral amygdala 5-HTergic neural circuit modulates restoration of consciousness during sevoflurane anesthesia. Biomed Pharmacother 2024; 176:116937. [PMID: 38870632 DOI: 10.1016/j.biopha.2024.116937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/06/2024] [Accepted: 06/09/2024] [Indexed: 06/15/2024] Open
Abstract
The advent of general anesthesia (GA) has significant implications for clinical practice. However, the exact mechanisms underlying GA-induced transitions in consciousness remain elusive. Given some similarities between GA and sleep, the sleep-arousal neural nuclei and circuits involved in sleep-arousal, including the 5-HTergic system, could be implicated in GA. Herein, we utilized pharmacology, optogenetics, chemogenetics, fiber photometry, and retrograde tracing to demonstrate that both endogenous and exogenous activation of the 5-HTergic neural circuit between the dorsal raphe nucleus (DR) and basolateral amygdala (BLA) promotes arousal and facilitates recovery of consciousness from sevoflurane anesthesia. Notably, the 5-HT1A receptor within this pathway holds a pivotal role. Our findings will be conducive to substantially expanding our comprehension of the neural circuit mechanisms underlying sevoflurane anesthesia and provide a potential target for modulating consciousness, ultimately leading to a reduction in anesthetic dose requirements and side effects.
Collapse
Affiliation(s)
- Qian Yu
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - YuLing Wang
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - LeYuan Gu
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - WeiHui Shao
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - JiaXuan Gu
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Lu Liu
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - XiTing Lian
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Qing Xu
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - YuanLi Zhang
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Yue Yang
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - ZhuoYue Zhang
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - YaXuan Wu
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - HaiXiang Ma
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Yue Shen
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China
| | - Wen Ye
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China
| | - YanHui Wu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China
| | - HuiFang Yang
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China
| | - LiHai Chen
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 606-8501, Japan
| | - HongHai Zhang
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou 310006, China; Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
19
|
Barra ME, Solt K, Yu X, Edlow BL. Restoring consciousness with pharmacologic therapy: Mechanisms, targets, and future directions. Neurotherapeutics 2024; 21:e00374. [PMID: 39019729 PMCID: PMC11452330 DOI: 10.1016/j.neurot.2024.e00374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 04/16/2024] [Accepted: 05/03/2024] [Indexed: 07/19/2024] Open
Abstract
Severe brain injury impairs consciousness by disrupting a broad spectrum of neurotransmitter systems. Emerging evidence suggests that pharmacologic modulation of specific neurotransmitter systems, such as dopamine, promotes recovery of consciousness. Clinical guidelines now endorse the use of amantadine in individuals with traumatic disorders of consciousness (DoC) based on level 1 evidence, and multiple neurostimulants are used off-label in clinical practice, including methylphenidate, modafinil, bromocriptine, levodopa, and zolpidem. However, the relative contributions of monoaminergic, glutamatergic, cholinergic, GABAergic, and orexinergic neurotransmitter systems to recovery of consciousness after severe brain injury are unknown, and personalized approaches to targeted therapy have yet to be developed. This review summarizes the state-of-the-science in the neurochemistry and neurobiology of neurotransmitter systems involved in conscious behaviors, followed by a discussion of how pharmacologic therapies may be used to modulate these neurotransmitter systems and promote recovery of consciousness. We consider pharmacologic modulation of consciousness at the synapse, circuit, and network levels, with a focus on the mesocircuit model that has been proposed to explain the consciousness-promoting effects of various monoaminergic, glutamatergic, and paradoxically, GABAergic therapies. Though fundamental questions remain about neurotransmitter mechanisms, target engagement and optimal therapy selection for individual patients, we propose that pharmacologic therapies hold great promise to promote recovery and improve quality of life for patients with severe brain injuries.
Collapse
Affiliation(s)
- Megan E Barra
- Department of Pharmacy, Massachusetts General Hospital, Boston, MA, USA; Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Boston, MA, USA
| | - Ken Solt
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Xin Yu
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Brian L Edlow
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Boston, MA, USA; Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
20
|
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
|
21
|
Mao R, Cavelli ML, Findlay G, Driessen K, Peterson MJ, Marshall W, Tononi G, Cirelli C. Behavioral and cortical arousal from sleep, muscimol-induced coma, and anesthesia by direct optogenetic stimulation of cortical neurons. iScience 2024; 27:109919. [PMID: 38812551 PMCID: PMC11134913 DOI: 10.1016/j.isci.2024.109919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/28/2023] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
The cerebral cortex is widely considered part of the neural substrate of consciousness, but direct causal evidence is missing. Here, we tested in mice whether optogenetic activation of cortical neurons in posterior parietal cortex (PtA) or medial prefrontal cortex (mPFC) is sufficient for arousal from three behavioral states characterized by progressively deeper unresponsiveness: sleep, a coma-like state induced by muscimol injection in the midbrain, and deep sevoflurane-dexmedetomidine anesthesia. We find that cortical stimulation always awakens the mice from both NREM sleep and REM sleep, with PtA requiring weaker/shorter light pulses than mPFC. Moreover, in most cases light pulses produce both cortical activation (decrease in low frequencies) and behavioral arousal (recovery of the righting reflex) from brainstem coma, as well as cortical activation from anesthesia. These findings provide evidence that direct activation of cortical neurons is sufficient for behavioral and/or cortical arousal from sleep, brainstem coma, and anesthesia.
Collapse
Affiliation(s)
- Rong Mao
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI 53719, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53719, USA
| | - Matias Lorenzo Cavelli
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI 53719, USA
- Departamento de Fisiología de Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Graham Findlay
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI 53719, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53719, USA
| | - Kort Driessen
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI 53719, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53719, USA
| | - Michael J. Peterson
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI 53719, USA
| | - William Marshall
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI 53719, USA
- Department of Mathematics and Statistics, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI 53719, USA
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI 53719, USA
| |
Collapse
|
22
|
Mashour GA. Anesthesia and the neurobiology of consciousness. Neuron 2024; 112:1553-1567. [PMID: 38579714 PMCID: PMC11098701 DOI: 10.1016/j.neuron.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 04/07/2024]
Abstract
In the 19th century, the discovery of general anesthesia revolutionized medical care. In the 21st century, anesthetics have become indispensable tools to study consciousness. Here, I review key aspects of the relationship between anesthesia and the neurobiology of consciousness, including interfaces of sleep and anesthetic mechanisms, anesthesia and primary sensory processing, the effects of anesthetics on large-scale functional brain networks, and mechanisms of arousal from anesthesia. I discuss the implications of the data derived from the anesthetized state for the science of consciousness and then conclude with outstanding questions, reflections, and future directions.
Collapse
Affiliation(s)
- George A Mashour
- Center for Consciousness Science, Department of Anesthesiology, Department of Pharmacology, Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
23
|
Rademacher K, Doric Z, Haddad D, Mamaligas A, Liao SC, Creed RB, Kano K, Chatterton Z, Fu Y, Garcia JH, Vance V, Sei Y, Kreitzer A, Halliday GM, Nelson AB, Margolis EB, Nakamura K. Chronic hyperactivation of midbrain dopamine neurons causes preferential dopamine neuron degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588321. [PMID: 38645054 PMCID: PMC11030348 DOI: 10.1101/2024.04.05.588321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Parkinson's disease (PD) is characterized by the death of substantia nigra (SNc) dopamine (DA) neurons, but the pathophysiological mechanisms that precede and drive their death remain unknown. The activity of DA neurons is likely altered in PD, but we understand little about if or how chronic changes in activity may contribute to degeneration. To address this question, we developed a chemogenetic (DREADD) mouse model to chronically increase DA neuron activity, and confirmed this increase using ex vivo electrophysiology. Chronic hyperactivation of DA neurons resulted in prolonged increases in locomotor activity during the light cycle and decreases during the dark cycle, consistent with chronic changes in DA release and circadian disturbances. We also observed early, preferential degeneration of SNc projections, recapitulating the PD hallmarks of selective vulnerability of SNc axons and the comparative resilience of ventral tegmental area axons. This was followed by eventual loss of midbrain DA neurons. Continuous DREADD activation resulted in a sustained increase in baseline calcium levels, supporting an important role for increased calcium in the neurodegeneration process. Finally, spatial transcriptomics from DREADD mice examining midbrain DA neurons and striatal targets, and cross-validation with human patient samples, provided insights into potential mechanisms of hyperactivity-induced toxicity and PD. Our results thus reveal the preferential vulnerability of SNc DA neurons to increased neural activity, and support a potential role for increased neural activity in driving degeneration in PD.
Collapse
Affiliation(s)
- Katerina Rademacher
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Zak Doric
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
| | - Dominik Haddad
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
| | - Aphroditi Mamaligas
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
| | - Szu-Chi Liao
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, CA
- Endocrinology Graduate Program, University of California Berkeley, Berkeley, CA
| | - Rose B. Creed
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA
| | - Kohei Kano
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Zac Chatterton
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Brain and Mind Centre, Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Yuhong Fu
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Brain and Mind Centre, Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Joseph H. Garcia
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- School of Medicine, University of California, San Francisco, California, USA
| | - Victoria Vance
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- College of Science, Northeastern University, Boston, MA
| | - Yoshitaka Sei
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
| | - Anatol Kreitzer
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- UCSF Department of Physiology, University of California San Francisco, CA
| | - Glenda M Halliday
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- Brain and Mind Centre, Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Alexandra B. Nelson
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA
| | - Elyssa B. Margolis
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA
| | - Ken Nakamura
- Gladstone Institute for Neurological Disease, Gladstone Institutes, San Francisco, CA
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco , CA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA
- Graduate Program in Biomedical Sciences, University of California San Francisco, San Francisco
| |
Collapse
|
24
|
Vincent KF, Zhang ER, Cho AJ, Kato-Miyabe R, Mallari OG, Moody OA, Obert DP, Park GH, Solt K. Electrical stimulation of the ventral tegmental area restores consciousness from sevoflurane-, dexmedetomidine-, and fentanyl-induced unconsciousness in rats. Brain Stimul 2024; 17:687-697. [PMID: 38821397 PMCID: PMC11212499 DOI: 10.1016/j.brs.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/15/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Dopaminergic neurons in the ventral tegmental area (VTA) are crucially involved in regulating arousal, making them a potential target for reversing general anesthesia. Electrical deep brain stimulation (DBS) of the VTA restores consciousness in animals anesthetized with drugs that primarily enhance GABAA receptors. However, it is unknown if VTA DBS restores consciousness in animals anesthetized with drugs that target other receptors. OBJECTIVE To evaluate the efficacy of VTA DBS in restoring consciousness after exposure to four anesthetics with distinct receptor targets. METHODS Sixteen adult Sprague-Dawley rats (8 female, 8 male) with bipolar electrodes implanted in the VTA were exposed to dexmedetomidine, fentanyl, ketamine, or sevoflurane to produce loss of righting, a proxy for unconsciousness. After receiving the dopamine D1 receptor antagonist, SCH-23390, or saline (vehicle), DBS was initiated at 30 μA and increased by 10 μA until reaching a maximum of 100 μA. The current that evoked behavioral arousal and restored righting was recorded for each anesthetic and compared across drug (saline/SCH-23390) condition. Electroencephalogram, heart rate and pulse oximetry were recorded continuously. RESULTS VTA DBS restored righting after sevoflurane, dexmedetomidine, and fentanyl-induced unconsciousness, but not ketamine-induced unconsciousness. D1 receptor antagonism diminished the efficacy of VTA stimulation following sevoflurane and fentanyl, but not dexmedetomidine. CONCLUSIONS Electrical DBS of the VTA restores consciousness in animals anesthetized with mechanistically distinct drugs, excluding ketamine. The involvement of the D1 receptor in mediating this effect is anesthetic-specific.
Collapse
Affiliation(s)
- Kathleen F Vincent
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA.
| | - Edlyn R Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Angel J Cho
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Risako Kato-Miyabe
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Olivia G Mallari
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Olivia A Moody
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - David P Obert
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Gwi H Park
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Ken Solt
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Edlow BL, Olchanyi M, Freeman HJ, Li J, Maffei C, Snider SB, Zöllei L, Iglesias JE, Augustinack J, Bodien YG, Haynes RL, Greve DN, Diamond BR, Stevens A, Giacino JT, Destrieux C, van der Kouwe A, Brown EN, Folkerth RD, Fischl B, Kinney HC. Multimodal MRI reveals brainstem connections that sustain wakefulness in human consciousness. Sci Transl Med 2024; 16:eadj4303. [PMID: 38691619 DOI: 10.1126/scitranslmed.adj4303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 04/10/2024] [Indexed: 05/03/2024]
Abstract
Consciousness is composed of arousal (i.e., wakefulness) and awareness. Substantial progress has been made in mapping the cortical networks that underlie awareness in the human brain, but knowledge about the subcortical networks that sustain arousal in humans is incomplete. Here, we aimed to map the connectivity of a proposed subcortical arousal network that sustains wakefulness in the human brain, analogous to the cortical default mode network (DMN) that has been shown to contribute to awareness. We integrated data from ex vivo diffusion magnetic resonance imaging (MRI) of three human brains, obtained at autopsy from neurologically normal individuals, with immunohistochemical staining of subcortical brain sections. We identified nodes of the proposed default ascending arousal network (dAAN) in the brainstem, hypothalamus, thalamus, and basal forebrain. Deterministic and probabilistic tractography analyses of the ex vivo diffusion MRI data revealed projection, association, and commissural pathways linking dAAN nodes with one another and with DMN nodes. Complementary analyses of in vivo 7-tesla resting-state functional MRI data from the Human Connectome Project identified the dopaminergic ventral tegmental area in the midbrain as a widely connected hub node at the nexus of the subcortical arousal and cortical awareness networks. Our network-based autopsy methods and connectivity data provide a putative neuroanatomic architecture for the integration of arousal and awareness in human consciousness.
Collapse
Affiliation(s)
- Brian L Edlow
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Mark Olchanyi
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Holly J Freeman
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Jian Li
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Chiara Maffei
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Samuel B Snider
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Lilla Zöllei
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - J Eugenio Iglesias
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Jean Augustinack
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Yelena G Bodien
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Robin L Haynes
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Douglas N Greve
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Bram R Diamond
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Allison Stevens
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Joseph T Giacino
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Christophe Destrieux
- UMR 1253, iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032, Tours, France
- CHRU de Tours, 2 Boulevard Tonnellé, Tours, France
| | - Andre van der Kouwe
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Emery N Brown
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | | | - Bruce Fischl
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Hannah C Kinney
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
26
|
Song XJ, Hu JJ. Neurobiological basis of emergence from anesthesia. Trends Neurosci 2024; 47:355-366. [PMID: 38490858 DOI: 10.1016/j.tins.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/25/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024]
Abstract
The suppression of consciousness by anesthetics and the emergence of the brain from anesthesia are complex and elusive processes. Anesthetics may exert their inhibitory effects by binding to specific protein targets or through membrane-mediated targets, disrupting neural activity and the integrity and function of neural circuits responsible for signal transmission and conscious perception/subjective experience. Emergence from anesthesia was generally thought to depend on the elimination of the anesthetic from the body. Recently, studies have suggested that emergence from anesthesia is a dynamic and active process that can be partially controlled and is independent of the specific molecular targets of anesthetics. This article summarizes the fundamentals of anesthetics' actions in the brain and the mechanisms of emergence from anesthesia that have been recently revealed in animal studies.
Collapse
Affiliation(s)
- Xue-Jun Song
- Department of Medical Neuroscience and SUSTech Center for Pain Medicine, Southern University of Science and Technology School of Medicine, Shenzhen, China.
| | - Jiang-Jian Hu
- Department of Medical Neuroscience and SUSTech Center for Pain Medicine, Southern University of Science and Technology School of Medicine, Shenzhen, China
| |
Collapse
|
27
|
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
|
28
|
Li M, Li W, Liang S, Liao X, Gu M, Li H, Chen X, Liu H, Qin H, Xiao J. BNST GABAergic neurons modulate wakefulness over sleep and anesthesia. Commun Biol 2024; 7:339. [PMID: 38503808 PMCID: PMC10950862 DOI: 10.1038/s42003-024-06028-5] [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: 03/30/2023] [Accepted: 03/08/2024] [Indexed: 03/21/2024] Open
Abstract
The neural circuits underlying sleep-wakefulness and general anesthesia have not been fully investigated. The GABAergic neurons in the bed nucleus of the stria terminalis (BNST) play a critical role in stress and fear that relied on heightened arousal. Nevertheless, it remains unclear whether BNST GABAergic neurons are involved in the regulation of sleep-wakefulness and anesthesia. Here, using in vivo fiber photometry combined with electroencephalography, electromyography, and video recordings, we found that BNST GABAergic neurons exhibited arousal-state-dependent alterations, with high activities in both wakefulness and rapid-eye movement sleep, but suppressed during anesthesia. Optogenetic activation of these neurons could initiate and maintain wakefulness, and even induce arousal from anesthesia. However, chronic lesion of BNST GABAergic neurons altered spontaneous sleep-wakefulness architecture during the dark phase, but not induction and emergence from anesthesia. Furthermore, we also discovered that the BNST-ventral tegmental area pathway might participate in promoting wakefulness and reanimation from steady-state anesthesia. Collectively, our study explores new elements in neural circuit mechanisms underlying sleep-wakefulness and anesthesia, which may contribute to a more comprehensive understanding of consciousness and the development of innovative anesthetics.
Collapse
Affiliation(s)
- Mengyao Li
- Advanced Institute for Brain and Intelligence, School of Medicine, Guangxi University, Nanning, 530004, China
| | - Wen Li
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Shanshan Liang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Xiang Liao
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, 400044, China
| | - Miaoqing Gu
- Advanced Institute for Brain and Intelligence, School of Medicine, Guangxi University, Nanning, 530004, China
| | - Huiming Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xiaowei Chen
- Advanced Institute for Brain and Intelligence, School of Medicine, Guangxi University, Nanning, 530004, China
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China
| | - Hongliang Liu
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| | - Han Qin
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, 400064, China.
| | - Jingyu Xiao
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| |
Collapse
|
29
|
Cao F, Guo Y, Guo S, Hao X, Yang L, Cao J, Zhou Z, Mi W, Tong L. Prelimbic cortical pyramidal neurons to ventral tegmental area projections promotes arousal from sevoflurane anesthesia. CNS Neurosci Ther 2024; 30:e14675. [PMID: 38488453 PMCID: PMC10941502 DOI: 10.1111/cns.14675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/27/2024] [Accepted: 02/28/2024] [Indexed: 03/18/2024] Open
Abstract
AIMS General anesthesia has been used in surgical procedures for approximately 180 years, yet the precise mechanism of anesthetic drugs remains elusive. There is significant anatomical connectivity between the ventral tegmental area (VTA) and the prelimbic cortex (PrL). Projections from VTA dopaminergic neurons (VTADA ) to the PrL play a role in the transition from sevoflurane anesthesia to arousal. It is still uncertain whether the prelimbic cortex pyramidal neuron (PrLPyr ) and its projections to VTA (PrLPyr -VTA) are involved in anesthesia-arousal regulation. METHODS We employed chemogenetics and optogenetics to selectively manipulate neuronal activity in the PrLPyr -VTA pathway. Electroencephalography spectra and burst-suppression ratios (BSR) were used to assess the depth of anesthesia. Furthermore, the loss or recovery of the righting reflex was monitored to indicate the induction or emergence time of general anesthesia. To elucidate the receptor mechanisms in the PrLPyr -VTA projection's impact on anesthesia and arousal, we microinjected NMDA receptor antagonists (MK-801) or AMPA receptor antagonists (NBQX) into the VTA. RESULTS Our findings show that chemogenetic or optogenetic activation of PrLPyr neurons prolonged anesthesia induction and promoted emergence. Additionally, chemogenetic activation of the PrLPyr -VTA neural pathway delayed anesthesia induction and promoted anesthesia emergence. Likewise, optogenetic activation of the PrLPyr -VTA projections extended the induction time and facilitated emergence from sevoflurane anesthesia. Moreover, antagonizing NMDA receptors in the VTA attenuates the delayed anesthesia induction and promotes emergence caused by activating the PrLPyr -VTA projections. CONCLUSION This study demonstrates that PrLPyr neurons and their projections to the VTA are involved in facilitating emergence from sevoflurane anesthesia, with the PrLPyr -VTA pathway exerting its effects through the activation of NMDA receptors within the VTA.
Collapse
Affiliation(s)
- Fuyang Cao
- Department of AnesthesiologyThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Department of AnesthesiologyThe Sixth Medical Center of Chinese PLA General HospitalBeijingChina
- Chinese PLA Medical SchoolBeijingChina
| | - Yongxin Guo
- Department of AnesthesiologyThe First Medical Center of Chinese PLA General HospitalBeijingChina
| | - Shuting Guo
- Department of AnesthesiologyThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Chinese PLA Medical SchoolBeijingChina
| | - Xinyu Hao
- Department of AnesthesiologyThe First Medical Center of Chinese PLA General HospitalBeijingChina
- Chinese PLA Medical SchoolBeijingChina
| | - Lujia Yang
- Department of AnesthesiologyThe First Medical Center of Chinese PLA General HospitalBeijingChina
| | - Jiangbei Cao
- Department of AnesthesiologyThe First Medical Center of Chinese PLA General HospitalBeijingChina
| | - Zhikang Zhou
- Department of AnesthesiologyThe First Medical Center of Chinese PLA General HospitalBeijingChina
| | - Weidong Mi
- Department of AnesthesiologyThe First Medical Center of Chinese PLA General HospitalBeijingChina
| | - Li Tong
- Department of AnesthesiologyThe First Medical Center of Chinese PLA General HospitalBeijingChina
| |
Collapse
|
30
|
Lu X, Xue J, Lai Y, Tang X. Heterogeneity of mesencephalic dopaminergic neurons: From molecular classifications, electrophysiological properties to functional connectivity. FASEB J 2024; 38:e23465. [PMID: 38315491 DOI: 10.1096/fj.202302031r] [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: 10/07/2023] [Revised: 01/06/2024] [Accepted: 01/22/2024] [Indexed: 02/07/2024]
Abstract
The mesencephalic dopamine (DA) system is composed of neuronal subtypes that are molecularly and functionally distinct, are responsible for specific behaviors, and are closely associated with numerous brain disorders. Existing research has made significant advances in identifying the heterogeneity of mesencephalic DA neurons, which is necessary for understanding their diverse physiological functions and disease susceptibility. Moreover, there is a conflict regarding the electrophysiological properties of the distinct subsets of midbrain DA neurons. This review aimed to elucidate recent developments in the heterogeneity of midbrain DA neurons, including subpopulation categorization, electrophysiological characteristics, and functional connectivity to provide new strategies for accurately identifying distinct subtypes of midbrain DA neurons and investigating the underlying mechanisms of these neurons in various diseases.
Collapse
Affiliation(s)
- Xiaying Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou, China
| | - Jinhua Xue
- Department of Pathophysiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou, China
| | - Yudong Lai
- Department of Human Anatomy, School of Basic Medical Sciences, Gannan Medical University, Ganzhou, China
| | - Xiaolu Tang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, China
| |
Collapse
|
31
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [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
|
32
|
Xia JM, Fan BQ, Yi XW, Ni WW, Zhou Y, Chen DD, Yi WJ, Feng LL, Xia Y, Li SS, Qu WM, Han Y, Huang ZL, Li WX. Medial Septal Glutamatergic Neurons Modulate States of Consciousness during Sevoflurane Anesthesia in Mice. Anesthesiology 2024; 140:102-115. [PMID: 37812765 DOI: 10.1097/aln.0000000000004798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
BACKGROUND Multiple neural structures involved in maintaining wakefulness have been found to promote arousal from general anesthesia. The medial septum is a critical region that modulates arousal behavior. This study hypothesized that glutamatergic neurons in the medial septum play a crucial role in regulating states of consciousness during sevoflurane general anesthesia. METHODS Adult male mice were used in this study. The effects of sevoflurane anesthesia on neuronal activity were determined by fiber photometry. Lesions and chemogenetic manipulations were used to study the effects of the altered activity of medial septal glutamatergic neurons on anesthesia induction, emergence, and sensitivity to sevoflurane. Optogenetic stimulation was used to observe the role of acute activation of medial septal glutamatergic neurons on cortical activity and behavioral changes during sevoflurane-induced continuous steady state of general anesthesia and burst suppression state. RESULTS The authors found that medial septal glutamatergic neuronal activity decreased during sevoflurane anesthesia induction and recovered in the early period of emergence. Chemogenetic activation of medial septal glutamatergic neurons prolonged the induction time (mean ± SD, hM3Dq-clozapine N-oxide vs. hM3Dq-saline, 297.5 ± 60.1 s vs. 229.4 ± 29.9 s, P < 0.001, n = 11) and decreased the emergence time (53.2 ± 11.8 s vs. 77.5 ± 33.5 s, P = 0.025, n = 11). Lesions or chemogenetic inhibition of these neurons produced the opposite effects. During steady state of general anesthesia and deep anesthesia-induced burst suppression state, acute optogenetic activation of medial septal glutamatergic neurons induced cortical activation and behavioral emergence. CONCLUSIONS The study findings reveal that activation of medial septal glutamatergic neurons has arousal-promoting effects during sevoflurane anesthesia in male mice. The activation of these neurons prolongs the induction and accelerates the emergence of anesthesia. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Jun-Ming Xia
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Bing-Qian Fan
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China; Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiu-Wen Yi
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Wen-Wen Ni
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Yu Zhou
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Dan-Dan Chen
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Wen-Jing Yi
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Li-Li Feng
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Ying Xia
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Shuang-Shuang Li
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yuan Han
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wen-Xian Li
- Department of Anesthesiology, Eye and Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
| |
Collapse
|
33
|
He J, Zhu Y, Wu C, Wu J, Chen Y, Yuan M, Cheng Z, Zeng L, Ji X. Transcranial ultrasound neuromodulation facilitates isoflurane-induced general anesthesia recovery and improves cognition in mice. ULTRASONICS 2023; 135:107132. [PMID: 37604030 DOI: 10.1016/j.ultras.2023.107132] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/13/2023] [Accepted: 08/05/2023] [Indexed: 08/23/2023]
Abstract
Delayed arousal and cognitive dysfunction are common, especially in older patients after general anesthesia (GA). Elevating central nervous system serotonin (5-HT) levels can promote recovery from GA and increase synaptic plasticity to improve cognition. Ultrasound neuromodulation has become a noninvasive physical intervention therapy with high spatial resolution and penetration depth, which can modulate neuronal excitability to treat psychiatric and neurodegenerative diseases. This study aims to use ultrasound to noninvasively modulate the brain 5-HT levels of mice to promote recovery from GA and improve cognition in mice. The dorsal raphe nucleus (DRN) of mice during GA was stimulated by the 1.1 MHz ultrasound with a negative pressure of 356 kPa, and the liquid chromatography coupled tandem mass spectrometry (LC-MS/MS) method was used to measure the DRN 5-HT concentrations. The mice's recovery time from GA was assessed, and the cognition was evaluated through spontaneous alternation Y-maze and novel object recognition (NOR) tests. After ultrasound stimulation, the mice's DRN 5-HT levels were significantly increased (control: 554.0 ± 103.2 ng/g, anesthesia + US: 664.2 ± 84.1 ng/g, *p = 0.0389); the GA recovery time (return of the righting reflex (RORR) emergence latency time) of mice was significantly reduced (anesthesia: 331.6 ± 70 s, anesthesia + US: 223.2 ± 67.7 s, *p = 0.0215); the spontaneous rotation behavior score of mice was significantly increased (anesthesia: 59.46 ± 5.26 %, anesthesia + US: 68.55 ± 5.24 %; *p = 0.0126); the recognition index was significantly increased (anesthesia: 55.02 ± 6.23 %, anesthesia + US: 78.52 ± 12.21 %; ***p = 0.0009). This study indicates that ultrasound stimulation of DRN increases serotonin levels, accelerates recovery from anesthesia, and improves cognition, which could be an important strategy for treating delayed arousal, postoperative delirium, or even lasting cognitive dysfunction after GA.
Collapse
Affiliation(s)
- Jiaru He
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Yiyue Zhu
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Canwen Wu
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Junwei Wu
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Yan Chen
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Maodan Yuan
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhongwen Cheng
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Lvming Zeng
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Xuanrong Ji
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
34
|
Piatkevich KD, Boyden ES. Optogenetic control of neural activity: The biophysics of microbial rhodopsins in neuroscience. Q Rev Biophys 2023; 57:e1. [PMID: 37831008 DOI: 10.1017/s0033583523000033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Optogenetics, the use of microbial rhodopsins to make the electrical activity of targeted neurons controllable by light, has swept through neuroscience, enabling thousands of scientists to study how specific neuron types contribute to behaviors and pathologies, and how they might serve as novel therapeutic targets. By activating a set of neurons, one can probe what functions they can initiate or sustain, and by silencing a set of neurons, one can probe the functions they are necessary for. We here review the biophysics of these molecules, asking why they became so useful in neuroscience for the study of brain circuitry. We review the history of the field, including early thinking, early experiments, applications of optogenetics, pre-optogenetics targeted neural control tools, and the history of discovering and characterizing microbial rhodopsins. We then review the biophysical attributes of rhodopsins that make them so useful to neuroscience - their classes and structure, their photocycles, their photocurrent magnitudes and kinetics, their action spectra, and their ion selectivity. Our hope is to convey to the reader how specific biophysical properties of these molecules made them especially useful to neuroscientists for a difficult problem - the control of high-speed electrical activity, with great precision and ease, in the brain.
Collapse
Affiliation(s)
- Kiryl D Piatkevich
- School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Edward S Boyden
- McGovern Institute and Koch Institute, Departments of Brain and Cognitive Sciences, Media Arts and Sciences, and Biological Engineering, K. Lisa Yang Center for Bionics and Center for Neurobiological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| |
Collapse
|
35
|
Lu K, Wang Z, Bai N, Zhao Z, Zhao X, He Y. Selective optogenetic modulation of the PBN terminals in the lateral hypothalamic area and basal forebrain regulates emergence from isoflurane anesthesia in mice. BMC Anesthesiol 2023; 23:328. [PMID: 37784027 PMCID: PMC10544560 DOI: 10.1186/s12871-023-02294-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/26/2023] [Indexed: 10/04/2023] Open
Abstract
While the mechanism of general anesthesia has been extensively studied, the underlying neural circuitry has yet to be fully understood. The parabrachial nucleus (PBN) plays a crucial role in modulating wakefulness and promoting arousal from general anesthesia. However, the specific role of PBN projections in the process of general anesthesia remains unclear. In this study, we bilaterally injected AAV-associated viruses encoding excitatory or inhibitory optogenetic probes into the PBN and implanted optical fibers in the LH or BF area. After four weeks, we optogenetically activated or inhibited the PBN-LH and PBN-BF pathways under 1.5 vol% isoflurane. We calculated the time it took for anesthesia induction and emergence, simultaneously monitoring changes in the burst-suppression ratio using electroencephalogram recording. Our findings indicate that optogenetic activation of the PBN-LH and PBN-BF projections plays a significant role in promoting both cortical and behavioral emergence from isoflurane inhalation, without significantly affecting the induction time. Conversely, photoinhibition of these pathways prolonged the recovery time, with no notable difference observed during the induction phase.In summary, our results demonstrate that the PBN-LH and PBN-BF pathways are crucial for promoting arousal from isoflurane general anesthesia, but do not have a pronounced impact on the induction phase.
Collapse
Affiliation(s)
- Kai Lu
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, Shaanxi, China
- Shaanxi Provincial Key Laboratory of Infection and Immunity, Shannxi, China
| | - Zhenhuan Wang
- Laboratory of Neurobiology, School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Ning Bai
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, Shaanxi, China
| | - Ziyu Zhao
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, Shaanxi, China
| | - Xinrong Zhao
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, Shaanxi, China
| | - Yun He
- Shaanxi Provincial Key Laboratory of Infection and Immunity, Shannxi, China.
- Department of Anesthesiology, Shannxi Provincial Cancer Hospital, Yanta District, 309 Yanta W Rd, Xi'An, 710063, Shaanxi, China.
| |
Collapse
|
36
|
Vincent KF, Solt K. Modulating anesthetic emergence with pathway-selective dopamine signaling. Curr Opin Anaesthesiol 2023; 36:468-475. [PMID: 37552017 PMCID: PMC10528732 DOI: 10.1097/aco.0000000000001293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
PURPOSE OF REVIEW To summarize the recent preclinical findings investigating dopaminergic circuits for their involvement in reversing anesthetic-induced unconsciousness. RECENT FINDINGS The release of dopamine from the ventral tegmental area onto dopamine D1 receptor-expressing neurons in the nucleus accumbens promotes emergence following general anesthesia. Two relevant targets of dopamine D1 receptor-expressing neurons in the nucleus accumbens include the lateral hypothalamus and ventral pallidum. Activating mesocortical dopaminergic projections from the ventral tegmental area to the prelimbic cortex has also been shown to hasten emergence from general anesthesia. In contrast, the nigrostriatal dopamine pathway is not involved in regulating anesthetic emergence. The role of the tuberoinfundibular endocrine dopamine pathway remains to be tested; however, recent studies have identified an important function of neuroendocrine signaling on modulating general anesthesia. SUMMARY Potential avenues for accelerating anesthetic emergence may be found through targeting specific arousal-promoting pathways in the brain. Accumulating evidence from rodent studies manipulating cell type- and circuit-specific signaling pathways have identified dopamine as a potent modulator of general anesthesia. Specifically, dopamine signaling along the mesolimbic and mesocortical pathways plays a fundamental role in regulating consciousness.
Collapse
Affiliation(s)
- Kathleen F. Vincent
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Ken Solt
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
37
|
Guo Y, Song Y, Cao F, Li A, Hao X, Shi W, Zhou Z, Cao J, Liu Y, Mi W, Tong L. Ventrolateral periaqueductal gray GABAergic neurons promote arousal of sevoflurane anesthesia through cortico-midbrain circuit. iScience 2023; 26:107486. [PMID: 37744409 PMCID: PMC10517397 DOI: 10.1016/j.isci.2023.107486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/24/2023] [Accepted: 07/24/2023] [Indexed: 09/26/2023] Open
Abstract
The mechanism of general anesthesia remains elusive. The ventrolateral periaqueductal gray (vlPAG) in the midbrain regulates sleep and awake states. However, the role of vlPAG and its circuits in anesthesia is unclear. We utilized opto/chemogenetics, righting reflex, and electroencephalographic recording to assess consciousness changes. We employed fiber photometry to measure the activity of neurons and neurotransmitters. As a result, photometry recording showed that the activity of GABA neurons in vlPAG decreased during sevoflurane anesthesia and was reactivated after anesthesia. Activating GABAergic neurons in vlPAG promoted arousal during anesthesia, while inhibiting them delayed this process. Furthermore, medial prefrontal cortex (mPFC) to vlPAG pyramidal neurons projections and vlPAG to ventral tegmental area (VTA) GABAergic projections played a prominent role in the anesthesia-awake transition. GABA neurotransmitter activity of VTA synchronized with mPFC-vlPAG pyramidal neuron projections. Therefore, the cortico-midbrain circuits centered on vlPAG GABAergic neurons exert an arousal-promoting effect during sevoflurane anesthesia.
Collapse
Affiliation(s)
- Yongxin Guo
- Department of Anaesthesiology, The First Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Yanping Song
- Department of Anaesthesiology, 922th Hospital of Joint Logistics Support Force, PLA, Hengyang, Hunan, China
| | - Fuyang Cao
- Department of Anaesthesiology, The Sixth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Ao Li
- Department of Anaesthesiology, The First Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Xinyu Hao
- Department of Anaesthesiology, The First Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Wenzhu Shi
- Department of Anaesthesiology, The First Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Zhikang Zhou
- Department of Anaesthesiology, The First Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Jiangbei Cao
- Department of Anaesthesiology, The First Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Yanhong Liu
- Department of Anaesthesiology, The First Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Weidong Mi
- Department of Anaesthesiology, The First Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Li Tong
- Department of Anaesthesiology, The First Medical Centre of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
38
|
Geisler CE, Hayes MR. Metabolic hormone action in the VTA: Reward-directed behavior and mechanistic insights. Physiol Behav 2023; 268:114236. [PMID: 37178855 PMCID: PMC10330780 DOI: 10.1016/j.physbeh.2023.114236] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/10/2023] [Accepted: 05/10/2023] [Indexed: 05/15/2023]
Abstract
Dysfunctional signaling in midbrain reward circuits perpetuates diseases characterized by compulsive overconsumption of rewarding substances such as substance abuse, binge eating disorder, and obesity. Ventral tegmental area (VTA) dopaminergic activity serves as an index for how rewarding stimuli are perceived and triggers behaviors necessary to obtain future rewards. The evolutionary linking of reward with seeking and consuming palatable foods ensured an organism's survival, and hormone systems that regulate appetite concomitantly developed to regulate motivated behaviors. Today, these same mechanisms serve to regulate reward-directed behavior around food, drugs, alcohol, and social interactions. Understanding how hormonal regulation of VTA dopaminergic output alters motivated behaviors is essential to leveraging therapeutics that target these hormone systems to treat addiction and disordered eating. This review will outline our current understanding of the mechanisms underlying VTA action of the metabolic hormones ghrelin, glucagon-like peptide-1, amylin, leptin, and insulin to regulate behavior around food and drugs of abuse, highlighting commonalities and differences in how these five hormones ultimately modulate VTA dopamine signaling.
Collapse
Affiliation(s)
- Caroline E Geisler
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
39
|
Edlow BL, Olchanyi M, Freeman HJ, Li J, Maffei C, Snider SB, Zöllei L, Iglesias JE, Augustinack J, Bodien YG, Haynes RL, Greve DN, Diamond BR, Stevens A, Giacino JT, Destrieux C, van der Kouwe A, Brown EN, Folkerth RD, Fischl B, Kinney HC. Sustaining wakefulness: Brainstem connectivity in human consciousness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.13.548265. [PMID: 37502983 PMCID: PMC10369992 DOI: 10.1101/2023.07.13.548265] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Consciousness is comprised of arousal (i.e., wakefulness) and awareness. Substantial progress has been made in mapping the cortical networks that modulate awareness in the human brain, but knowledge about the subcortical networks that sustain arousal is lacking. We integrated data from ex vivo diffusion MRI, immunohistochemistry, and in vivo 7 Tesla functional MRI to map the connectivity of a subcortical arousal network that we postulate sustains wakefulness in the resting, conscious human brain, analogous to the cortical default mode network (DMN) that is believed to sustain self-awareness. We identified nodes of the proposed default ascending arousal network (dAAN) in the brainstem, hypothalamus, thalamus, and basal forebrain by correlating ex vivo diffusion MRI with immunohistochemistry in three human brain specimens from neurologically normal individuals scanned at 600-750 μm resolution. We performed deterministic and probabilistic tractography analyses of the diffusion MRI data to map dAAN intra-network connections and dAAN-DMN internetwork connections. Using a newly developed network-based autopsy of the human brain that integrates ex vivo MRI and histopathology, we identified projection, association, and commissural pathways linking dAAN nodes with one another and with cortical DMN nodes, providing a structural architecture for the integration of arousal and awareness in human consciousness. We release the ex vivo diffusion MRI data, corresponding immunohistochemistry data, network-based autopsy methods, and a new brainstem dAAN atlas to support efforts to map the connectivity of human consciousness.
Collapse
Affiliation(s)
- Brian L. Edlow
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
| | - Mark Olchanyi
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Holly J. Freeman
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
| | - Jian Li
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
| | - Chiara Maffei
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
| | - Samuel B. Snider
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Lilla Zöllei
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
| | - J. Eugenio Iglesias
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
| | - Jean Augustinack
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
| | - Yelena G. Bodien
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital and Harvard Medical School, Charlestown, MA 02129 USA
| | - Robin L. Haynes
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Douglas N. Greve
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
| | - Bram R. Diamond
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
| | - Allison Stevens
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
| | - Joseph T. Giacino
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital and Harvard Medical School, Charlestown, MA 02129 USA
| | - Christophe Destrieux
- UMR 1253, iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032, Tours, France
- CHRU de Tours, 2 Boulevard Tonnellé, Tours, France
| | - Andre van der Kouwe
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
| | - Emery N. Brown
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Bruce Fischl
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hannah C. Kinney
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
40
|
Vincent KF, Mallari OG, Dillon EJ, Stewart VG, Cho AJ, Dong Y, Edlow AG, Ichinose F, Xie Z, Solt K. Oestrous cycle affects emergence from anaesthesia with dexmedetomidine, but not propofol, isoflurane, or sevoflurane, in female rats. Br J Anaesth 2023; 131:67-78. [PMID: 37142466 PMCID: PMC10308440 DOI: 10.1016/j.bja.2023.03.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/17/2023] [Accepted: 03/13/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Although sex differences in anaesthetic sensitivity have been reported, what underlies these differences is unknown. In rodents, one source of variability in females is the oestrous cycle. Here we test the hypothesis that the oestrous cycle impacts emergence from general anaesthesia. METHODS Time to emergence was measured after isoflurane (2 vol% for 1 h), sevoflurane (3 vol% for 20 min), dexmedetomidine (50 μg kg-1 i.v., infused over 10 min), or propofol (10 mg kg-1 i.v. bolus) during proestrus, oestrus, early dioestrus, and late dioestrus in female Sprague-Dawley rats (n=24). EEG recordings were taken during each test for power spectral analysis. Serum was analysed for 17β-oestradiol and progesterone concentrations. The effect of oestrous cycle stage on return of righting latency was assessed using a mixed model. The association between righting latency and serum hormone concentration was tested by linear regression. Mean arterial blood pressure and arterial blood gases were assessed in a subset of rats after dexmedetomidine and compared in a mixed model. RESULTS Oestrous cycle did not affect righting latency after isoflurane, sevoflurane, or propofol. When in the early dioestrus stage, rats emerged more rapidly from dexmedetomidine than in the proestrus (P=0.0042) or late dioestrus (P=0.0230) stage and showed reduced overall power in frontal EEG spectra 30 min after dexmedetomidine (P=0.0049). 17β-Oestradiol and progesterone serum concentrations did not correlate with righting latency. Oestrous cycle did not affect mean arterial blood pressure or blood gases during dexmedetomidine. CONCLUSIONS In female rats, the oestrous cycle significantly impacts emergence from dexmedetomidine-induced unconsciousness. However, 17β-oestradiol and progesterone serum concentrations do not correlate with the observed changes.
Collapse
Affiliation(s)
- Kathleen F Vincent
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Olivia G Mallari
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Emmaline J Dillon
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Brigham Young University, Provo, UT, USA
| | - Victoria G Stewart
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Brigham Young University, Provo, UT, USA
| | - Angel J Cho
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Touro College of Osteopathic Medicine, New York, NY, USA
| | - Yuanlin Dong
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Andrea G Edlow
- Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | - Fumito Ichinose
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Ken Solt
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Anaesthesia, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Bao W, Ding J, Jiang S, Yao Z, Qu W, Li W, Huang Z, Han Y. Selective Activation of NAc D1R-VP/LH Circuits Promotes Reanimation From Sevoflurane Anesthesia in Mice. Anesth Analg 2023; 137:87-97. [PMID: 36944111 DOI: 10.1213/ane.0000000000006436] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
BACKGROUND Emerging evidence has uncovered a vital role of nucleus accumbens (NAc) neurons that express the dopamine D1 receptor (D1R) and its upstream neural circuit in general anesthesia (GA) regulation. However, the underlying downstream neural basis of the modulation of GA emergence by NAc D1R neurons remains unknown. In the present study, we explored the downstream neural mechanism of NAc D1R neurons in the modulation of emergence from sevoflurane GA. METHODS We traced the axonal projections of NAc D1R neurons using a cell type-specific anterograde tracing method and immunohistochemical techniques in D1R-Cre mice. Optogenetic stimulations combined with electroencephalogram/electromyogram recordings and behavioral tests were used to determine the effects of optogenetic activation of the axonal terminals of NAc D1R neurons on sevoflurane emergence during sevoflurane-induced continuous, steady-state general anesthesia (CSSGA) or burst-suppression oscillations. RESULTS Labeled efferent fibers of NAc D1R neurons were highly distributed in the ventral pallidum (VP), lateral hypothalamus (LH), and substantia nigra pars compacta. Optogenetic activation of the NAc D1R -VP circuit during CSSGA with sevoflurane induced cortical activation (mean ± standard deviation [SD]; delta power: prestimulation versus during stimulation, 48.7% ± 5.7% vs 35.1% ± 3.3%, P < .0001; beta power: 7.1% ± 2.7% vs 14.2% ± 3.3%, P = .0264) and behavioral emergence, and restored the righting reflex in 66.7% of ChR2 mice. Optogenetic stimulation of the NAc D1R -LH circuit also produced cortical activation (delta power: prestimulation versus during stimulation, 45.0% ± 6.5% vs 36.1% ± 4.6%, P = .0016) and behavioral emergence, and restored the righting reflex in 100% of the ChR2 mice during CSSGA with sevoflurane. Under a sevoflurane-induced burst-suppression state, NAc D1R -VP/LH circuit activation produced evidence of cortical activation (burst-suppression ratio [BSR]: NAc D1R -VP circuit, prestimulation versus during stimulation, 42.4% ± 4.0% vs 26.3% ± 6.0%, P = .0120; prestimulation versus poststimulation, 42.4% ± 4.0% vs 5.9% ± 5.6%, P = .0002; BSR: NAc D1R -LH circuit, prestimulation versus during stimulation, 33.3% ± 13.4% vs 5.1% ± 4.9%, P = .0177; prestimulation vs poststimulation, 33.3% ± 13.4% vs 3.2% ± 4.0%, P = .0105) and behavioral emergence. CONCLUSIONS Both NAc D1R -VP and NAc D1R -LH circuits are sufficient to promote reanimation from sevoflurane GA by simultaneously inducing cortical and behavioral emergence.
Collapse
Affiliation(s)
- Weiwei Bao
- From the Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiahui Ding
- From the Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Shan Jiang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen Yao
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weimin Qu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenxian Li
- From the Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Zhili Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuan Han
- From the Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Xu Z, Hu SW, Zhou Y, Guo Q, Wang D, Gao YH, Zhao WN, Tang HM, Yang JX, Yu X, Ding HL, Cao JL. Corticotropin-releasing factor neurones in the paraventricular nucleus of the hypothalamus modulate isoflurane anaesthesia and its responses to acute stress in mice. Br J Anaesth 2023; 130:446-458. [PMID: 36737387 DOI: 10.1016/j.bja.2022.12.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 11/23/2022] [Accepted: 12/23/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Corticotropin-releasing factor (CRF) neurones in the paraventricular nucleus (PVN) of the hypothalamus (PVNCRF neurones) can promote wakefulness and are activated under anaesthesia. However, whether these neurones contribute to anaesthetic effects is unknown. METHODS With a combination of chemogenetic and molecular approaches, we examined the roles of PVNCRF neurones in isoflurane anaesthesia in mice and further explored the underlying cellular and molecular mechanisms. RESULTS PVN neurones exhibited increased Fos expression during isoflurane anaesthesia (mean [standard deviation], 218 [69.3] vs 21.3 [7.3]; P<0.001), and ∼75% were PVNCRF neurones. Chemogenetic inhibition of PVNCRF neurones facilitated emergence from isoflurane anaesthesia (11.7 [1.1] vs 13.9 [1.2] min; P=0.001), whereas chemogenetic activation of these neurones delayed emergence from isoflurane anaesthesia (16.9 [1.2] vs 13.9 [1.3] min; P=0.002). Isoflurane exposure increased CRF protein expression in PVN (4.0 [0.1] vs 2.2 [0.3], respectively; P<0.001). Knockdown of CRF in PVNCRF neurones mimicked the effects of chemogenetic inhibition of PVNCRF neurones in facilitating emergence (9.6 [1.1] vs 13.0 [1.4] min; P=0.003) and also abolished the effects of chemogenetic activation of PVNCRF neurones on delaying emergence from isoflurane anaesthesia (10.3 [1.3] vs 16.0 [2.6] min; P<0.001). Acute, but not chronic, stress delayed emergence from isoflurane anaesthesia (15.5 [1.5] vs 13.0 [1.4] min; P=0.004). This effect was reversed by chemogenetic inhibition of PVNCRF neurones (11.7 [1.6] vs 14.7 [1.4] min; P=0.001) or knockdown of CRF in PVNCRF neurones (12.3 [1.5] vs 15.3 [1.6] min; P=0.002). CONCLUSIONS CRF neurones in the PVN of the hypothalamus neurones modulate isoflurane anaesthesia and acute stress effects on anaesthesia through CRF signalling.
Collapse
Affiliation(s)
- Zheng Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Su-Wan Hu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Yu Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Qingchen Guo
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Di Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Yi-Hong Gao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Wei-Nan Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Hui-Mei Tang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Jun-Xia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Xiaolu Yu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China; Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
43
|
Wang J, Miao X, Sun Y, Li S, Wu A, Wei C. Dopaminergic System in Promoting Recovery from General Anesthesia. Brain Sci 2023; 13:brainsci13040538. [PMID: 37190503 DOI: 10.3390/brainsci13040538] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/22/2023] [Accepted: 03/22/2023] [Indexed: 05/17/2023] Open
Abstract
Dopamine is an important neurotransmitter that plays a biological role by binding to dopamine receptors. The dopaminergic system regulates neural activities, such as reward and punishment, memory, motor control, emotion, and sleep-wake. Numerous studies have confirmed that the dopaminergic system has the function of maintaining wakefulness in the body. In recent years, there has been increasing evidence that the sleep-wake cycle in the brain has similar neurobrain network mechanisms to those associated with the loss and recovery of consciousness induced by general anesthesia. With the continuous development and innovation of neurobiological techniques, the dopaminergic system has now been proved to be involved in the emergence from general anesthesia through the modulation of neuronal activity. This article is an overview of the dopaminergic system and the research progress into its role in wakefulness and general anesthesia recovery. It provides a theoretical basis for interpreting the mechanisms regulating consciousness during general anesthesia.
Collapse
Affiliation(s)
- Jinxu Wang
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaolei Miao
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yi Sun
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Sijie Li
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
44
|
Weiss E, Kann M, Wang Q. Neuromodulation of Neural Oscillations in Health and Disease. BIOLOGY 2023; 12:371. [PMID: 36979063 PMCID: PMC10045166 DOI: 10.3390/biology12030371] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/16/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023]
Abstract
Using EEG and local field potentials (LFPs) as an index of large-scale neural activities, research has been able to associate neural oscillations in different frequency bands with markers of cognitive functions, goal-directed behavior, and various neurological disorders. While this gives us a glimpse into how neurons communicate throughout the brain, the causality of these synchronized network activities remains poorly understood. Moreover, the effect of the major neuromodulatory systems (e.g., noradrenergic, cholinergic, and dopaminergic) on brain oscillations has drawn much attention. More recent studies have suggested that cross-frequency coupling (CFC) is heavily responsible for mediating network-wide communication across subcortical and cortical brain structures, implicating the importance of neurotransmitters in shaping coordinated actions. By bringing to light the role each neuromodulatory system plays in regulating brain-wide neural oscillations, we hope to paint a clearer picture of the pivotal role neural oscillations play in a variety of cognitive functions and neurological disorders, and how neuromodulation techniques can be optimized as a means of controlling neural network dynamics. The aim of this review is to showcase the important role that neuromodulatory systems play in large-scale neural network dynamics, informing future studies to pay close attention to their involvement in specific features of neural oscillations and associated behaviors.
Collapse
Affiliation(s)
| | | | - Qi Wang
- Department of Biomedical Engineering, Columbia University, ET 351, 500 W. 120th Street, New York, NY 10027, USA
| |
Collapse
|
45
|
Wang YL, Wang L, Xu W, He M, Dong H, Shi HY, Chen YQ, Huang ZL. Paraventricular thalamus controls consciousness transitions during propofol anaesthesia in mice. Br J Anaesth 2023; 130:698-708. [PMID: 36828739 DOI: 10.1016/j.bja.2023.01.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/20/2022] [Accepted: 01/04/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND The neuronal mechanisms underlying propofol-induced modulation of consciousness are poorly understood. Neuroimaging studies suggest a potential role for non-specific thalamic nuclei in propofol-induced loss of consciousness. We investigated the contribution of the paraventricular thalamus (PVT), a midline thalamic nucleus that has been implicated in arousal control and general anaesthesia with inhaled anaesthetics, to loss and recovery of consciousness during propofol anaesthesia. METHODS Polysomnographic recordings and righting reflex test were used to determine the transitions of loss and recovery of righting reflex, used as a measure of consciousness in mice, during propofol anaesthesia in mice under conditions mimicking clinical propofol administration. PVT neuronal activities were monitored using fibre photometry and regulated using optogenetic and chemogenetic methods. RESULTS Population activities of PVT glutamatergic neurones began to decrease before propofol-induced loss of consciousness and rapidly increased to a peak at the onset of recovery of consciousness. Chemogenetic inhibition of PVT calretinin-expressing (PVTCR) neurones shortened onset (from 176 [35] to 127 [26] s; P=0.001) and prolonged return (from 1568 [611] to 3126 [1616] s; P=0.002) of righting reflex. Conversely, chemogenetic activation of PVTCR neurones exerted opposite effects. Furthermore, optogenetic silencing of PVTCR neurones accelerated transitions to loss of consciousness (from 205 [35] to 158 [44] s; P=0.027) and slowed transitions to recovery of consciousness (from 230 [78] to 370 [99] s; P=0.041). During a steady period of unconsciousness maintained with continuous propofol infusion, brief optical activation of PVTCR neurones restored cortical activity and arousal with a latency of about 5 s. CONCLUSIONS The paraventricular thalamus contributes to the control of consciousness transitions in propofol anaesthesia in mice. This provides a potential neuroanatomical target for controlling consciousness to reduce anaesthetic dose requirements and side effects.
Collapse
Affiliation(s)
- Yu-Long Wang
- Department of Anaesthesiology, Yijishan Hospital, Wannan Medical College, Wuhu, China; Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lu Wang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China.
| | - Wei Xu
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Miao He
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui Dong
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Huan-Ying Shi
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yong-Quan Chen
- Department of Anaesthesiology, Yijishan Hospital, Wannan Medical College, Wuhu, China.
| | - Zhi-Li Huang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
46
|
Fifel K, Yanagisawa M, Deboer T. Mechanisms of Sleep/Wake Regulation under Hypodopaminergic State: Insights from MitoPark Mouse Model of Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2203170. [PMID: 36515271 PMCID: PMC9929135 DOI: 10.1002/advs.202203170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 11/16/2022] [Indexed: 06/17/2023]
Abstract
Sleep/wake alterations are predominant in neurological and neuropsychiatric disorders involving dopamine dysfunction. Unfortunately, specific, mechanisms-based therapies for these debilitating sleep problems are currently lacking. The pathophysiological mechanisms of sleep/wake alterations within a hypodopaminergic MitoPark mouse model of Parkinson's disease (PD) are investigated. MitoPark mice replicate most PD-related sleep alterations, including sleep fragmentation, hypersomnia, and daytime sleepiness. Surprisingly, these alterations are not accounted for by a dysfunction in the circadian or homeostatic regulatory processes of sleep, nor by acute masking effects of light or darkness. Rather, the sleep phenotype is linked with the impairment of instrumental arousal and sleep modulation by behavioral valence. These alterations correlate with changes in high-theta (8-11.5 Hz) electroencephalogram power density during motivationally-charged wakefulness. These results demonstrate that sleep/wake alterations induced by dopamine dysfunction are mediated by impaired modulation of sleep by motivational valence and provide translational insights into sleep problems associated with disorders linked to dopamine dysfunction.
Collapse
Affiliation(s)
- Karim Fifel
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of Tsukuba1‐1‐1 TennodaiTsukubaIbaraki305–8575Japan
- Department of Cell and Chemical BiologyLaboratory of NeurophysiologyLeiden University Medical CenterP.O. Box 9600Leiden2300 RCThe Netherlands
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of Tsukuba1‐1‐1 TennodaiTsukubaIbaraki305–8575Japan
| | - Tom Deboer
- Department of Cell and Chemical BiologyLaboratory of NeurophysiologyLeiden University Medical CenterP.O. Box 9600Leiden2300 RCThe Netherlands
| |
Collapse
|
47
|
Lu Z, Huo T, Deng J, Guo F, Liu K, Liu P, Wang Q, Xiong L. Transcutaneous electrical acupoint stimulation induced sedative effects in healthy volunteers: A resting-state fMRI study. Front Hum Neurosci 2023; 16:843186. [PMID: 36741778 PMCID: PMC9893780 DOI: 10.3389/fnhum.2022.843186] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 12/06/2022] [Indexed: 01/21/2023] Open
Abstract
Background Previous studies indicated the sedative effect of acupoint stimulation. However, its mechanism remains unclear. This study aimed to investigate the sedative effect of transcutaneous electrical acupoint stimulation (TEAS) and to explore the brain regions involved in this effect in healthy volunteers using functional magnetic resonance imaging (fMRI) techniques. Methods In this randomized trial, 26 healthy volunteers were randomly assigned to the TEAS group (receiving 30 min of acupoint stimulation at HT7/PC4) and the control group. fMRI was conducted before and after the intervention. The primary outcome was the BIS value during the intervention. Secondary outcomes included the amplitude of low-frequency fluctuation (ALFF) and region of interest (ROI)-based functional connectivity (FC) showed by fMRI. Results In healthy volunteers, compared with the control group, ALFF values in the TEAS-treated volunteers decreased in the left thalamus, right putamen, and midbrain, while they increased in the left orbitofrontal cortex. More FC existed between the thalamus and the insula, middle cingulate cortex, somatosensory cortex, amygdala, and putamen in subjects after TEAS treatment compared with subjects that received non-stimulation. In addition, ALFF values of the thalamus positively correlated with BIS in both groups. Conclusion Transcutaneous electrical acupoint stimulation could induce a sedative effect in healthy volunteers, and inhibition of the thalamus was among its possible mechanisms. Clinical trial registration www.ClinicalTrials.gov; identifier: NCT01896063.
Collapse
Affiliation(s)
- Zhihong Lu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China,*Correspondence: Lize Xiong ✉
| | - Tingting Huo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiao Deng
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fan Guo
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kang Liu
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Peng Liu
- Life Sciences Research Center, School of Life Sciences and Technology, Xidian University, Xi'an, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lize Xiong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China,Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China,Zhihong Lu ✉
| |
Collapse
|
48
|
Liu PC, Yao W, Chen XY, Su WK, Zheng ZH, Yan XB, Deng YL, Shi KG, Liu X, Gao YW, Lin TT, Zhu YX, Lin YX, Zhu ZH, Cai P, Zhang LC, Chen L. Parabrachial nucleus astrocytes regulate wakefulness and isoflurane anesthesia in mice. Front Pharmacol 2023; 13:991238. [PMID: 36712675 PMCID: PMC9880442 DOI: 10.3389/fphar.2022.991238] [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: 07/11/2022] [Accepted: 12/19/2022] [Indexed: 01/15/2023] Open
Abstract
Background: The parabrachial nucleus (PBN) is an important structure regulating the sleep-wake behavior and general anesthesia. Astrocytes in the central nervous system modulate neuronal activity and consequential behavior. However, the specific role of the parabrachial nucleus astrocytes in regulating the sleep-wake behavior and general anesthesia remains unclear. Methods: We used chemogenetic approach to activate or inhibit the activity of PBN astrocytes by injecting AAV-GFAabc1d-hM3Dq-eGFP or AAV-GFAabc1d-hM4Di-eGFP into the PBN. We investigated the effects of intraperitoneal injection of CNO or vehicle on the amount of wakefulness, NREM sleep and REM sleep in sleep-wake behavior, and on the time of loss of righting reflex, time of recovery of righting reflex, sensitivity to isoflurane, electroencephalogram (EEG) power spectrum and burst suppression ratio (BSR) in isoflurane anesthesia. Results: The activation of PBN astrocytes increased wakefulness amount for 4 h, while the inhibition of PBN astrocytes decreased total amount of wakefulness during the 3-hour post-injection period. Chemogenetic activation of PBN astrocytes decreased isoflurane sensitivity and shortened the emergence time from isoflurane-induced general anesthesia. Cortical EEG recordings revealed that PBN astrocyte activation decreased the EEG delta power and BSR during isoflurane anesthesia. Chemogenetic Inhibition of PBN astrocytes increased the EEG delta power and BSR during isoflurane anesthesia. Conclusion: PBN astrocytes are a key neural substrate regulating wakefulness and emergence from isoflurane anesthesia.
Collapse
Affiliation(s)
- Pei-Chang Liu
- Department of Anesthesiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Wei Yao
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Xing-Yu Chen
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Wei-Kun Su
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Ze-Hong Zheng
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiong-Bin Yan
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Ya-Ling Deng
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Kai-Ge Shi
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Xin Liu
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Yu-Wei Gao
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Tian-Tian Lin
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Yun-Xi Zhu
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Ying-Xuan Lin
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhong-Hua Zhu
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Ping Cai
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China,*Correspondence: Li Chen, ; Liang-Cheng Zhang, ; Ping Cai,
| | - Liang-Cheng Zhang
- Department of Anesthesiology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China,*Correspondence: Li Chen, ; Liang-Cheng Zhang, ; Ping Cai,
| | - Li Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China,*Correspondence: Li Chen, ; Liang-Cheng Zhang, ; Ping Cai,
| |
Collapse
|
49
|
Luo M, Fei X, Liu X, Jin Z, Wang Y, Xu M. Divergent Neural Activity in the VLPO During Anesthesia and Sleep. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2203395. [PMID: 36461756 PMCID: PMC9839870 DOI: 10.1002/advs.202203395] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/10/2022] [Indexed: 05/27/2023]
Abstract
The invention of general anesthesia (GA) represents a significant advance in modern clinical practices. However, the exact mechanisms of GA are not entirely understood. Because of the multitude of similarities between GA and sleep, one intriguing hypothesis is that anesthesia may engage the sleep-wake regulation circuits. Here, using fiber photometry and micro-endoscopic imaging of Ca2+ signals at both population and single-cell levels, it investigates how various anesthetics modulate the neural activity in the ventrolateral preoptic nucleus (vLPO), a brain region essential for the initiation of sleep. It is found that different anesthetics primarily induced suppression of neural activity and tended to recruit a similar group of vLPO neurons; however, each anesthetic caused comparable modulations of both wake-active and sleep-active neurons. These results demonstrate that anesthesia creates a different state of neural activity in the vLPO than during natural sleep, suggesting that anesthesia may not engage the same vLPO circuits for sleep generation.
Collapse
Affiliation(s)
- Mengqiang Luo
- Department of AnesthesiologyHuashan HospitalFudan UniversityShanghai200040China
| | - Xiang Fei
- Institute of NeuroscienceState Key Laboratory of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
| | - Xiaotong Liu
- Institute of NeuroscienceState Key Laboratory of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
| | - Zikang Jin
- Institute of NeuroscienceState Key Laboratory of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
| | - Yingwei Wang
- Department of AnesthesiologyHuashan HospitalFudan UniversityShanghai200040China
| | - Min Xu
- Institute of NeuroscienceState Key Laboratory of NeuroscienceCenter for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
- Shanghai Center for Brain Science and Brain‐Inspired Intelligence TechnologyShanghai201210China
| |
Collapse
|
50
|
Wang Y, Song Y, Tong L, Wang L, Cao J, Qin G, Liu X, Mi W, Wang E, Guo Y. GABAergic neurons in the dorsomedial hypothalamus regulate states of consciousness in sevoflurane anesthesia. iScience 2022; 26:105913. [PMID: 36686391 PMCID: PMC9852568 DOI: 10.1016/j.isci.2022.105913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/12/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
The neural inhibitory gamma-aminobutyric acid (GABA) system in the regulation of anesthetic consciousness is heterogeneous, and the medial hypothalamus (MH), consisting of ventromedial hypothalamus (VMH) and dorsomedial hypothalamus (DMH), plays an important role in sleep and circadian rhythm. However, the role of MH GABAergic neurons (MHGABA) in anesthesia remains unclear. In this study, we used righting reflex, electroencephalogram (EEG), and arousal behavioral score to evaluate the sevoflurane anesthesia. Activation of MHGABA or DMHGABA neurons prolonged the anesthesia induction time, shortened the anesthesia emergence time, and induced EEG arousal and body movement during anesthesia; meanwhile, VMHGABA neurons activated only induced EEG changes during 1.5% sevoflurane anesthesia. Furthermore, inhibition of DMHGABA neurons significantly deepened sevoflurane anesthesia. Therefore, DMHGABA neurons exert a strong emergence-promoting effect on induction, maintenance, and arousal during sevoflurane general anesthesia, which helps to reveal the mechanism of anesthesia.
Collapse
Affiliation(s)
- Yanfeng Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yanping Song
- Department of Anesthesia, 922 Hospital of PLA, Hengyang, Hunan 421002, China
| | - Li Tong
- Anesthesia and Operation Centre, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Lu Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jiangbei Cao
- Anesthesia and Operation Centre, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - Gang Qin
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xingyang Liu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Weidong Mi
- Anesthesia and Operation Centre, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China
| | - E. Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China,Corresponding author
| | - Yongxin Guo
- Anesthesia and Operation Centre, the First Medical Centre of Chinese PLA General Hospital, Beijing 100853, China,Corresponding author
| |
Collapse
|