1
|
Su CW, Yang F, Lai R, Li Y, Naeem H, Yao N, Zhang SP, Zhang H, Li Y, Huang ZG. Unraveling the functional complexity of the locus coeruleus-norepinephrine system: insights from molecular anatomy to neurodynamic modeling. Cogn Neurodyn 2025; 19:29. [PMID: 39866663 PMCID: PMC11757662 DOI: 10.1007/s11571-024-10208-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/08/2024] [Accepted: 09/29/2024] [Indexed: 01/28/2025] Open
Abstract
The locus coeruleus (LC), as the primary source of norepinephrine (NE) in the brain, is central to modulating cognitive and behavioral processes. This review synthesizes recent findings to provide a comprehensive understanding of the LC-NE system, highlighting its molecular diversity, neurophysiological properties, and role in various brain functions. We discuss the heterogeneity of LC neurons, their differential responses to sensory stimuli, and the impact of NE on cognitive processes such as attention and memory. Furthermore, we explore the system's involvement in stress responses and pain modulation, as well as its developmental changes and susceptibility to stressors. By integrating molecular, electrophysiological, and theoretical modeling approaches, we shed light on the LC-NE system's complex role in the brain's adaptability and its potential relevance to neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Chun-Wang Su
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Fan Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Runchen Lai
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Yanhai Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Hadia Naeem
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Nan Yao
- Department of Applied Physics, Xi’an University of Technology, 710054 Shaanxi, China
| | - Si-Ping Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Haiqing Zhang
- Xi’an Children’s Hospital, Xi’an, 710003 Shaanxi China
| | - Youjun Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| | - Zi-Gang Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Institute of Health and Rehabilitation Science, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
- Research Center for Brain-Inspired Intelligence, Xi’an Jiaotong University, Xi’an, 710049 Shaanxi China
| |
Collapse
|
2
|
Li Z, Wu Y, Manyande A, Wu D, Xiang H. Odorgenetics with 2-pentanone: a novel cell manipulation technique. Med Gas Res 2025; 15:450-451. [PMID: 40072256 PMCID: PMC12054665 DOI: 10.4103/mgr.medgasres-d-25-00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 01/27/2025] [Accepted: 02/15/2025] [Indexed: 04/20/2025] Open
Affiliation(s)
- Zhixiao Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, Hubei Province, China
| | - Yanqiong Wu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Institute of Anesthesiology & Pain (IAP), Department of Anesthesiology, Taihe Hospital, College of Pharmacy, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, UK
| | - Duozhi Wu
- Department of Anesthesiology, Hainan General Hospital (Hainan Hospital Affiliated to Hainan Medical University), Haikou, Hainan Province, China
| | - Hongbing Xiang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
3
|
Valcarce-Aspegren M, Paszkowski P, Liu S, Wu Q, McGill S, Sieu LA, Blumenfeld H. Decreased locus coeruleus multiunit activity in a mouse model of temporal lobe seizures with impaired consciousness. Exp Neurol 2025; 389:115233. [PMID: 40189126 PMCID: PMC12083539 DOI: 10.1016/j.expneurol.2025.115233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 04/13/2025]
Abstract
People with temporal lobe epilepsy often suffer debilitating loss of consciousness during seizures. Rodent models have previously implicated the inhibition of brainstem and basal forebrain cholinergic neurons in the cortical impairment during these periods of impaired consciousness. However, there are still other subcortical pathways that remain largely unexplored. Our goal was to record multiunit activity in the locus coeruleus (LC) in an awake mouse model to help elucidate its potential role in this pathophysiology. Recordings were performed using head-fixed mice running on a wheel with chronically implanted bipolar electrodes in the right orbitofrontal cortex and bilateral dorsal hippocampi. Focal limbic seizures were induced via the application of current pulses into the HC and multiunit recordings were simultaneously obtained from the LC. We observed a significant decrease in firing of LC neurons during ictal impairment of running wheel behavior. There was also a concurrent, significant increase in power in the 1-4 Hz band in the OFC. This provides evidence of a LC noradrenergic pathway contributing to depressed arousal in focal limbic seizures. Further elucidation of these, and other pathways, will contribute better mechanistic understanding of ictal unconsciousness and may lead to novel, improved treatments for people with epilepsy.
Collapse
Affiliation(s)
- Marcus Valcarce-Aspegren
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| | - Patrick Paszkowski
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| | - Shixin Liu
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| | - Qian Wu
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Sarah McGill
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| | - Lim-Anna Sieu
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| | - Hal Blumenfeld
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
4
|
Torres AS, Robison MK, Brewer GA. The Role of the LC-NE System in Attention: From Cells, to Systems, to Sensory-Motor Control. Neurosci Biobehav Rev 2025:106233. [PMID: 40412462 DOI: 10.1016/j.neubiorev.2025.106233] [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: 03/07/2025] [Revised: 05/13/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Attention control is a fundamental cognitive function that enables individuals to sustain focus, shift attention flexibly, and filter distractions in a goal-directed manner. The locus coeruleus-norepinephrine (LC-NE) system plays a pivotal role in this process by dynamically regulating arousal, prioritizing salient stimuli, and optimizing cognitive performance. This review synthesizes evidence from molecular, cellular, systems, cognitive neuroscience, and behavioral studies to elucidate the LC-NE system's role in attention control. We first examine the neurophysiological mechanisms of the LC, highlighting its distinct firing patterns-tonic and phasic activity-and their impact on attention. Next, we integrate findings from animal models, human neuroimaging, electrophysiology, and computational modeling, demonstrating how LC-NE activity influences sensory processing, cognitive flexibility, and executive function. We interpret these findings through the lens of three major theoretical frameworks: Adaptive Gain Theory (AGT), which describes how LC activity optimizes task engagement, the Network Reset Hypothesis (NRH), which describes how optimizes network connectivity, and the Glutamate Amplifies NE Effects (GANE) model, which explains how NE enhances neural selectivity and suppresses irrelevant signals. Collectively, the evidence underscores the LC-NE system's role in modulating the signal-to-noise ratio in cortical and subcortical circuits, thereby shaping attention and behavior. We conclude by discussing implications for individual differences, age-related cognitive decline, and emphasizing the need for interdisciplinary research that integrates emerging technologies to further unravel the complexities of LC function.
Collapse
|
5
|
Gu BM, Kim JG, Hossain A, Cron GO, Lee JH. Substantia nigra modulates breathing rate via locus coeruleus. iScience 2025; 28:112423. [PMID: 40343266 PMCID: PMC12059658 DOI: 10.1016/j.isci.2025.112423] [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: 05/22/2023] [Revised: 02/04/2025] [Accepted: 04/09/2025] [Indexed: 05/11/2025] Open
Abstract
Breathing is an automatic and rhythmic act primarily controlled by hindbrain neural circuits, including the pre-Bötzinger complex. While the basal ganglia are known to regulate a wide range of behaviors through downstream projections, their role in breathing control remains unclear. In this study, we demonstrate that outputs from the substantia nigra (SN) can modulate breathing rate in a cell-type specific manner. Specifically, optogenetic activation of glutamic acid decarboxylase 2 (Gad2)-expressing neurons in the SN, but not parvalbumin (Pvalb)-expressing neurons, decreased breathing rate in lightly anesthetized mice. Importantly, this effect was mediated through the inhibition of neural activity in the locus coeruleus (LC), suggesting a relationship between the decrease in breathing rate and the baseline firing rate of LC neurons. These findings provide evidence that the basal ganglia play an important role in the control of breathing rate through modulation of LC neural activity.
Collapse
Affiliation(s)
- Bon-Mi Gu
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Jae Gon Kim
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Aronee Hossain
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Greg O. Cron
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Jin Hyung Lee
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
6
|
Luskin AT, Li L, Fu X, Martin MM, Barcomb K, Girven KS, Blackburn T, Wells BA, Thai ST, Li EM, Rana AN, Simon RC, Sun L, Gao L, Murry AD, Golden SA, Stuber GD, Ford CP, Gu L, Bruchas MR. Heterogeneous pericoerulear neurons tune arousal and exploratory behaviours. Nature 2025:10.1038/s41586-025-08952-w. [PMID: 40335695 DOI: 10.1038/s41586-025-08952-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 03/27/2025] [Indexed: 05/09/2025]
Abstract
As the primary source of noradrenaline in the brain, the locus coeruleus (LC) regulates arousal, avoidance and stress responses1,2. However, how local neuromodulatory inputs control LC function remains unresolved. Here we identify a population of transcriptionally, spatially and functionally diverse GABAergic (γ-aminobutyric acid-producing) neurons in the LC dendritic field that receive distant inputs and modulate modes of LC firing to control global arousal levels and arousal-related processing and behaviours. We define peri-LC anatomy using viral tracing and combine single-cell RNA sequencing with spatial transcriptomics to molecularly define both LC noradrenaline-producing and peri-LC cell types. We identify several neuronal cell types that underlie peri-LC functional diversity using a series of complementary neural circuit approaches in behaving mice. Our findings indicate that LC and peri-LC neurons are transcriptionally, functionally and anatomically heterogenous neuronal populations that modulate arousal and avoidance states. Defining the molecular, cellular and functional diversity of the LC and peri-LC provides a roadmap for understanding the neurobiological basis of arousal, motivation and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Andrew T Luskin
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
- Laboratory of Neural Dynamics and Cognition, The Rockefeller University, New York, NY, USA
| | - Li Li
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Xiaonan Fu
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Madison M Martin
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
| | - Kelsey Barcomb
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Kasey S Girven
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Taylor Blackburn
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Bailey A Wells
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Sarah T Thai
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Esther M Li
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Psychology, University of Washington, Seattle, WA, USA
| | - Akshay N Rana
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Rhiana C Simon
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
| | - Li Sun
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- TopoGene Inc., Seattle, WA, USA
| | - Lei Gao
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Alexandria D Murry
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Sam A Golden
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Garret D Stuber
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Liangcai Gu
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Michael R Bruchas
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA.
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA.
- Department of Pharmacology, University of Washington, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
7
|
Petersen N, McCann KE, Stavarache MA, Kim LY, Weinshenker D, Winder DG. Adenosine A 2A Receptors Link Astrocytic α 1-Adrenergic Signaling to Wake-Promoting Dopamine Neurons. Biol Psychiatry 2025; 97:915-928. [PMID: 39419462 PMCID: PMC11991893 DOI: 10.1016/j.biopsych.2024.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Sleep and arousal disorders are common, but the underlying physiology of wakefulness is not fully understood. The locus coeruleus promotes arousal via α1-adrenergic receptor (α1AR)-driven recruitment of wake-promoting dopamine neurons in the ventral periaqueductal gray (vPAGDA neurons). α1AR expression is enriched on vPAG astrocytes, and chemogenetic activation of astrocytic Gq signaling promotes wakefulness. Astrocytes can release extracellular gliotransmitters, such as ATP and adenosine, but the mechanism underlying how vPAG astrocytic α1ARs influence sleep/wake behavior and vPAGDA neuron physiology is unknown. METHODS In this study, we utilized genetic manipulations with ex vivo calcium imaging in vPAGDA neurons and astrocytes, patch-clamp electrophysiology, and behavioral experiments in mice to test our hypothesis that astrocytic α1ARs mediate noradrenergic modulation of wake-promoting vPAGDA neurons via adenosine signaling. RESULTS Activation of α1ARs with phenylephrine increased calcium transients in vPAGDA neurons and vPAG astrocytes and increased vPAGDA neuron excitability ex vivo. Chemogenetic Gq-DREADD (designer receptor exclusively activated by designer drugs) activation of vPAG astrocytes similarly increased vPAGDA neuron calcium activity and intrinsic excitability. Conversely, short hairpin RNA knockdown of vPAG astrocytic α1ARs reduced the excitatory effect of phenylephrine on vPAGDA neurons and blunted arousal during the wake phase. Pharmacological blockade of adenosine A2A receptors precluded the α1AR-induced increase in vPAGDA calcium activity and excitability in brain slices, as well as the wake-promoting effects of vPAG α1AR activation in vivo. CONCLUSIONS We have identified a crucial role for vPAG astrocytic α1ARs in sustaining arousal through heightened excitability and activity of vPAGDA neurons mediated by local A2A receptors.
Collapse
Affiliation(s)
- Nicholas Petersen
- Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, Tennessee; Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee
| | - Katharine E McCann
- Human Genetics, Emory University School of Medicine, Atlanta, Georgia; Neuroscience Undergraduate Program, School of Psychology, Georgia Institute of Technology, Atlanta, Georgia
| | | | - Lisa Y Kim
- School for Science and Math at Vanderbilt, Nashville, Tennessee
| | - David Weinshenker
- Human Genetics, Emory University School of Medicine, Atlanta, Georgia
| | - Danny G Winder
- Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, Tennessee; Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee; Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts.
| |
Collapse
|
8
|
Foustoukos G, Lüthi A. Monoaminergic signaling during mammalian NREM sleep - Recent insights and next-level questions. Curr Opin Neurobiol 2025; 92:103025. [PMID: 40267623 DOI: 10.1016/j.conb.2025.103025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 04/25/2025]
Abstract
Subcortical neuromodulatory activity in the mammalian brain enables flexible wake behaviors, which are essential for survival in an ever-changing external environment. With the suppression of such behaviors in sleep, this activity is, on average, much reduced. Recent discoveries, enabled by unprecedented technical advancements, challenge the long-standing view that monoaminergic systems-noradrenaline (NA), dopamine (DA), and serotonin (5-HT)-remain largely inactive during sleep. This review highlights recent technological and scientific progress in this field, summarizing evidence that monoaminergic signaling in the brain supplements sleep with essential wake-related functions. Stress and/or neuropsychiatric conditions negatively impact on monoaminergic signaling, which can lead to sleep disruptions. Furthermore, subcortical neuromodulatory systems are vulnerable to neurodegenerative pathologies, which implies them in sleep disruptions at early stages of disease. We propose that future research will be well-invested in elucidating the spatiotemporal organization, cellular mechanisms, and functional relevance of neuromodulatory dynamics across species, and in identifying the molecular and physiological processes that sustain their integrity throughout the lifespan.
Collapse
Affiliation(s)
- Georgios Foustoukos
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, CH, Switzerland.
| | - Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, CH, Switzerland.
| |
Collapse
|
9
|
Lee YT, Chang YH, Barquero C, Wu CS, Chao SP, Chen DYT, Chen JT, Cherng YG, Wang CA. Pupil and Eye Blink Response Abnormalities During Emotional Conflict Processing in Late-Life Depression. J Geriatr Psychiatry Neurol 2025:8919887251334999. [PMID: 40227571 DOI: 10.1177/08919887251334999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
IntroductionThis study aims to investigate the locus coeruleus-norepinephrine system (LC-NE) function in late-life depression (LLD) patients by examining task-evoked pupil dilation in the emotional face-word Stroop task, given the recently established coupling between task-evoked pupil dilation and LC-NE activation.Materials and MethodsUsing video-based eye-tracking and principal component analysis, we explored task-evoked pupil responses and eye blinks in LLD patients (N = 25) and older healthy controls (CTRL) (N = 29) to determine whether there were alterations in pupil responses and eye blinks in LLD compared to CTRL.ResultsLLD patients exhibited significantly different pupil and eye-blink behavior compared to CTRL, with dampened task-evoked pupil dilation associated with emotional congruency and valence processing mediated by the sympathetic system compared to CTRL. Eye-blink rates associated with emotional valence were also altered in LLD compared to CTRL Moreover, Geriatric Depression Scale-15 scores in LLD correlated with emotional congruency effects revealed by task-evoked pupil dilation.ConclusionThe findings demonstrate that LLD patients display altered pupil behavior compared to CTRL. These altered responses correlated with the severity of depressive symptoms, indicating their potential as objective biomarkers for use in large at-risk populations for LLD.
Collapse
Affiliation(s)
- Yao-Tung Lee
- Department of Psychiatry, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Psychiatry, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Graduate School of Criminology, National Taipei University, New Taipei City, Taiwan
| | - Yi-Hsuan Chang
- Eye-Tracking Laboratory, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Institute of Cognitive Neuroscience, College of Health Science and Technology, National Central University, Taoyuan City, Taiwan
| | - Cesar Barquero
- Department of Physical Activity and Sport Science, Universidad Peruana de Ciencias Aplicadas, Lima, Peru
| | - Chi-Shin Wu
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Miaoli, Taiwan
- Department of Psychiatry, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Shu-Ping Chao
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City, Taiwan
- Dementia Center and Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - David Yen-Ting Chen
- Department of Medical Image, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Jui-Tai Chen
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yih-Giun Cherng
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chin-An Wang
- Eye-Tracking Laboratory, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- College of Medical Science and Technology, Taipei Medical University, Taipei City, Taiwan
| |
Collapse
|
10
|
Deng Z, Fei X, Zhang S, Xu M. A time window for memory consolidation during NREM sleep revealed by cAMP oscillation. Neuron 2025:S0896-6273(25)00220-X. [PMID: 40233747 DOI: 10.1016/j.neuron.2025.03.020] [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: 08/22/2024] [Revised: 01/29/2025] [Accepted: 03/14/2025] [Indexed: 04/17/2025]
Abstract
Memory formation requires specific neural activity in coordination with intracellular signaling mediated by second messengers such as cyclic adenosine monophosphate (cAMP). However, the real-time dynamics of cAMP remain largely unknown. Here, using a genetically encoded cAMP sensor with high temporal resolution, we found neural-activity-dependent rapid cAMP elevation during learning. Interestingly, in slow-wave sleep, during which memory consolidation occurs, the cAMP level in mice was anti-correlated with neural activity and exhibited norepinephrine β1 receptor-dependent infra-slow oscillations that were synchronized across the hippocampus and cortex. Furthermore, the hippocampal-cortical interactions increased during the narrow time-window of the peak cAMP level; suppressing hippocampal activity specifically during this window impaired spatial memory consolidation. Thus, hippocampal-dependent memory consolidation occurs within a specific time window of high cAMP activity during slow-wave sleep.
Collapse
Affiliation(s)
- Ziru Deng
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiang Fei
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siyu Zhang
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Min Xu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
11
|
Reyes B. The Locus Coeruleus: Anatomy, Physiology, and Stress-Related Neuropsychiatric Disorders. Eur J Neurosci 2025; 61:e70111. [PMID: 40219735 PMCID: PMC11992612 DOI: 10.1111/ejn.70111] [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: 10/23/2024] [Revised: 03/18/2025] [Accepted: 03/29/2025] [Indexed: 04/14/2025]
Abstract
The locus coeruleus-norepinephrine (LC-NE) system is involved in mediating a wide array of functions, including attention, arousal, cognition, and stress response. Dysregulation of the LC-NE system is strongly linked with several stress-induced neuropsychiatric disorders, highlighting the LC's pivotal role in the development of these disorders. Located in the dorsal pontine tegmental area, the LC contains noradrenergic neurons that serve as the main source of NE in the central nervous system. Activation of the LC and subsequent release of NE at different levels of the neuroaxis is adaptive, allowing the body to adjust appropriately amid a challenging stimulus. However, prolonged and repeated LC activation leads to maladaptive responses that implicate LC-NE dysfunction in stress-induced neuropsychiatric disorders. As the primary initiator of the stress response, corticotropin-releasing factor (CRF) activates the hypothalamic-pituitary-adrenal axis. Following the discovery of CRF more than four decades ago, numerous studies established that CRF also acts as a neurotransmitter that governs the activity of other neurotransmitters in the brain neurotransmitter system. The LC-NE system receives abundant CRF afferents arising from several brain nuclei. CRF afferents to LC-NE are activated and recruited in the pathogenesis of stress-induced neuropsychiatric disorders. Presented in this review are the CRF neuroanatomical connectivity and physiological characteristics that modulate LC-NE function, which may contribute to the pathogenesis of stress-induced neuropsychiatric disorders. Additionally, this review illustrates the contribution of LC-NE to the apparent sex-dependent differences in stress-induced neuropsychiatric disorders. Hence, the LC-NE system is a promising target for the development of therapeutic strategies for stress-induced neuropsychiatric disorders.
Collapse
Affiliation(s)
- Beverly A. S. Reyes
- Department of Pharmacology & PhysiologyDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| |
Collapse
|
12
|
Moreira TS, Takakura AC, Falquetto B, Ramirez JM, Oliveira LM, Silva PE, Araujo EV. Neuroanatomical and neurochemical organization of brainstem and forebrain circuits involved in breathing regulation. J Neurophysiol 2025; 133:1116-1137. [PMID: 40059616 DOI: 10.1152/jn.00475.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/19/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Breathing regulation depends on a highly intricate and precise network within the brainstem, requiring the identification of all neuronal elements in the brainstem respiratory circuits and a comprehensive understanding of their organization into distinct functional compartments. These compartments play a pivotal role by providing essential input to three main targets: cranial motoneurons that regulate airway control, spinal motoneurons that activate the inspiratory and expiratory muscles, and higher brain structures that influence breathing behavior and integrate it with other physiological and behavioral processes. This review offers a comprehensive examination of the phenotypes, connections, and functional roles of the major compartments within the brainstem and forebrain respiratory circuits. In addition, it summarizes the diverse neurotransmitters used by neurons in these regions, highlighting their contributions to the coordination and modulation of respiratory activity.
Collapse
Affiliation(s)
- Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Barbara Falquetto
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, United States
- Department of Neurological Surgery, University of Washington, Seattle, Washington, United States
| | - Luiz M Oliveira
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, United States
| | - Phelipe E Silva
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Emmanuel V Araujo
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
13
|
Zhou W, Chu HY. Progressive noradrenergic degeneration and motor cortical dysfunction in Parkinson's disease. Acta Pharmacol Sin 2025; 46:829-835. [PMID: 39592731 PMCID: PMC11950218 DOI: 10.1038/s41401-024-01428-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024]
Abstract
The locus coeruleus norepinephrine (LC-NE) system plays an important role in regulating brain function, and its neuronal loss has been well-documented in Parkinson's disease (PD). The LC-NE neurodegeneration is believed to underlie various nonmotor symptoms in people with PD, including neuropsychiatric deficits, sleep disruptions, and cognitive impairments. Of particular interest, LC-NE neurons send intensive axonal projections to the motor regions of the cerebral cortex. However, how NE depletion in the motor cortex contributes to PD pathophysiology remains poorly understood. In addition, recent studies provided increasing mechanistic insights into secondary changes in the cerebral cortex as LC-NE degenerates, which might involve its interaction with dopaminergic signaling during the chronic course of the disease. In the present article, we briefly discuss clinical and preclinical studies that support the critical roles of LC-NE neurodegeneration and motor cortical dysfunction in both motor and nonmotor deficits in Parkinsonian states. We focus our discussion on the potential impact of LC-NE neurodegeneration on motor cortical function and the subsequent symptom manifestation. Last, we propose future research directions that can advance our understanding of cortical pathophysiology in PD by integrating noradrenergic degeneration.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington D.C., WA, 20007, USA
| | - Hong-Yuan Chu
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington D.C., WA, 20007, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, USA.
| |
Collapse
|
14
|
Hu DD, Shi W, Jia X, Shao FM, Zhang L. Alpha-2 receptor mediates the endogenous antagonistic regulation of itch and pain via descending noradrenaline pathway from the locus coeruleus. Brain Res Bull 2025; 223:111270. [PMID: 39999937 DOI: 10.1016/j.brainresbull.2025.111270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/07/2025] [Accepted: 02/23/2025] [Indexed: 02/27/2025]
Abstract
Pain and itch are sensations that are regulated antagonistically; painful stimulation suppresses itch, while the inhibition of pain enhances itch. However, the central neural circuit underlying this antagonistic regulation remains elusive. The noradrenaline (NA) pathway from the locus coeruleus (LC) to the spinal cord (SC) constitutes an important component of endogenous descending pain inhibitory system. While the pathway of LC:SC has been extensively studied on pain modulation, its role in itch regulation remains poorly understood. We employed behavioral assays for itch and pain, immunofluorescence, electrophysiology, and chemogenetic techniques to investigate the role of noradrenergic (NAergic) neurons of LC (LCNA neurons)and their pathways in modulating itch and pain. Our study has demonstrated that LCNA neurons encode signals for both itch and pain. Inhibition of LCNA neurons had no effect on itch but enhanced pain behaviour. Surprisingly, inhibition of the NAergic projection of LC:SC increased pain and suppressed itch. Furthermore, intrathecal injection of an α2 adrenergic receptor antagonist, but not α1 or β receptor antagonists, produced effects similar to those observed when the LC:SC pathway was inhibited. Our research suggests that the descending NAergic pathway from LC to SC exerts endogenous antagonistic regulation on itch and pain through α2 receptors.
Collapse
Affiliation(s)
- Dan-Dan Hu
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai 201619, China; Clinical Center For Brain And Spinal Cord Research, Tongji University, Shanghai 200331, China
| | - Wu Shi
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai 201619, China; Clinical Center For Brain And Spinal Cord Research, Tongji University, Shanghai 200331, China
| | - Xin Jia
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai 201619, China; Clinical Center For Brain And Spinal Cord Research, Tongji University, Shanghai 200331, China
| | - Fu-Ming Shao
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai 201619, China; Clinical Center For Brain And Spinal Cord Research, Tongji University, Shanghai 200331, China
| | - Ling Zhang
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai 201619, China; Clinical Center For Brain And Spinal Cord Research, Tongji University, Shanghai 200331, China.
| |
Collapse
|
15
|
Parhizkar S, Holtzman DM. The night's watch: Exploring how sleep protects against neurodegeneration. Neuron 2025; 113:817-837. [PMID: 40054454 PMCID: PMC11925672 DOI: 10.1016/j.neuron.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/15/2024] [Accepted: 02/04/2025] [Indexed: 03/21/2025]
Abstract
Sleep loss is often regarded as an early manifestation of neurodegenerative diseases given its common occurrence and link to cognitive dysfunction. However, the precise mechanisms by which sleep disturbances contribute to neurodegeneration are not fully understood, nor is it clear why some individuals are more susceptible to these effects than others. This review addresses critical unanswered questions in the field, including whether sleep disturbances precede or result from neurodegenerative diseases, the functional significance of sleep changes during the preclinical disease phase, and the potential role of sleep homeostasis as an adaptive mechanism enhancing resilience against cognitive decline and neurodegeneration.
Collapse
Affiliation(s)
- Samira Parhizkar
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO 63110, USA.
| |
Collapse
|
16
|
Li N, Huang L, Zhang B, Zhu W, Dai W, Li S, Xu H. The mechanism of different orexin/hypocretin neuronal projections in wakefulness and sleep. Brain Res 2025; 1850:149408. [PMID: 39706239 DOI: 10.1016/j.brainres.2024.149408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Since the discovery of orexin/hypocretin, numerous studies have accumulated evidence demonstrating its key role in various aspects of neuromodulation, including addiction, motivation, and arousal. This paper focuses on the projection of orexin neurons to specific target brain regions through distinct neural pathways to regulate sleep and arousal. We provide a detailed discussion of the projection mechanisms of orexin neurons to downstream neurons, particularly emphasizing their activation of monoaminergic and cholinergic neurons associated with arousal. Additionally, we briefly explore the immune response and inflammatory factors linked to the loss of orexin neurons. Our findings underscore the significance of understanding specific neural projections in the generation and maintenance of arousal, which could guide advancements in neuroscience and lead to new therapeutic opportunities for treating insomnia or narcolepsy.
Collapse
Affiliation(s)
- Nanxi Li
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Lishan Huang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Bin Zhang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Wenwen Zhu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Wenbin Dai
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Sen Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University.
| | - Houping Xu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
17
|
Yu H, Wang J, Pang R, Chen P, Luo T, Zhang X, Liao Y, Hu C, Gu M, Luo B, Shi Z, Li M, Zhang Y, Wei Q, Yuan W, Xie H, Chen Z, Liu H, Ren S, Chen X, Zhou Y. Temporal Association Cortex Gates Sound-Evoked Arousal from NREM Sleep. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414271. [PMID: 39887927 PMCID: PMC11948000 DOI: 10.1002/advs.202414271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/27/2024] [Indexed: 02/01/2025]
Abstract
Sound-evoked wakefulness from sleep is crucial in daily life, yet its neural mechanisms remain poorly understood. It is found that CaMKIIα+ neurons in the temporal association cortex (TeA) of mice are not essential for natural awakening from sleep. However, optogenetic activation of these neurons reliably induces wakefulness from non-rapid eye movement (NREM) sleep but not from rapid eye movement (REM) sleep. In vivo electrophysiological and calcium recordings further demonstrated that TeA neurons are monotonically tuned to sound intensity but not frequency. More importantly, it is found that the activity of CaMKIIα+ neurons in TeA can gate sound-evoked arousal from NREM sleep, which is further confirmed by optogenetic manipulations. Further investigation reveals that the baseline excitability of TeA CaMKIIα+ neurons and the delta oscillations in the electroencephalogram are particularly important in regulating the evoked activity of TeA neurons. Anatomical and functional screening of downstream targets of TeA reveals that excitatory projections from TeA glutamatergic neurons to glutamatergic neurons in the basolateral/lateral amygdala are critical for modulating sound-evoked arousal from NREM sleep. These findings uncover a top-down regulatory circuit that selectively governs sound-evoked arousal from NREM sleep, with the TeA functioning as a key connecting cortex to subcortical regions.
Collapse
Affiliation(s)
- Haipeng Yu
- Advanced Institute for Brain and IntelligenceSchool of Physical Science and TechnologyGuangxi UniversityNanning530004China
- Department of NeurobiologyCollege of Basic MedicineArmy Medical UniversityChongqing400038China
| | - Jincheng Wang
- Department of NeurobiologyCollege of Basic MedicineArmy Medical UniversityChongqing400038China
| | - Ruiqi Pang
- Guangxi Key Laboratory of Special BiomedicineSchool of MedicineGuangxi UniversityNanning530004China
| | - Penghui Chen
- Department of NeurobiologyCollege of Basic MedicineArmy Medical UniversityChongqing400038China
| | - Tiantian Luo
- Department of NeurobiologyCollege of Basic MedicineArmy Medical UniversityChongqing400038China
| | - Xuan Zhang
- Department of NeurobiologyCollege of Basic MedicineArmy Medical UniversityChongqing400038China
| | - Yatao Liao
- Department of NeurobiologyCollege of Basic MedicineArmy Medical UniversityChongqing400038China
| | - Chao Hu
- Department of NeurobiologyCollege of Basic MedicineArmy Medical UniversityChongqing400038China
| | - Miaoqing Gu
- Advanced Institute for Brain and IntelligenceSchool of Physical Science and TechnologyGuangxi UniversityNanning530004China
| | - Bingmin Luo
- Department of NeurosciencesCase Western Reserve University School of MedicineClevelandOH44106USA
| | - Zhiyue Shi
- Department of NeurobiologyCollege of Basic MedicineArmy Medical UniversityChongqing400038China
| | - Mengyao Li
- Guangxi Key Laboratory of Special BiomedicineSchool of MedicineGuangxi UniversityNanning530004China
| | - Yueting Zhang
- Guangxi Key Laboratory of Special BiomedicineSchool of MedicineGuangxi UniversityNanning530004China
| | - Qiaoqian Wei
- Guangxi Key Laboratory of Special BiomedicineSchool of MedicineGuangxi UniversityNanning530004China
| | - Wei Yuan
- Department of OtolaryngologyChongqing General HospitalChongqing UniversityChongqing400038China
| | - Hui Xie
- School of Architecture and Urban PlanningChongqing UniversityChongqing400044China
| | - Zhiyi Chen
- Experimental Research Center for Medical and Psychological ScienceSchool of PsychologyArmy Medical UniversityChongqing400038China
| | - Hongbang Liu
- Advanced Institute for Brain and IntelligenceSchool of Physical Science and TechnologyGuangxi UniversityNanning530004China
| | - Shuancheng Ren
- Department of PhysiologyCollege of Basic MedicineArmy Medical UniversityChongqing400038China
| | - Xiaowei Chen
- Brain Research Center and State Key Laboratory of Trauma and Chemical PoisoningCollege of Basic MedicineArmy Medical UniversityChongqing400038China
| | - Yi Zhou
- Department of NeurobiologyCollege of Basic MedicineArmy Medical UniversityChongqing400038China
| |
Collapse
|
18
|
Basgol H, Dayan P, Franz VH. Violation of auditory regularities is reflected in pupil dynamics. Cortex 2025; 183:66-86. [PMID: 39616966 DOI: 10.1016/j.cortex.2024.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 09/16/2024] [Accepted: 10/25/2024] [Indexed: 02/21/2025]
Abstract
The brain builds and maintains internal models and uses them to make predictions. When predictions are violated, the current model can either be updated or replaced by a new model. The latter is accompanied by pupil dilation responses (PDRs) related to locus coeruleus activity/norepinephrine release (LC-NE). Following earlier research, we investigated PDRs associated with transitions between regular and random patterns of tones in auditory sequences. We presented these sequences to participants and instructed them to find gaps (to maintain attention). Transitions from regular to random patterns induced PDRs, suggesting that an internal model attuned to the regular pattern is reset. Transitions from one regular pattern to another regular pattern also induced PDRs, suggesting that they also led to a model reset. In contrast, transitions from random patterns to regular patterns did not induce PDRs, suggesting a gradual update of model parameters. We modelled these findings, using pupil response functions to show how ongoing PDRs and pupil event rates were sensitive to the trial-by-trial changes in the information content of the auditory sequences. Expanding on previous research, we suggest that PDRs-as biomarkers for LC-NE activation-may indicate the extent of prediction violations.
Collapse
Affiliation(s)
- Hamit Basgol
- Department of Computer Science, University of Tübingen, Tübingen, Germany; Experimental Cognitive Science, University of Tübingen, Tübingen, Germany; The Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, Germany.
| | - Peter Dayan
- Department of Computer Science, University of Tübingen, Tübingen, Germany; Max Planck Institute for Biological Cybernetics, Tübingen, Germany.
| | - Volker H Franz
- Department of Computer Science, University of Tübingen, Tübingen, Germany; Experimental Cognitive Science, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
19
|
Lee YT, Tai YH, Chang YH, Barquero C, Chao SP, Wang CA. Disrupted microsaccade responses in late-life depression. Sci Rep 2025; 15:2827. [PMID: 39843485 PMCID: PMC11754810 DOI: 10.1038/s41598-025-86399-9] [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: 08/03/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025] Open
Abstract
Late-life depression (LLD) is a psychiatric disorder in older adults, characterized by high prevalence and significant mortality rates. Thus, it is imperative to develop objective and cost-effective methods for detecting LLD. Individuals with depression often exhibit disrupted levels of arousal, and microsaccades, as a type of fixational eye movement that can be measured non-invasively, are known to be modulated by arousal. This makes microsaccades a promising candidate as biomarkers for LLD. In this study, we used a high-resolution, video-based eye-tracker to examine microsaccade behavior in a visual fixation task between LLD patients and age-matched healthy controls (CTRL). Our goal was to determine whether microsaccade responses are disrupted in LLD compared to CTRL. LLD patients exhibited significantly higher microsaccade peak velocities and larger amplitudes compared to CTRL. Although microsaccade rates were lower in LLD than in CTRL, these differences were not statistically significant. Additionally, while both groups displayed microsaccadic inhibition and rebound in response to changes in background luminance, this modulation was significantly blunted in LLD patients, suggesting dysfunction in the neural circuits responsible for microsaccade generation. Together, these findings, for the first time, demonstrate significant alterations in microsaccade behavior in LLD patients compared to CTRL, highlighting the potential of these disrupted responses as behavioral biomarkers for identifying individuals at risk for LLD.
Collapse
Affiliation(s)
- Yao-Tung Lee
- Department of Psychiatry, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Psychiatry, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Ying-Hsuan Tai
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yi-Hsuan Chang
- Eye-Tracking Laboratory, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Institute of Cognitive Neuroscience, College of Health Science and Technology, National Central University, Taoyuan City, Taiwan
| | - Cesar Barquero
- Department of Physical Activity and Sport Science, Peruvian University of Applied Sciences, Lima, Peru
| | - Shu-Ping Chao
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City, Taiwan
- Dementia Center, Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chin-An Wang
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan.
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Eye-Tracking Laboratory, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|
20
|
Silverman D, Chen C, Chang S, Bui L, Zhang Y, Raghavan R, Jiang A, Le A, Darmohray D, Sima J, Ding X, Li B, Ma C, Dan Y. Activation of locus coeruleus noradrenergic neurons rapidly drives homeostatic sleep pressure. SCIENCE ADVANCES 2025; 11:eadq0651. [PMID: 39823324 PMCID: PMC11740930 DOI: 10.1126/sciadv.adq0651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025]
Abstract
Homeostatic sleep regulation is essential for optimizing the amount and timing of sleep for its revitalizing function, but the mechanism underlying sleep homeostasis remains poorly understood. Here, we show that optogenetic activation of locus coeruleus (LC) noradrenergic neurons immediately increased sleep propensity following a transient wakefulness, contrasting with many other arousal-promoting neurons whose activation induces sustained wakefulness. Fiber photometry showed that repeated optogenetic or sensory stimulation caused a rapid reduction of calcium activity in LC neurons and steep declines in noradrenaline/norepinephrine (NE) release in both the LC and medial prefrontal cortex (mPFC). Knockdown of α2A adrenergic receptors in LC neurons mitigated the decline of NE release induced by repetitive stimulation and extended wakefulness, demonstrating an important role of α2A receptor-mediated auto-suppression of NE release. Together, these results suggest that functional fatigue of LC noradrenergic neurons, which reduces their wake-promoting capacity, contributes to sleep pressure.
Collapse
Affiliation(s)
- Daniel Silverman
- Department of Neuroscience, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Changwan Chen
- Department of Neuroscience, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Shuang Chang
- Department of Neuroscience, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | | | - Rishi Raghavan
- Department of Neuroscience, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Anna Jiang
- Department of Neuroscience, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - April Le
- Department of Neuroscience, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Dana Darmohray
- Department of Neuroscience, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jiao Sima
- Department of Neuroscience, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Xinlu Ding
- Department of Neuroscience, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | | | - Yang Dan
- Department of Neuroscience, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
21
|
Rae RJ, Baker NL, Irwin ZT, Shea SD, McMahon LL. Diurnal Modulation of Locus Coeruleus Noradrenergic Neurons in Anesthetized Adult Male Rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631545. [PMID: 39829806 PMCID: PMC11741345 DOI: 10.1101/2025.01.06.631545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The locus coeruleus (LC) is the primary source of noradrenaline (NA) in brain and its activity is essential for learning, memory, stress, arousal, and mood. LC-NA neuron activity varies over the sleep-wake cycle, with higher activity during wakefulness, correlating with increased CSF NA levels. Whether spontaneous and burst firing of LC-NA neurons during active and inactive periods is controlled by mechanisms independent of wakefulness and natural sleep, is currently unknown. Here, using multichannel in vivo electrophysiology under anesthesia, we assessed LC-NA neuron firing in adult male Fisher 344 rats at two different times of day- ZT4- the inactive period (light phase) and ZT16-the active period (dark phase)- independent of contributions from behavioral arousal and natural sleep. In the dark phase, LC-NA neurons exhibit increased average firing rate during baseline compared to the light phase. Using a relatively weak foot shock paradigm, we observed distinct populations of LC-NA neurons with some increasing, and others decreasing, their firing rate compared to baseline. Additionally, while spike frequency during spontaneous and evoked bursts is consistent across the dark-light phase, units recorded during the dark phase have more frequent bursts with a longer duration than those during the light phase. Our findings show that independent of wake state, LC-NA neurons exhibit intrinsic diurnal activity, and that the variability of response to foot shock stimulation demonstrates a physiological heterogeneity of LC-NA neurons that is just beginning to be appreciated. NEW & NOTEWORTHY Multichannel in vivo electrophysiology assesses activity of large populations of NA neurons within an intact LC. Recording activity under anesthesia eliminates influence of behavior and sleep on LC-NA neuron physiology. Our data show that LC-NA neurons have heightened firing and burst activity during the dark phase, suggesting a hardwired diurnal rhythm. Additionally, LC-NA neurons have variable evoked firing highlighting heterogeneity, consistent with a contemporary view that LC physiology is more complex than previously appreciated.
Collapse
|
22
|
Chang YH, Yep R, Wang CA. Pupil size correlates with heart rate, skin conductance, pulse wave amplitude, and respiration responses during emotional conflict and valence processing. Psychophysiology 2025; 62:e14726. [PMID: 39533166 DOI: 10.1111/psyp.14726] [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: 05/29/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Pupil size is a non-invasive index for autonomic arousal mediated by the locus coeruleus-norepinephrine (LC-NE) system. While pupil size and its derivative (velocity) are increasingly used as indicators of arousal, limited research has investigated the relationships between pupil size and other well-known autonomic responses. Here, we simultaneously recorded pupillometry, heart rate, skin conductance, pulse wave amplitude, and respiration signals during an emotional face-word Stroop task, in which task-evoked (phasic) pupil dilation correlates with LC-NE responsivity. We hypothesized that emotional conflict and valence would affect pupil and other autonomic responses, and trial-by-trial correlations between pupil and other autonomic responses would be observed during both tonic and phasic epochs. Larger pupil dilations, higher pupil size derivative, and lower heart rates were observed in the incongruent condition compared to the congruent condition. Additionally, following incongruent trials, the congruency effect was reduced, and arousal levels indexed by previous-trial pupil dilation were correlated with subsequent reaction times. Furthermore, linear mixed models revealed that larger pupil dilations correlated with higher heart rates, higher skin conductance responses, higher respiration amplitudes, and lower pulse wave amplitudes on a trial-by-trial basis. Similar effects were seen between positive and negative valence conditions. Moreover, tonic pupil size before stimulus presentation significantly correlated with all other tonic autonomic responses, whereas tonic pupil size derivative correlated with heart rates and skin conductance responses. These results demonstrate a trial-by-trial relationship between pupil dynamics and other autonomic responses, highlighting pupil size as an effective real-time index for autonomic arousal during emotional conflict and valence processing.
Collapse
Affiliation(s)
- Yi-Hsuan Chang
- Eye-Tracking Laboratory, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Institute of Cognitive Neuroscience, National Central University, Taoyuan City, Taiwan
| | - Rachel Yep
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Chin-An Wang
- Eye-Tracking Laboratory, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
23
|
Osorio-Forero A, Foustoukos G, Cardis R, Cherrad N, Devenoges C, Fernandez LMJ, Lüthi A. Infraslow noradrenergic locus coeruleus activity fluctuations are gatekeepers of the NREM-REM sleep cycle. Nat Neurosci 2025; 28:84-96. [PMID: 39587312 DOI: 10.1038/s41593-024-01822-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/15/2024] [Indexed: 11/27/2024]
Abstract
The noradrenergic locus coeruleus (LC) regulates arousal levels during wakefulness, but its role in sleep remains unclear. Here, we show in mice that fluctuating LC neuronal activity partitions non-rapid-eye-movement sleep (NREMS) into two brain-autonomic states that govern the NREMS-REMS cycle over ~50-s periods; high LC activity induces a subcortical-autonomic arousal state that facilitates cortical microarousals, whereas low LC activity is required for NREMS-to-REMS transitions. This functional alternation regulates the duration of the NREMS-REMS cycle by setting permissive windows for REMS entries during undisturbed sleep while limiting these entries to maximally one per ~50-s period during REMS restriction. A stimulus-enriched, stress-promoting wakefulness was associated with longer and shorter levels of high and low LC activity, respectively, during subsequent NREMS, resulting in more microarousal-induced NREMS fragmentation and delayed REMS onset. We conclude that LC activity fluctuations are gatekeepers of the NREMS-REMS cycle and that this role is influenced by adverse wake experiences.
Collapse
Affiliation(s)
- Alejandro Osorio-Forero
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Georgios Foustoukos
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Romain Cardis
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Najma Cherrad
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Christiane Devenoges
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Laura M J Fernandez
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
24
|
Kosciessa JQ, Mayr U, Lindenberger U, Garrett DD. Broadscale dampening of uncertainty adjustment in the aging brain. Nat Commun 2024; 15:10717. [PMID: 39715747 PMCID: PMC11666723 DOI: 10.1038/s41467-024-55416-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/10/2024] [Indexed: 12/25/2024] Open
Abstract
The ability to prioritize among input features according to relevance enables adaptive behaviors across the human lifespan. However, relevance often remains ambiguous, and such uncertainty increases demands for dynamic control. While both cognitive stability and flexibility decline during healthy ageing, it is unknown whether aging alters how uncertainty impacts perception and decision-making, and if so, via which neural mechanisms. Here, we assess uncertainty adjustment across the adult lifespan (N = 100; cross-sectional) via behavioral modeling and a theoretically informed set of EEG-, fMRI-, and pupil-based signatures. On the group level, older adults show a broad dampening of uncertainty adjustment relative to younger adults. At the individual level, older individuals whose modulation more closely resembled that of younger adults also exhibit better maintenance of cognitive control. Our results highlight neural mechanisms whose maintenance plausibly enables flexible task-set, perception, and decision computations across the adult lifespan.
Collapse
Affiliation(s)
- Julian Q Kosciessa
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Berlin, Germany.
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany.
- Radboud University, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.
| | - Ulrich Mayr
- Department of Psychology, University of Oregon, Eugene, OR, USA
| | - Ulman Lindenberger
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Berlin, Germany
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | - Douglas D Garrett
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Berlin, Germany.
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany.
| |
Collapse
|
25
|
Choi A, Kim B, Labriola E, Wiest A, Wang Y, Smith J, Shin H, Jin X, An I, Hong J, Antila H, Thomas S, Bhattarai JP, Beier K, Ma M, Weber F, Chung S. Circuit mechanism underlying fragmented sleep and memory deficits in 16p11.2 deletion mouse model of autism. iScience 2024; 27:111285. [PMID: 39628570 PMCID: PMC11612818 DOI: 10.1016/j.isci.2024.111285] [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: 05/29/2024] [Revised: 08/26/2024] [Accepted: 10/25/2024] [Indexed: 12/06/2024] Open
Abstract
Sleep disturbances are prevalent in children with autism spectrum disorder (ASD). Strikingly, sleep problems are positively correlated with the severity of ASD symptoms, such as memory impairment. However, the neural mechanisms underlying sleep disturbances and cognitive deficits in ASD are largely unexplored. Here, we show that non-rapid eye movement sleep (NREMs) is fragmented in the 16p11.2 deletion mouse model of ASD. The degree of sleep fragmentation is reflected in an increased number of calcium transients in the activity of locus coeruleus noradrenergic (LC-NE) neurons during NREMs. In contrast, optogenetic inhibition of LC-NE neurons and pharmacological blockade of noradrenergic transmission using clonidine consolidate sleep. Furthermore, inhibiting LC-NE neurons restores memory. Finally, rabies-mediated screening of presynaptic neurons reveals altered connectivity of LC-NE neurons with sleep- and memory-regulatory regions in 16p11.2 deletion mice. Our findings identify a crucial role of the LC-NE system in regulating sleep stability and memory in ASD.
Collapse
Affiliation(s)
- Ashley Choi
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bowon Kim
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eleanor Labriola
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alyssa Wiest
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yingqi Wang
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jennifer Smith
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hyunsoo Shin
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xi Jin
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Isabella An
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jiso Hong
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hanna Antila
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven Thomas
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Janardhan P. Bhattarai
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kevin Beier
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA 92617, USA
| | - Minghong Ma
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Franz Weber
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shinjae Chung
- Department of Neuroscience, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
26
|
Dai Y, Shi K, Liu Q, Shen C, Lu X, Qiu X, Sun J. Intraoperative Sleep Spindle Activity and Postoperative Sleep Disturbance in Elderly Patients Undergoing Orthopedic Surgery: A Prospective Cohort Study. Nat Sci Sleep 2024; 16:2083-2097. [PMID: 39712881 PMCID: PMC11662682 DOI: 10.2147/nss.s486625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 11/26/2024] [Indexed: 12/24/2024] Open
Abstract
Purpose This study aimed to investigate the relationship between intraoperative sleep spindle activity and postoperative sleep disturbance (PSD) in elderly orthopedic surgery patients. Patients and Methods In this prospective observational cohort study, we collected intraoperative electroencephalography (EEG) data from 212 elderly patients undergoing orthopedic surgery from May 2023 to December 2023. We used the Athens Insomnia Scale to assess sleep quality on postoperative day (POD) 1 and POD 3 and analyzed the correlation between intraoperative sleep spindle activity and PSD through logistic regression. Results The incidence of PSD was 65.6% on POD 1 and 41.9% on POD 3. On the first day, there were no significant differences in intraoperative sleep spindle characteristics between PSD and non-postoperative sleep disturbance (non-PSD) patients. However, by the third day, PSD patients showed lower sigma power compared to non-PSD patients, as well as lower spindle density in the bilateral frontopolar (Fp1/Fp2) and bilateral temporal (F7/F8) channels, with shorter average spindle duration (P < 0.05). Multivariate logistic regression analysis suggested that the average spindle density in F7/F8 channels (OR 0.543, 95% CI 0.375-0.786; P = 0.001) was an independent risk factor for PSD on POD 3. Furthermore, Mini-Mental State Examination (MMSE) could independently predict PSD on POD 1 (OR 0.806, 95% CI 0.656-0.991; P = 0.041) and POD 3 (OR 0.701, 95% CI 0.562-0.875; P = 0.002). Pain on movement and at rest were independently associated with PSD on POD 1 (OR 1.480, 95% CI 1.200-1.824; P < 0.001) and POD 3 (OR 1.848, 95% CI 1.166-2.927; P = 0.009), respectively. Conclusion Intraoperative mean spindle density in the F7/F8 channels was an independent risk factor for PSD on POD 3 in elderly patients undergoing orthopedic surgery. MMSE and postoperative pain also independently increased the risk of PSD.
Collapse
Affiliation(s)
- Yuchen Dai
- Department of Anesthesiology & Key Laboratory of Clinical Science and Research, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Kaikai Shi
- Department of Anesthesiology & Key Laboratory of Clinical Science and Research, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Qingren Liu
- Department of Anesthesiology, Xishan People’s Hospital of Wuxi City, Wuxi, Jiangsu, People’s Republic of China
| | - Changli Shen
- Department of Anesthesiology, Xinxiang Central Hospital, Xinxiang, Henan, People’s Republic of China
| | - Xinjian Lu
- Department of Anesthesiology & Key Laboratory of Clinical Science and Research, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Xiaodong Qiu
- Department of Anesthesiology & Key Laboratory of Clinical Science and Research, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| | - Jie Sun
- Department of Anesthesiology & Key Laboratory of Clinical Science and Research, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
27
|
Hiraga T, Hata T, Soya S, Shimoda R, Takahashi K, Soya M, Inoue K, Johansen JP, Okamoto M, Soya H. Light-exercise-induced dopaminergic and noradrenergic stimulation in the dorsal hippocampus: Using a rat physiological exercise model. FASEB J 2024; 38:e70215. [PMID: 39668509 PMCID: PMC11638517 DOI: 10.1096/fj.202400418rrr] [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: 02/25/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024]
Abstract
Exercise activates the dorsal hippocampus that triggers synaptic and cellar plasticity and ultimately promotes memory formation. For decades, these benefits have been explored using demanding and stress-response-inducing exercise at moderate-to-vigorous intensities. In contrast, our translational research with animals and humans has focused on light-intensity exercise (light exercise) below the lactate threshold (LT), which almost anyone can safely perform with minimal stress. We found that even light exercise can stimulate hippocampal activity and enhance memory performance. Although the circuit mechanism of this boost remains unclear, arousal promotion even with light exercise implies the involvement of the ascending monoaminergic system that is essential to modulate hippocampal activity and impact memory. To test this hypothesis, we employed our physiological exercise model based on the LT of rats and immunohistochemically assessed the neuronal activation of the dorsal hippocampal sub-regions and brainstem monoaminergic neurons. Also, we monitored the extracellular concentration of monoamines in the dorsal hippocampus using in vivo microdialysis. We found that even light exercise increased neuronal activity in the dorsal hippocampal sub-regions and elevated the extracellular concentrations of noradrenaline and dopamine. Furthermore, we found that tyrosine hydroxylase-positive neurons in the locus coeruleus (LC) and the ventral tegmental area (VTA) were activated even by light exercise and were both positively correlated with the dorsal hippocampal activation. In conclusion, our findings demonstrate that light exercise stimulates dorsal hippocampal neurons, which are associated with LC-noradrenergic and VTA-dopaminergic activation. This shed light on the circuit mechanisms responsible for hippocampal neural activation during exercise, consequently enhancing memory function.
Collapse
Affiliation(s)
- Taichi Hiraga
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Toshiaki Hata
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Shingo Soya
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaJapan
- Department of Molecular Behavioral Physiology, Institute of MedicineUniversity of TsukubaTsukubaJapan
| | - Ryo Shimoda
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Kanako Takahashi
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Mariko Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Department of Anatomy and Neuroscience, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Koshiro Inoue
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Center for Education in Liberal Arts and SciencesHealth Sciences University of HokkaidoIshikariJapan
| | - Joshua P. Johansen
- Laboratory for Neural Circuitry of MemoryRIKEN Center for Brain ScienceSaitamaJapan
| | - Masahiro Okamoto
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Hideaki Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| |
Collapse
|
28
|
Do AD, Portet C, Goutagny R, Jackson J. The claustrum and synchronized brain states. Trends Neurosci 2024; 47:1028-1040. [PMID: 39488479 DOI: 10.1016/j.tins.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/25/2024] [Accepted: 10/09/2024] [Indexed: 11/04/2024]
Abstract
Cortical activity is constantly fluctuating between distinct spatiotemporal activity patterns denoted by changes in brain state. States of cortical desynchronization arise during motor generation, increased attention, and high cognitive load. Synchronized brain states comprise spatially widespread, coordinated low-frequency neural activity during rest and sleep when disengaged from the external environment or 'offline'. The claustrum is a small subcortical structure with dense reciprocal connections with the cortex suggesting modulation by, or participation in, brain state regulation. Here, we highlight recent work suggesting that neural activity in the claustrum supports cognitive processes associated with synchronized brain states characterized by increased low-frequency network activity. As an example, we outline how claustrum activity could support episodic memory consolidation during sleep.
Collapse
Affiliation(s)
- Alison D Do
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | - Coline Portet
- University of Strasbourg, Strasbourg, France; Laboratoire de Neurosciences Cognitives et Adaptatives, CNRS UMR7364, Strasbourg, France
| | - Romain Goutagny
- University of Strasbourg, Strasbourg, France; Laboratoire de Neurosciences Cognitives et Adaptatives, CNRS UMR7364, Strasbourg, France
| | - Jesse Jackson
- Department of Physiology, University of Alberta, Edmonton, AB, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
29
|
Cankar N, Beschorner N, Tsopanidou A, Qvist FL, Colaço AR, Andersen M, Kjaerby C, Delle C, Lambert M, Mundt F, Weikop P, Jucker M, Mann M, Skotte NH, Nedergaard M. Sleep deprivation leads to non-adaptive alterations in sleep microarchitecture and amyloid-β accumulation in a murine Alzheimer model. Cell Rep 2024; 43:114977. [PMID: 39541211 DOI: 10.1016/j.celrep.2024.114977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/09/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Impaired sleep is a common aspect of aging and often precedes the onset of Alzheimer's disease. Here, we compare the effects of sleep deprivation in young wild-type mice and their APP/PS1 littermates, a murine model of Alzheimer's disease. After 7 h of sleep deprivation, both genotypes exhibit an increase in EEG slow-wave activity. However, only the wild-type mice demonstrate an increase in the power of infraslow norepinephrine oscillations, which are characteristic of healthy non-rapid eye movement sleep. Notably, the APP/PS1 mice fail to enhance norepinephrine oscillations 24 h after sleep deprivation, coinciding with an accumulation of cerebral amyloid-β protein. Proteome analysis of cerebrospinal fluid and extracellular fluid further supports these findings by showing altered protein clearance in APP/PS1 mice. We propose that the suppression of infraslow norepinephrine oscillations following sleep deprivation contributes to increased vulnerability to sleep loss and heightens the risk of developing amyloid pathology in early stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Neža Cankar
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Natalie Beschorner
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Anastasia Tsopanidou
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Filippa L Qvist
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ana R Colaço
- Proteomics Research Infrastructure, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mie Andersen
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Celia Kjaerby
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Christine Delle
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Marius Lambert
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Filip Mundt
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Matthias Mann
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department for Proteomics and Signal Transduction, Max-Planck Institute for Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Niels Henning Skotte
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA.
| |
Collapse
|
30
|
Hata T, Grenier F, Hiraga T, Soya M, Okamoto M, Soya H. Promoting arousal associated with physical activity with the vitamin B 1 derivative TTFD. J Physiol Sci 2024; 75:100001. [PMID: 40008856 PMCID: PMC11979667 DOI: 10.1016/j.jphyss.2024.100001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 02/27/2025]
Abstract
Physical inactivity, which is a global issue, reduces physical and mental vitality, particularly impairing prefrontal-cortex-based mental health. This may trigger social withdrawal and depression, hindering the ability to have an active lifestyle. However, we have identified a beneficial agent, a vitamin B1 derivative called thiamine tetrahydrofurfuryl disulfide (TTFD), that enhances physical activity through dopaminergic regulation in the medial prefrontal cortex (mPFC) of rats. Since the brain dopaminergic system also regulates the sleep-wake cycle via the ascending arousal system, we postulated that TTFD may promote arousal. To test this, we performed electroencephalograms and electromyograms in rats, monitoring their physical activity and sleep-wake cycles after TTFD injection. Analysis revealed that TTFD acutely promotes arousal, reduces slow-wave sleep (SWS) and rapid eye movement (REM) sleep, and promotes increased physical activity. TTFD not only promotes physical activity but also increases arousal, thereby potentially contributing to enhanced mental health.
Collapse
Affiliation(s)
- Toshiaki Hata
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan; Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan
| | - François Grenier
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan
| | - Taichi Hiraga
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan
| | - Mariko Soya
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Fukuoka 812-8582, Japan
| | - Masahiro Okamoto
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan; Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan
| | - Hideaki Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan; Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8574, Japan.
| |
Collapse
|
31
|
Gu L, Shao W, Liu L, Xu Q, Wang Y, Gu J, Yang Y, Zhang Z, Wu Y, Shen Y, Yu Q, Lian X, Ma H, Zhang Y, Zhang H. NE contribution to rebooting unconsciousness caused by midazolam. eLife 2024; 13:RP97954. [PMID: 39565190 DOI: 10.7554/elife.97954] [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: 11/21/2024] Open
Abstract
The advent of midazolam holds profound implications for modern clinical practice. The hypnotic and sedative effects of midazolam afford it broad clinical applicability. However, the specific mechanisms underlying the modulation of altered consciousness by midazolam remain elusive. Herein, using pharmacology, optogenetics, chemogenetics, fiber photometry, and gene knockdown, this in vivo research revealed the role of locus coeruleus (LC)-ventrolateral preoptic nucleus noradrenergic neural circuit in regulating midazolam-induced altered consciousness. This effect was mediated by α1 adrenergic receptors. Moreover, gamma-aminobutyric acid receptor type A (GABAA-R) represents a mechanistically crucial binding site in the LC for midazolam. These findings will provide novel insights into the neural circuit mechanisms underlying the recovery of consciousness after midazolam administration and will help guide the timing of clinical dosing and propose effective intervention targets for timely recovery from midazolam-induced loss of consciousness.
Collapse
Affiliation(s)
- LeYuan Gu
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou, China
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - WeiHui Shao
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Liu
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qing Xu
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou, China
| | - YuLing Wang
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - JiaXuan Gu
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yue Yang
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - ZhuoYue Zhang
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou, China
| | - YaXuan Wu
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yue Shen
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Qian Yu
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - XiTing Lian
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou, China
| | - HaiXiang Ma
- Medical College of Jining Medical University, Shandong, China
| | - YuanLi Zhang
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - HongHai Zhang
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou, China
- Department of Anesthesiology, the Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| |
Collapse
|
32
|
Kim DI, Kang SJ, Jhang J, Jo YS, Park S, Ye M, Pyeon GH, Im GH, Kim SG, Han S. Encoding opposing valences through frequency-dependent transmitter switching in single peptidergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.09.622790. [PMID: 39574736 PMCID: PMC11581014 DOI: 10.1101/2024.11.09.622790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Peptidergic neurons often co-express fast transmitters and neuropeptides in separate vesicles with distinct release properties. However, the release dynamics of each transmitter in various contexts have not been fully understood in behaving animals. Here, we demonstrate that calcitonin gene-related peptide (CGRP) neurons in the external lateral subdivision of the parabrachial nucleus (CGRPPBel) encode opposing valence via differential release, rather than co-release, of glutamate and neuropeptides, according to firing rate. Glutamate is released preferentially at lower firing rates with minimal release at higher firing rates, whereas neuropeptides are released at higher firing rates, resulting in frequency-dependent switching of transmitters. Aversive stimuli evoke high frequency responses with accompanying neuropeptide release to encode negative valence, whereas appetitive stimuli evoke low frequency responses with glutamate release to encode positive valence. Our study reveals a previously unknown capability of single CGRPPBel neurons to bidirectionally encode valence via frequency-dependent differential release of transmitters in vivo.
Collapse
Affiliation(s)
- Dong-Il Kim
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Sukjae J. Kang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Jinho Jhang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Yong S. Jo
- School of Psychology, Korea University; Seoul, Republic of Korea
| | - Seahyung Park
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Mao Ye
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
| | - Gyeong Hee Pyeon
- School of Psychology, Korea University; Seoul, Republic of Korea
| | - Geun-Ho Im
- Center for Neuroscience Imaging Research, Institute for Basic Science; Suwon, Republic of Korea
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science; Suwon, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University; Suwon, Republic of Korea
| | - Sung Han
- Peptide Biology Laboratory, The Salk Institute for Biological Studies; La Jolla, CA 92037, USA
- Center for Neuroscience Imaging Research, Institute for Basic Science; Suwon, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University; Suwon, Republic of Korea
| |
Collapse
|
33
|
Bombassaro B, Araujo EP, Velloso LA. The hypothalamus as the central regulator of energy balance and its impact on current and future obesity treatments. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e240082. [PMID: 39876968 PMCID: PMC11771753 DOI: 10.20945/2359-4292-2024-0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/10/2024] [Indexed: 01/31/2025]
Abstract
The hypothalamus is a master regulator of energy balance in the body. First-order hypothalamic neurons localized in the arcuate nucleus sense systemic signals that indicate the energy stores in the body. Through distinct projections, arcuate nucleus neurons communicate with second-order neurons, which are mostly localized in the paraventricular nucleus and in the lateral hypothalamus. The signals then proceed to third- and fourth-order neurons that activate complex responses aimed at maintaining whole-body energy homeostasis. During the last 30 years, since the identification of leptin in 1994, there has been a great advance in the unveiling of the hypothalamic and extra-hypothalamic neuronal networks that control energy balance. This has contributed to the characterization of the mechanisms by which glucagon-like peptide-1 receptor agonists promote body mass reduction and has opened new windows of opportunity for the development of drugs to treat obesity. This review presents an overview of the mechanisms involved in the hypothalamic regulation of energy balance and discusses how advancements in this field are contributing to the development of new pharmacological strategies to treat obesity.
Collapse
Affiliation(s)
- Bruna Bombassaro
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| | - Eliana P Araujo
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| | - Licio A Velloso
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| |
Collapse
|
34
|
Heer C, Sheffield M. Distinct catecholaminergic pathways projecting to hippocampal CA1 transmit contrasting signals during navigation in familiar and novel environments. eLife 2024; 13:RP95213. [PMID: 39504262 PMCID: PMC11540301 DOI: 10.7554/elife.95213] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
Neuromodulatory inputs to the hippocampus play pivotal roles in modulating synaptic plasticity, shaping neuronal activity, and influencing learning and memory. Recently, it has been shown that the main sources of catecholamines to the hippocampus, ventral tegmental area (VTA) and locus coeruleus (LC), may have overlapping release of neurotransmitters and effects on the hippocampus. Therefore, to dissect the impacts of both VTA and LC circuits on hippocampal function, a thorough examination of how these pathways might differentially operate during behavior and learning is necessary. We therefore utilized two-photon microscopy to functionally image the activity of VTA and LC axons within the CA1 region of the dorsal hippocampus in head-fixed male mice navigating linear paths within virtual reality (VR) environments. We found that within familiar environments some VTA axons and the vast majority of LC axons showed a correlation with the animals' running speed. However, as mice approached previously learned rewarded locations, a large majority of VTA axons exhibited a gradual ramping-up of activity, peaking at the reward location. In contrast, LC axons displayed a pre-movement signal predictive of the animal's transition from immobility to movement. Interestingly, a marked divergence emerged following a switch from the familiar to novel VR environments. Many LC axons showed large increases in activity that remained elevated for over a minute, while the previously observed VTA axon ramping-to-reward dynamics disappeared during the same period. In conclusion, these findings highlight distinct roles of VTA and LC catecholaminergic inputs in the dorsal CA1 hippocampal region. These inputs encode unique information, with reward information in VTA inputs and novelty and kinematic information in LC inputs, likely contributing to differential modulation of hippocampal activity during behavior and learning.
Collapse
Affiliation(s)
- Chad Heer
- The Department of Neurobiology, The University of ChicagoChicagoUnited States
| | - Mark Sheffield
- The Department of Neurobiology, The University of ChicagoChicagoUnited States
| |
Collapse
|
35
|
Martin KA, Papadoyannis ES, Schiavo JK, Fadaei SS, Issa HA, Song SC, Valencia SO, Temiz NZ, McGinley MJ, McCormick DA, Froemke RC. Vagus nerve stimulation recruits the central cholinergic system to enhance perceptual learning. Nat Neurosci 2024; 27:2152-2166. [PMID: 39284963 PMCID: PMC11932732 DOI: 10.1038/s41593-024-01767-4] [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: 01/28/2022] [Accepted: 08/15/2024] [Indexed: 11/07/2024]
Abstract
Perception can be refined by experience, up to certain limits. It is unclear whether perceptual limits are absolute or could be partially overcome via enhanced neuromodulation and/or plasticity. Recent studies suggest that peripheral nerve stimulation, specifically vagus nerve stimulation (VNS), can alter neural activity and augment experience-dependent plasticity, although little is known about central mechanisms recruited by VNS. Here we developed an auditory discrimination task for mice implanted with a VNS electrode. VNS applied during behavior gradually improved discrimination abilities beyond the level achieved by training alone. Two-photon imaging revealed VNS induced changes to auditory cortical responses and activated cortically projecting cholinergic axons. Anatomical and optogenetic experiments indicated that VNS can enhance task performance through activation of the central cholinergic system. These results highlight the importance of cholinergic modulation for the efficacy of VNS and may contribute to further refinement of VNS methodology for clinical conditions.
Collapse
Affiliation(s)
- Kathleen A Martin
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Eleni S Papadoyannis
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Jennifer K Schiavo
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Saba Shokat Fadaei
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Habon A Issa
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Soomin C Song
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Sofia Orrey Valencia
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Nesibe Z Temiz
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Matthew J McGinley
- Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | | | - Robert C Froemke
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA.
- Department of Otolaryngology, New York University School of Medicine, New York, NY, USA.
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA.
- Center for Neural Science, New York University, New York, NY, USA.
| |
Collapse
|
36
|
Brannan S, Garbe L, Richardson BD. Early life stress induced sex-specific changes in behavior is paralleled by altered locus coeruleus physiology in BALB/cJ mice. Neurobiol Stress 2024; 33:100674. [PMID: 39385751 PMCID: PMC11462065 DOI: 10.1016/j.ynstr.2024.100674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Adverse childhood experiences have been associated with many neurodevelopmental and affective disorders including attention deficit hyperactivity disorder and generalized anxiety disorder, with more exposures increasing negative risk. Sex and genetic background are biological variables involved in adverse psychiatric outcomes due to early life trauma. Females in general have an increased prevalence of stress-related psychopathologies beginning after adolescence, indicative of adolescence being a female-specific sensitive period. To understand the underlying neuronal mechanisms potentially responsible for this relationship between genetic background, sex, stress/trauma, and cognitive/affective behaviors, we assessed behavioral and neuronal changes in a novel animal model of early life stress exposure. Male and female BALB/cJ mice that express elevated basal anxiety-like behaviors and differences in monoamine signaling-associated genes, were exposed to an early life variable stress protocol that combined deprivation in early life with unpredictability in adolescence. Stress exposure produced hyperlocomotion and attention deficits (5-choice serial reaction time task) in male and female mice along with female-specific increased anxiety-like behavior. These behavioral changes were paralleled by reduced excitability of locus coeruleus (LC) neurons, due to resting membrane potential hyperpolarization in males and a female-specific increase in action potential delay time. These data describe a novel interaction between sex, genetic background, and early life stress that results in behavioral changes in clinically relevant domains and potential underlying mechanistic lasting changes in physiological properties of neurons in the LC.
Collapse
Affiliation(s)
- Savannah Brannan
- Department of Pharmacology, Southern Illinois University – School of Medicine, Springfield, IL, 62702, USA
| | - Lauren Garbe
- Department of Pharmacology, Southern Illinois University – School of Medicine, Springfield, IL, 62702, USA
| | - Ben D. Richardson
- Department of Pharmacology, Southern Illinois University – School of Medicine, Springfield, IL, 62702, USA
| |
Collapse
|
37
|
Grimm C, Duss SN, Privitera M, Munn BR, Karalis N, Frässle S, Wilhelm M, Patriarchi T, Razansky D, Wenderoth N, Shine JM, Bohacek J, Zerbi V. Tonic and burst-like locus coeruleus stimulation distinctly shift network activity across the cortical hierarchy. Nat Neurosci 2024; 27:2167-2177. [PMID: 39284964 PMCID: PMC11537968 DOI: 10.1038/s41593-024-01755-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 08/07/2024] [Indexed: 11/07/2024]
Abstract
Noradrenaline (NA) release from the locus coeruleus (LC) changes activity and connectivity in neuronal networks across the brain, modulating multiple behavioral states. NA release is mediated by both tonic and burst-like LC activity. However, it is unknown whether the functional changes in target areas depend on these firing patterns. Using optogenetics, photometry, electrophysiology and functional magnetic resonance imaging in mice, we show that tonic and burst-like LC firing patterns elicit brain responses that hinge on their distinct NA release dynamics. During moderate tonic LC activation, NA release engages regions associated with associative processing, while burst-like stimulation biases the brain toward sensory processing. These activation patterns locally couple with increased astrocytic and inhibitory activity and change the brain's topological configuration in line with the hierarchical organization of the cerebral cortex. Together, these findings reveal how the LC-NA system achieves a nuanced regulation of global circuit operations.
Collapse
Affiliation(s)
- Christina Grimm
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Neuro-X institute, School of Engineering (STI), EPFL, Lausanne, Switzerland
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
| | - Sian N Duss
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Mattia Privitera
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Brandon R Munn
- School of Physics, The University of Sydney, Sydney, New South Wales, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Nikolaos Karalis
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Stefan Frässle
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zürich & ETH Zürich, Zürich, Switzerland
| | - Maria Wilhelm
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Tommaso Patriarchi
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
- Chemical Neuropharmacology, Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | - Daniel Razansky
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zürich, Zürich, Switzerland
- Institute of Biological and Medical Imaging (IBMI), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Nicole Wenderoth
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - James M Shine
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich, Switzerland.
| | - Valerio Zerbi
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.
- Neuro-X institute, School of Engineering (STI), EPFL, Lausanne, Switzerland.
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland.
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
38
|
Wei J, Xiao C, Zhang GW, Shen L, Tao HW, Zhang LI. A distributed auditory network mediated by pontine central gray underlies ultra-fast awakening in response to alerting sounds. Curr Biol 2024; 34:4597-4611.e5. [PMID: 39265569 PMCID: PMC11521200 DOI: 10.1016/j.cub.2024.08.020] [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: 03/11/2024] [Revised: 07/12/2024] [Accepted: 08/13/2024] [Indexed: 09/14/2024]
Abstract
Sleeping animals can be woken up rapidly by external threat signals, which is an essential defense mechanism for survival. However, neuronal circuits underlying the fast transmission of sensory signals for this process remain unclear. Here, we report in mice that alerting sound can induce rapid awakening within hundreds of milliseconds and that glutamatergic neurons in the pontine central gray (PCG) play an important role in this process. These neurons exhibit higher sensitivity to auditory stimuli in sleep than wakefulness. Suppressing these neurons results in reduced sound-induced awakening and increased sleep in intrinsic sleep/wake cycles, whereas their activation induces ultra-fast awakening from sleep and accelerates awakening from anesthesia. Additionally, the sound-induced awakening can be attributed to the propagation of auditory signals from the PCG to multiple arousal-related regions, including the mediodorsal thalamus, lateral hypothalamus, and ventral tegmental area. Thus, the PCG serves as an essential distribution center to orchestrate a global auditory network to promote rapid awakening.
Collapse
Affiliation(s)
- Jinxing Wei
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cuiyu Xiao
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Guang-Wei Zhang
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Li Shen
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Huizhong W Tao
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
39
|
Zhou Z, Tang Y, Li R, Wang W, Dai Z. Hovering flight regulation of pigeon robots in laboratory and field. iScience 2024; 27:110927. [PMID: 39391728 PMCID: PMC11465124 DOI: 10.1016/j.isci.2024.110927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/11/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Compared to traditional bio-mimic robots, animal robots show superior locomotion, energy efficiency, and adaptability to complex environments but most remained in laboratory stage, needing further development for practical applications like exploration and inspection. Our pigeon robots validated in both laboratory and field, tested with an electrical stimulus unit (2-s duration, 0.5 ms pulse width, 80 Hz frequency). In a fixed stimulus procedure, hovering flight was conducted with 8 stimulus units applied every 2 s after flew over the trigger boundary. In a flexible procedure, stimulus was applied whenever they deviated from a virtual circle, with pulse width gains of 0.1 ms or 0.2 ms according to the trajectory angle. These optimized protocols achieved a success hovering rate of 87.5% and circle curvatures of 0.008 m-1-0.024 m-1, largely advancing the practical application of animal robots.
Collapse
Affiliation(s)
- Zhengyue Zhou
- Institute of Bio-inspired Structure and Surface Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
| | - Yezhong Tang
- Institute of Bio-inspired Structure and Surface Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
- Chengdu Institute of Biology, Chinese Academy of Sciences. No.9 Section 4, Renmin Nan Road, Chengdu 610041, Sichuan, China
| | - Rongxun Li
- Institute of Bio-inspired Structure and Surface Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
| | - Wenbo Wang
- Institute of Bio-inspired Structure and Surface Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
| | - Zhendong Dai
- Institute of Bio-inspired Structure and Surface Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, China
| |
Collapse
|
40
|
Kelberman MA, Rodberg E, Arabzadeh E, Bair-Marshall CJ, Berridge CW, Berrocoso E, Breton-Provencher V, Chandler DJ, Che A, Davy O, Devilbiss DM, Downs AM, Drummond G, Dvorkin R, Fazlali Z, Froemke RC, Glennon E, Gold JI, Ito H, Jiang X, Johansen JP, Kaye AP, Kim JR, Kuo CC, Liu RJ, Liu Y, Llorca-Torralba M, McCall JG, McElligott ZA, McKinney AM, Miguelez C, Min MY, Nowlan AC, Omrani M, Poe GR, Pickering AE, Ranjbar-Slamloo Y, Razquin J, Rodenkirch C, Sales AC, Satyasambit R, Shea SD, Sur M, Tkaczynski JA, Torres-Sanchez S, Uematsu A, Vazquez CR, Vreven A, Wang Q, Waterhouse BD, Yang HW, Yang JH, Zhao L, Zouridis IS, Weinshenker D, Vazey E, Totah NK. Diversity of ancestral brainstem noradrenergic neurons across species and multiple biological factors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618224. [PMID: 39464004 PMCID: PMC11507722 DOI: 10.1101/2024.10.14.618224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The brainstem region, locus coeruleus (LC), has been remarkably conserved across vertebrates. Evolution has woven the LC into wide-ranging neural circuits that influence functions as broad as autonomic systems, the stress response, nociception, sleep, and high-level cognition among others. Given this conservation, there is a strong possibility that LC activity is inherently similar across species, and furthermore that age, sex, and brain state influence LC activity similarly across species. The degree to which LC activity is homogenous across these factors, however, has never been assessed due to the small sample size of individual studies. Here, we pool data from 20 laboratories (1,855 neurons) and show diversity across both intrinsic and extrinsic factors such as species, age, sex and brain state. We use a negative binomial regression model to compare activity from male monkeys, and rats and mice of both sexes that were recorded across brain states from brain slices ex vivo or under different anesthetics or during wakefulness in vivo. LC activity differed due to complex interactions of species, sex, and brain state. The LC became more active during aging, independent of sex. Finally, in contrast to the foundational principle that all species express two distinct LC firing modes ("tonic" or "phasic"), we discovered great diversity within spontaneous LC firing patterns. Different factors were associated with higher incidence of some firing modes. We conclude that the activity of the evolutionarily-ancient LC is not conserved. Inherent differences due to age and species-sex-brain state interactions have implications for understanding the role of LC in species-specific naturalistic behavior, as well as in psychiatric disorders, cardiovascular disease, immunology, and metabolic disorders.
Collapse
Affiliation(s)
- Michael A. Kelberman
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Ellen Rodberg
- Department of Biology, University of Massachusetts Amherst, Amherst, MA, USA
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Ehsan Arabzadeh
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, The Australian National University, Canberra, AUS
| | - Chloe J. Bair-Marshall
- Neuroscience Institute, NYU Langone Medical Center, New York University, New York, New York, USA
| | - Craig W. Berridge
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Esther Berrocoso
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, School of Medicine, Biomedical Research and Innovation Institute of Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Alicia Che
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Oscar Davy
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | | | - Anthony M. Downs
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabrielle Drummond
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roman Dvorkin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Zeinab Fazlali
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
- Department of Psychiatry, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Robert C. Froemke
- Neuroscience Institute, NYU Langone Medical Center, New York University, New York, New York, USA
- Department of Otolaryngology, NYU Grossman School of Medicine, New York, NY, USA
| | - Erin Glennon
- Neuroscience Institute, NYU Langone Medical Center, New York University, New York, New York, USA
- Department of Neurology, Weill Cornell Medicine, New York
| | - Joshua I. Gold
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Hiroki Ito
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Xiaolong Jiang
- Department of Neuroscience, Baylor College of Medicine Neurological Research Institute at Texas Children’s Hospital, 1250, Houston, TX, USA
- Department of Ophthalmology, Baylor College of Medicine Neurological Research Institute at Texas Children’s Hospital, 1250, Houston, TX, USA
| | | | - Alfred P. Kaye
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
- Clinical Neurosciences Division, VA National Center for PTSD, West Haven, CT, USA
| | - Jenny R. Kim
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Chao-Cheng Kuo
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Rong-Jian Liu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yang Liu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Meritxell Llorca-Torralba
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, School of Medicine, Biomedical Research and Innovation Institute of Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordan G. McCall
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Zoe A. McElligott
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew M. McKinney
- Department of Neuroscience, Baylor College of Medicine Neurological Research Institute at Texas Children’s Hospital, 1250, Houston, TX, USA
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Ming-Yuan Min
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Alexandra C. Nowlan
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mohsen Omrani
- Department of Psychiatry, Queen’s University, Kingston, ON, Canada
| | - Gina R. Poe
- Integrative Biology and Physiology, UCLA, Los Angeles, CA, USA
| | - Anthony Edward Pickering
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Yadollah Ranjbar-Slamloo
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Jone Razquin
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Charles Rodenkirch
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Anna C. Sales
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Rath Satyasambit
- RIKEN Center for Brain Science, Wako-shi Saitama, Japan
- Department of Computer Science, Tokyo Institute of Technology, Midori, Yokohama, Japan
| | | | - Mriganka Sur
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Sonia Torres-Sanchez
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, School of Medicine, Biomedical Research and Innovation Institute of Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Akira Uematsu
- Human Informatics and Information Research Institute, National Institute of Advanced Industrial Science and Technology, Japan
| | - Chayla R. Vazquez
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Amelien Vreven
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Qi Wang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | | - Hsiu-Wen Yang
- Department of Biomedical Sciences, Chung-Shan Medical University, Taichung, Taiwan
| | - Jen-Hau Yang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Doctoral Program of Clinical and Experimental Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Liping Zhao
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA USA
| | - Ioannis S. Zouridis
- Graduate Training Centre of Neuroscience, International Max Planck Research School (IMPRS), University of Tübingen, Tübingen, Germany
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | | | - Elena Vazey
- Department of Biology, University of Massachusetts Amherst, Amherst, MA, USA
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Nelson K. Totah
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| |
Collapse
|
41
|
Murphy KR, Farrell JS, Bendig J, Mitra A, Luff C, Stelzer IA, Yamaguchi H, Angelakos CC, Choi M, Bian W, DiIanni T, Pujol EM, Matosevich N, Airan R, Gaudillière B, Konofagou EE, Butts-Pauly K, Soltesz I, de Lecea L. Optimized ultrasound neuromodulation for non-invasive control of behavior and physiology. Neuron 2024; 112:3252-3266.e5. [PMID: 39079529 PMCID: PMC11709124 DOI: 10.1016/j.neuron.2024.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 05/09/2024] [Accepted: 07/02/2024] [Indexed: 08/09/2024]
Abstract
Focused ultrasound can non-invasively modulate neural activity, but whether effective stimulation parameters generalize across brain regions and cell types remains unknown. We used focused ultrasound coupled with fiber photometry to identify optimal neuromodulation parameters for four different arousal centers of the brain in an effort to yield overt changes in behavior. Applying coordinate descent, we found that optimal parameters for excitation or inhibition are highly distinct, the effects of which are generally conserved across brain regions and cell types. Optimized stimulations induced clear, target-specific behavioral effects, whereas non-optimized protocols of equivalent energy resulted in substantially less or no change in behavior. These outcomes were independent of auditory confounds and, contrary to expectation, accompanied by a cyclooxygenase-dependent and prolonged reduction in local blood flow and temperature with brain-region-specific scaling. These findings demonstrate that carefully tuned and targeted ultrasound can exhibit powerful effects on complex behavior and physiology.
Collapse
Affiliation(s)
- Keith R Murphy
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Jordan S Farrell
- Department of Neurosurgery, Stanford University, Stanford, CA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA; F.M. Kirby Neurobiology Center, Harvard Medical School, Boston, MA, USA
| | - Jonas Bendig
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Anish Mitra
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Charlotte Luff
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Ina A Stelzer
- Department of Anesthesia, Stanford University, Stanford, CA, USA
| | - Hiroshi Yamaguchi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Department of Neuroscience, Nagoya University, Nagoya, Japan
| | | | - Mihyun Choi
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Wenjie Bian
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Tommaso DiIanni
- Department of Radiology, Stanford University, Stanford, CA, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Esther Martinez Pujol
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Noa Matosevich
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Raag Airan
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Brice Gaudillière
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Kim Butts-Pauly
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
42
|
Kong D, Kong L, Liu C, Wu Q, Wang J, Dai C. Commissural and monosynaptic inputs to medial vestibular nucleus GABAergic neurons in mice. Front Neurol 2024; 15:1484488. [PMID: 39440253 PMCID: PMC11493639 DOI: 10.3389/fneur.2024.1484488] [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: 08/21/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
Objective MVN GABAergic neurons is involved in the rebalance of commissural system contributing to alleviating acute peripheral vestibular dysfunction syndrome. This study aims to depict monosynaptic inputs to MVN GABAergic neurons. Methods The modified rabies virus-based retrogradation method combined with the VGAT-IRES-Cre mice was used in this study. Moreover, the commissural connections with MVN GABAergic neurons were analyzed. Results We identified 60 nuclei projecting to MVN GABAergic neurons primarily distributed in the cerebellum and the medulla. The uvula-nodulus, gigantocellular reticular nucleus, prepositus nucleus, intermediate reticular nucleus, and three other nuclei sent dense inputs to MVN GABAergic neurons. The medial (fastigial) cerebellar nucleus, dorsal paragigantocellular nucleus, lateral paragigantocellular nucleus and 10 other nuclei sent moderate inputs to MVN GABAergic neurons. Sparse inputs to MVN GABAergic neurons originated from the nucleus of the solitary tract, lateral reticular nucleus, pedunculopontine tegmental nucleus and 37 other nuclei. The MVN GABAergic neurons were regulated by the contralateral MVN, lateral vestibular nucleus, superior vestibular nucleus, and inferior vestibular nucleus. Conclusion Our study contributes to further understanding of the vestibular dysfunction in terms of neural circuits and search for new strategies to facilitate vestibular compensation.
Collapse
Affiliation(s)
- Dedi Kong
- Department of Otology and Skull Base Surgery, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
- Key Laboratory of Hearing Medicine, Ministry of Health, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Lingxi Kong
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Chengwei Liu
- Department of Otology and Skull Base Surgery, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
- Key Laboratory of Hearing Medicine, Ministry of Health, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Qianru Wu
- Department of Otology and Skull Base Surgery, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
- Key Laboratory of Hearing Medicine, Ministry of Health, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Jing Wang
- Department of Otology and Skull Base Surgery, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
- Key Laboratory of Hearing Medicine, Ministry of Health, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Chunfu Dai
- Department of Otology and Skull Base Surgery, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
- Key Laboratory of Hearing Medicine, Ministry of Health, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
| |
Collapse
|
43
|
Sulaman BA, Zhang Y, Matosevich N, Kjærby C, Foustoukos G, Andersen M, Eban-Rothschild A. Emerging Functions of Neuromodulation during Sleep. J Neurosci 2024; 44:e1277242024. [PMID: 39358018 PMCID: PMC11450531 DOI: 10.1523/jneurosci.1277-24.2024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 10/04/2024] Open
Abstract
Neuromodulators act on multiple timescales to affect neuronal activity and behavior. They function as synaptic fine-tuners and master coordinators of neuronal activity across distant brain regions and body organs. While much research on neuromodulation has focused on roles in promoting features of wakefulness and transitions between sleep and wake states, the precise dynamics and functions of neuromodulatory signaling during sleep have received less attention. This review discusses research presented at our minisymposium at the 2024 Society for Neuroscience meeting, highlighting how norepinephrine, dopamine, and acetylcholine orchestrate brain oscillatory activity, control sleep architecture and microarchitecture, regulate responsiveness to sensory stimuli, and facilitate memory consolidation. The potential of each neuromodulator to influence neuronal activity is shaped by the state of the synaptic milieu, which in turn is influenced by the organismal or systemic state. Investigating the effects of neuromodulator release across different sleep substates and synaptic environments offers unique opportunities to deepen our understanding of neuromodulation and explore the distinct computational opportunities that arise during sleep. Moreover, since alterations in neuromodulatory signaling and sleep are implicated in various neuropsychiatric disorders and because existing pharmacological treatments affect neuromodulatory signaling, gaining a deeper understanding of the less-studied aspects of neuromodulators during sleep is of high importance.
Collapse
Affiliation(s)
- Bibi Alika Sulaman
- Department of Psychology, University of Michigan, Ann Arbor, Michigan 48109
| | - Yiyao Zhang
- Neuroscience Institute, New York University, New York, New York 10016
| | - Noa Matosevich
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Celia Kjærby
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Georgios Foustoukos
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne 1005, Switzerland
| | - Mie Andersen
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen 2200, Denmark
| | | |
Collapse
|
44
|
Metha J, Ji Y, Braun C, Nicholson JR, De Lecea L, Murawski C, Hoyer D, Jacobson LH. Hypocretin-1 receptor antagonism improves inhibitory control during the Go/No-Go task in highly motivated, impulsive male mice. Psychopharmacology (Berl) 2024; 241:2171-2187. [PMID: 38886189 PMCID: PMC11442560 DOI: 10.1007/s00213-024-06628-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 05/28/2024] [Indexed: 06/20/2024]
Abstract
RATIONALE Motivation and inhibitory control are dominantly regulated by the dopaminergic (DA) and noradrenergic (NA) systems, respectively. Hypothalamic hypocretin (orexin) neurons provide afferent inputs to DA and NA nuclei and hypocretin-1 receptors (HcrtR1) are implicated in reward and addiction. However, the role of the HcrtR1 in inhibitory control is not well understood. OBJECTIVES To determine the effects of HcrtR1 antagonism and motivational state in inhibitory control using the go/no-go task in mice. METHODS n = 23 male C57Bl/6JArc mice were trained in a go/no-go task. Decision tree dendrogram analysis of training data identified more and less impulsive clusters of animals. A HcrtR1 antagonist (BI001, 12.5 mg/kg, per os) or vehicle were then administered 30 min before go/no-go testing, once daily for 5 days, under high (food-restricted) and low (free-feeding) motivational states in a latin-square crossover design. Compound exposure levels were assessed in a satellite group of animals. RESULTS HcrtR1 antagonism increased go accuracy and decreased no-go accuracy in free-feeding animals overall, whereas it decreased go accuracy and increased no-go accuracy only in more impulsive, food restricted mice. HcrtR1 antagonism also showed differential effects in premature responding, which was increased in response to the antagonist in free-feeding, less impulsive animals, and decreased in food restricted, more impulsive animals. HcrtR1 receptor occupancy by BI001 was estimated at ~ 66% during the task. CONCLUSIONS These data indicate that hypocretin signalling plays roles in goal-directed behaviour and inhibitory control in a motivational state-dependant manner. While likely not useful in all settings, HcrtR1 antagonism may be beneficial in improving inhibitory control in impulsive subpopulations.
Collapse
Affiliation(s)
- Jeremy Metha
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Department of Finance, The University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Yijun Ji
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia
- Circadian Misalignment and Shift Work Laboratory, Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Notting Hill, VIC, 3162, Australia
| | - Clemens Braun
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| | - Janet R Nicholson
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| | - Luis De Lecea
- Department of Psychiatry and Behavioural Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Carsten Murawski
- Department of Finance, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Daniel Hoyer
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Laura H Jacobson
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia.
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3052, Australia.
| |
Collapse
|
45
|
Mir HD, Yang Q, Maximin E, Montardy Q, Ji S, Cheng Q, Shan X, Wang L, Naudon L, Rabot S, Li L. Indole induces anxiety-like behaviour in mice mediated by brainstem locus coeruleus activation. Neurobiol Dis 2024; 200:106606. [PMID: 39019292 DOI: 10.1016/j.nbd.2024.106606] [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: 04/04/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024] Open
Abstract
The gut microbiota produces metabolites that enrich the host metabolome and play a part in host physiology, including brain functions. Yet the biological mediators of this gut-brain signal transduction remain largely unknown. In this study, the possible role of the gut microbiota metabolite indole, originating from tryptophan, was investigated. Oral administration of indole to simulate microbial overproduction of this compound in the gut consistently led to impaired locomotion and anxiety-like behaviour in both C3H/HeN and C57BL/6J mice. By employing c-Fos protein expression mapping in mice, we observed a noticeable increase in brain activation within the dorsal motor nucleus of the vagus nerve (DMX) and the locus coeruleus (LC) regions in a dose-dependent manner. Further immune co-labelling experiments elucidated that the primary cells activated within the LC were tyrosine hydroxylase positive. To delve deeper into the mechanistic aspects, we conducted chemogenetic activation experiments on LC norepinephrine neurons with two doses of clozapine N-oxide (CNO). Low dose of CNO at 0.5 mg/kg induced no change in locomotion but anxiety-like behaviour, while high dose of CNO at 2 mg/kg resulted in locomotion impairment and anxiety-like behaviour. These findings support the neuroactive roles of indole in mediating gut-brain communication. It also highlights the LC as a novel hub in the gut-brain axis, encouraging further investigations.
Collapse
Affiliation(s)
- Hayatte-Dounia Mir
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Qingning Yang
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Brain Connectome and Behaviour, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Elise Maximin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Quentin Montardy
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Brain Connectome and Behaviour, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Shuqin Ji
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Brain Connectome and Behaviour, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Qi Cheng
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Brain Connectome and Behaviour, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Xiaochun Shan
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Brain Connectome and Behaviour, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Liping Wang
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Brain Connectome and Behaviour, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Laurent Naudon
- Université Paris-Saclay, INRAE, AgroParisTech, CNRS, Micalis Institute, Jouy-en-Josas, France
| | - Sylvie Rabot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France.
| | - Lei Li
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Brain Connectome and Behaviour, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
| |
Collapse
|
46
|
Nassar MR. Toward a computational role for locus coeruleus/norepinephrine arousal systems. Curr Opin Behav Sci 2024; 59:101407. [PMID: 39070697 PMCID: PMC11280330 DOI: 10.1016/j.cobeha.2024.101407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Brain and behavior undergo measurable changes in their underlying state and neuromodulators are thought to contribute to these fluctuations. Why do we undergo such changes, and what function could the underlying neuromodulatory systems perform? Here we examine theoretical answers to these questions with respect to the locus coeruleus/norepinephrine system focusing on peripheral markers for arousal, such as pupil diameter, that are thought to provide a window into brain wide noradrenergic signaling. We explore a computational role for arousal systems in facilitating internal state transitions that facilitate credit assignment and promote accurate perceptions in non-stationary environments. We summarize recent work that supports this idea and highlight open questions as well as alternative views of how arousal affects cognition.
Collapse
Affiliation(s)
- M R Nassar
- Brown University, Dept of Neuroscience and Carney Institute for Brain Science
| |
Collapse
|
47
|
Sawada T, Iino Y, Yoshida K, Okazaki H, Nomura S, Shimizu C, Arima T, Juichi M, Zhou S, Kurabayashi N, Sakurai T, Yagishita S, Yanagisawa M, Toyoizumi T, Kasai H, Shi S. Prefrontal synaptic regulation of homeostatic sleep pressure revealed through synaptic chemogenetics. Science 2024; 385:1459-1465. [PMID: 39325885 DOI: 10.1126/science.adl3043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 06/28/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024]
Abstract
Sleep is regulated by homeostatic processes, yet the biological basis of sleep pressure that accumulates during wakefulness, triggers sleep, and dissipates during sleep remains elusive. We explored a causal relationship between cellular synaptic strength and electroencephalography delta power indicating macro-level sleep pressure by developing a theoretical framework and a molecular tool to manipulate synaptic strength. The mathematical model predicted that increased synaptic strength promotes the neuronal "down state" and raises the delta power. Our molecular tool (synapse-targeted chemically induced translocation of Kalirin-7, SYNCit-K), which induces dendritic spine enlargement and synaptic potentiation through chemically induced translocation of protein Kalirin-7, demonstrated that synaptic potentiation of excitatory neurons in the prefrontal cortex (PFC) increases nonrapid eye movement sleep amounts and delta power. Thus, synaptic strength of PFC excitatory neurons dictates sleep pressure in mammals.
Collapse
Affiliation(s)
- Takeshi Sawada
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yusuke Iino
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kensuke Yoshida
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Hitoshi Okazaki
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shinnosuke Nomura
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Chika Shimizu
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tomoki Arima
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Motoki Juichi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Siqi Zhou
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | | | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Molecular Behavioral Physiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Sho Yagishita
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Taro Toyoizumi
- RIKEN Center for Brain Science, Wako, Saitama, Japan
- Department of Mathematical Informatics, Graduate School of Information Science and Technology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Haruo Kasai
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shoi Shi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
48
|
Mir FA, Jha SK. The Kir channel in the nucleus tractus solitarius integrates the chemosensory system with REM sleep executive machinery for homeostatic balance. Sci Rep 2024; 14:21651. [PMID: 39289431 PMCID: PMC11408532 DOI: 10.1038/s41598-024-71818-0] [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] [Accepted: 08/30/2024] [Indexed: 09/19/2024] Open
Abstract
The locus coeruleus (LC), nucleus tractus solitarius (NTS), and retrotrapezoid nucleus (RTN) are critical chemosensory regions in the brainstem. In the LC, acid-sensing ion channels and proton pumps serve as H+ sensors and facilitate the transition from non-rapid eye movement (NREM) to rapid eye movement (REM) sleep. Interestingly, the potassium inward rectifier (KIR) channels in the LC, NTS, and RTN also act as H+-sensors and are a primary target for improving sleep in obstructive sleep apnea and Rett syndrome patients. However, the role of Kir channels in NREM to REM sleep transition for H+ homeostasis is not known. Male Wistar rats were surgically prepared for chronic sleep-wake recording and drug delivery into the LC, NTS, and RTN. In different animal cohorts, microinjections of the Kir channel inhibitor, barium chloride (BaCl2), at concentrations of 1 mM (low dose) and 2 mM (high dose) in the LC and RTN significantly increased wakefulness and decreased NREM sleep. However, BaCl2 microinjection into the LC notably reduced REM sleep, whereas it didn't change in the RTN-injected group. Interestingly, BaCl2 microinjections into the NTS significantly decreased wakefulness and increased the percent amount of NREM and REM sleep. Additionally, with the infusion of BaCl2 into the NTS, the mean REM sleep episode numbers significantly increased, but the length of the REM sleep episode didn't change. These findings suggest that the Kir channels in the NTS, but not in the LC and RTN, modulate state transition from NREM to REM sleep.
Collapse
Affiliation(s)
- Fayaz A Mir
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
- Department of Anesthesia, Critical Care & Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Sushil K Jha
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
49
|
Wang X, Li Z, Wang X, Chen J, Guo Z, Qiao B, Qin L. Effects of Phasic Activation of Locus Ceruleus on Cortical Neural Activity and Auditory Discrimination Behavior. J Neurosci 2024; 44:e1296232024. [PMID: 39134421 PMCID: PMC11391501 DOI: 10.1523/jneurosci.1296-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 09/13/2024] Open
Abstract
Although the locus ceruleus (LC) is recognized as a crucial modulator for attention and perception by releasing norepinephrine into various cortical regions, the impact of LC-noradrenergic (LC-NE) modulation on auditory discrimination behavior remains elusive. In this study, we firstly recorded local field potential and single-unit activity in multiple cortical regions associated with auditory-motor processing, including the auditory cortex, posterior parietal cortex, secondary motor cortex, anterior cingulate cortex, prefrontal cortex, and orbitofrontal cortex (OFC), in response to optogenetic activation (40 Hz and 0.5 s) of the LC-NE neurons in awake mice (male). We found that phasic LC stimulation induced a persistent high gamma oscillation (50-80 Hz) in the OFC. Phasic activation of LC-NE neurons also resulted in a corresponding increase in norepinephrine levels in the OFC, accompanied by a pupillary dilation response. Furthermore, when mice were performing a go/no-go auditory discrimination task, we optogeneticaly activated the neural projections from LC to OFC and revealed a shortened latency in behavioral responses to sound stimuli and an increased false alarm rate. These impulsive behavioral responses may be associated with the gamma neural activity in the OFC. These findings have broadened our understanding of the neural mechanisms involved in the role of LC in auditory-motor processing.
Collapse
Affiliation(s)
- Xuejiao Wang
- Department of Physiology, China Medical University, Shenyang 110122, China
| | - Zijie Li
- Department of Physiology, China Medical University, Shenyang 110122, China
| | - Xueru Wang
- School of Life Sciences, China Medical University, Shenyang 110122, China
| | - Jingyu Chen
- Department of Physiology, China Medical University, Shenyang 110122, China
| | - Ziyu Guo
- School of Life Sciences, China Medical University, Shenyang 110122, China
| | - Bingqing Qiao
- School of Life Sciences, China Medical University, Shenyang 110122, China
| | - Ling Qin
- School of Life Sciences, China Medical University, Shenyang 110122, China
| |
Collapse
|
50
|
Quan P, Mao T, Zhang X, Wang R, Lei H, Wang J, Liu W, Dinges DF, Jiang C, Rao H. Locus coeruleus microstructural integrity is associated with vigilance vulnerability to sleep deprivation. Hum Brain Mapp 2024; 45:e70013. [PMID: 39225144 PMCID: PMC11369684 DOI: 10.1002/hbm.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/29/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024] Open
Abstract
Insufficient sleep compromises cognitive performance, diminishes vigilance, and disrupts daily functioning in hundreds of millions of people worldwide. Despite extensive research revealing significant variability in vigilance vulnerability to sleep deprivation, the underlying mechanisms of these individual differences remain elusive. Locus coeruleus (LC) plays a crucial role in the regulation of sleep-wake cycles and has emerged as a potential marker for vigilance vulnerability to sleep deprivation. In this study, we investigate whether LC microstructural integrity, assessed by fractional anisotropy (FA) through diffusion tensor imaging (DTI) at baseline before sleep deprivation, can predict impaired psychomotor vigilance test (PVT) performance during sleep deprivation in a cohort of 60 healthy individuals subjected to a rigorously controlled in-laboratory sleep study. The findings indicate that individuals with high LC FA experience less vigilance impairment from sleep deprivation compared with those with low LC FA. LC FA accounts for 10.8% of the variance in sleep-deprived PVT lapses. Importantly, the relationship between LC FA and impaired PVT performance during sleep deprivation is anatomically specific, suggesting that LC microstructural integrity may serve as a biomarker for vigilance vulnerability to sleep loss.
Collapse
Affiliation(s)
- Peng Quan
- The First Dongguan Affiliated Hospital, School of Humanities and ManagementGuangdong Medical UniversityDongguanChina
- Center for Functional Neuroimaging, Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Tianxin Mao
- Center for Functional Neuroimaging, Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain‐Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and ManagementShanghai International Studies UniversityShanghaiChina
| | - Xiaocui Zhang
- Center for Functional Neuroimaging, Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ruosi Wang
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain‐Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and ManagementShanghai International Studies UniversityShanghaiChina
| | - Hui Lei
- Center for Functional Neuroimaging, Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jieqiong Wang
- Center for Functional Neuroimaging, Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Wanting Liu
- Center for Functional Neuroimaging, Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - David F. Dinges
- Chronobiology and Sleep InstituteUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Caihong Jiang
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain‐Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and ManagementShanghai International Studies UniversityShanghaiChina
| | - Hengyi Rao
- Center for Functional Neuroimaging, Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain‐Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and ManagementShanghai International Studies UniversityShanghaiChina
- Chronobiology and Sleep InstituteUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|