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
|
Geissler CF, Frings C, Domes G. The effects of stress on working-memory-related prefrontal processing: an fNIRS study. Stress 2025; 28:2472067. [PMID: 40034019 DOI: 10.1080/10253890.2025.2472067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/20/2025] [Indexed: 03/05/2025] Open
Abstract
Acute stress causes a shift from executive to automated behavior. A key executive function suffering from this shift is working memory. Working memory is mainly negatively affected in the first 10 and more than 25 minutes after acute stress. These phases coincide with increased central levels of noradrenaline and cortisol. Increased levels of both hormones can cause a relative deactivation in prefrontal areas related to working memory processing. However, so far, there is little research that investigates the complete relationship between acute stress and resulting changes in stress hormones, neural activation, and working memory processing, over time. In this study, we used functional near-infrared spectroscopy to measure prefrontal activity during an nback task in a stress (28 subjects, 7 female/21 male) and a control group (28 subjects, 10 female/18 male) once (20 minutes) before and twice (4 and 24 minutes) after a socially evaluated cold pressor test or a warm water control condition. Additionally, we regularly measured changes in salivary cortisol and α-amylase (a correlate of central noradrenaline) during the experiment. While salivary cortisol was increased starting 14 minutes after acute stress, no effect of stress on salivary α-amylase or working memory performance was found. On a neural level, we found a marginally stronger decline in 3-back-related prefrontal activity from the first to the third measurement point in the stress than in the control group. These results present tentative evidence for a negative effect of acute stress on working-memory-related prefrontal processing mediated by central cortisol levels.
Collapse
Affiliation(s)
| | - Christian Frings
- Institute for Cognitive & Affective Neuroscience, Trier University, Trier, Germany
- Department of Cognitive Psychology, Trier University, Trier, Germany
| | - Gregor Domes
- Institute for Cognitive & Affective Neuroscience, Trier University, Trier, Germany
- Department of Biological and Clinical Psychology, Trier University, Trier, Germany
| |
Collapse
|
3
|
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; 175:106233. [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] [MESH Headings] [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
|
4
|
Yang X, Fedumenti FT, Niethard N, Hallschmid M, Born J, Rauss K. Regulation of peripheral glucose levels during human sleep. Sleep 2025; 48:zsaf042. [PMID: 39987458 PMCID: PMC12163131 DOI: 10.1093/sleep/zsaf042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/09/2025] [Indexed: 02/25/2025] Open
Abstract
Studies in rats indicate that oscillatory signatures of memory processing during sleep, specifically hippocampal sharp wave-ripples, also regulate peripheral glucose concentration. Here, we examined whether there is a similar link between such signatures of memory processing and glucose regulation during sleep in healthy humans. We obtained polysomnographic recordings and continuous recordings of peripheral interstitial glucose levels (1 sample/minute) from 10 participants (5 females) during two consecutive nights. Temporal relationships between electroencephalography (EEG) events of interest and glucose levels were examined using cross-correlation functions and peri-event time histograms. Confirming the findings in rats, we found that sleep spindles, a core signature of sleep-dependent memory processing, were followed within 1-6 minutes by a robust decrease in glucose concentrations. By contrast, slow oscillation events hallmarking slow wave sleep were followed, with a lag of 5-11 minutes, by an increase in glucose levels. Transitions into rapid eye movement sleep were followed by a glucose decrease after 10-14 minutes, whereas awakenings and microarousals were linked to immediate glucose increases. These temporal relationships indicate a sleep-specific regulation of peripheral glucose concentrations that is linked to both signatures of sleep-dependent memory processing as well as the macro-architecture of sleep. They possibly reflect noradrenergic regulation of sympathetic activity via the brainstem locus coeruleus and may be of relevance in clinical conditions with concurrent disturbances of sleep and glucose regulation.
Collapse
Affiliation(s)
- Xuefeng Yang
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
- Graduate School of Neural and Behavioural Science, International Max Planck Research School, Tübingen, Germany
| | | | - Niels Niethard
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
- Department of Cognitive Sciences, University of California, Irvine, Irvine, CA, USA
| | - Manfred Hallschmid
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Tübingen, Germany
- German Center for Mental Health (DZPG), Tübingen, Germany
| | - Jan Born
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Tübingen, Germany
- German Center for Mental Health (DZPG), Tübingen, Germany
- Werner Reichardt Center for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - Karsten Rauss
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
5
|
Tran S, Barker GRI, Mathiasen ML, Aggleton JP, Warburton EC. Functional and Regional Specificity of Noradrenergic Signaling for Encoding and Retrieval of Associative Recognition Memory in the Rat. J Neurosci 2025; 45:e2408242025. [PMID: 40312130 PMCID: PMC12160406 DOI: 10.1523/jneurosci.2408-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/24/2025] [Accepted: 04/27/2025] [Indexed: 05/03/2025] Open
Abstract
Recognition of a familiar object in a novel location requires retrieval of the former object-place association and encoding of novel information. Such object-in-place (OiP) memory recruits a neural network including the hippocampus (HPC), medial prefrontal cortex (mPFC), and nucleus reuniens of the thalamus (NRe); however, the underlying cellular mechanisms are not understood. Locus ceruleus (LC) noradrenergic neurons signal novelty; thus here we focused on the contribution of LC-forebrain projections and noradrenaline (NA) receptor subtypes to OiP encoding compared with retrieval, using an arena-based OiP task in male rats. The NRe was found to receive a catecholaminergic input from LC, with the strongest innervation directed to rostral NRe. Interestingly optogenetic inactivation of the LC→NRe pathway impaired OiP retrieval but was without effect on encoding, while inactivation of the LC→HPC selectively impaired encoding. Consistent with this double dissociation, pharmacological blockade of NRe α1-adrenoreceptors selectively impaired memory retrieval, while blockade of HPC β-adrenoreceptors impaired encoding. Finally, pharmacological attenuation of noradrenergic signaling in the NRe and HPC through the infusion of the α2-adrenergic receptor agonist UK 14,304 impaired retrieval and encoding, respectively. Surprisingly, antagonism or agonism of adrenoreceptor subtypes in the mPFC had no effect on memory performance. Together these results reveal the importance of NA within the HPC and NRe for OiP, whereby selectivity of function is achieved via spatially distinct LC output projections and NA receptor subtypes consistent with a modular view of NA function. These results are also important in demonstrating the distinct neuronal mechanisms by which encoding and retrieval are achieved.
Collapse
Affiliation(s)
- Stephanie Tran
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Gareth R I Barker
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, United Kingdom
| | | | - John P Aggleton
- School of Psychology, Cardiff University, Cardiff CF10 3AT, United Kingdom
| | - Elizabeth Clea Warburton
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, United Kingdom
| |
Collapse
|
6
|
Xu C, Chen S, Sun W, Xu R, Yin X, Guo D, Yang Y. Plasmon-enhanced fluorescence sensor based on Au nanocages for sensitive detection of norepinephrine. Anal Chim Acta 2025; 1354:343995. [PMID: 40253060 DOI: 10.1016/j.aca.2025.343995] [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: 01/03/2025] [Revised: 03/06/2025] [Accepted: 03/29/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Norepinephrine (NE) as a crucial monoamine neurotransmitter in the central and sympathetic nervous system, plays an important role in different physiological and pathophysiological processes. Brain NE can modulate cerebrospinal fluid flux and neurovascular coupling, regulate cortical and hippocampal neuronal circuitry, and participate the immune system. In addition, the reduced concentration of NE in brain was currently deemed to be the internal reason of major depression. However, development of detection method of NE with high spatiotemporal resolution in living systems remains a great challenge. RESULTS Herein, a plasmon-enhanced fluorescence (PEF) sensor based on Au nanocages (Aucages) were designed and prepared for ultra-sensitive detection of NE. Aucages with porous walls, hollow interior and systematically tunable localized surface plasmon resonance (LSPR) wavelengths (536 nm, 654 nm, 754 nm) were prepared to obtain a highly fluorescent enhancement of Au nanoclusters (Au NCs). Moreover, polyethylene glycol (PEG) with different molecular weight (1000, 5000, 10000 Da) were applied to control the distance between the Aucages and Au NCs. 3D-FDTD simulation results indicated that the fluorescence enhancement was primarily due to the internal and external enhanced electric field effects of Aucages. This sensor was applied for the turn-on detection of NE in commonly used clinical injectable norepinephrine bitartrate with the recovery rate of 98.06-105.34 %. Meanwhile, real-time fluorescence imaging of NE in living pheochromocytoma (PC-12) cells was explored with a red-emitted fluorescence. SIGNIFICANCE This study first employed Aucages with more "hot spot" for red-emitted Au NCs to realize fluorescence enhancement. It provides a new method for the development of more sensitive, accurate and convenient analysis of NE in clinical drug analysis, cell monitor and metabolism study.
Collapse
Affiliation(s)
- Chenye Xu
- College of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, PR China
| | - Shilin Chen
- College of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, PR China
| | - Wen Sun
- College of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, PR China
| | - Rong Xu
- College of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, PR China
| | - Xiangyuan Yin
- College of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, PR China
| | - Dongsheng Guo
- College of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, PR China.
| | - Yaqiong Yang
- College of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, PR China.
| |
Collapse
|
7
|
Evans WR, Baskar SS, Vellore A, Costa ARCE, Jacob C, Ravoori S, Arigbe A, Huda R. Chemogenetic Control of Striatal Astrocytes Improves Parkinsonian Motor Deficits in Mice. Glia 2025; 73:1188-1202. [PMID: 39902809 PMCID: PMC12012328 DOI: 10.1002/glia.24679] [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: 05/24/2024] [Revised: 12/22/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Parkinson's disease (PD) is characterized by the degeneration of dopaminergic nigrostriatal inputs, which causes striatal network dysfunction and leads to pronounced motor deficits. Recent evidence highlights astrocytes as a potential local source for striatal neuromodulation. There is substantial evidence for norepinephrine-mediated recruitment of cortical astrocyte activity during movement and locomotion. However, it is unclear how astrocytes in the striatum, a region devoid of norepinephrine neuromodulatory inputs, respond during locomotion. Moreover, it remains unknown how dopamine loss affects striatal astrocyte activity and whether astrocyte activity regulates behavioral deficits in PD. We addressed these questions by performing astrocyte-specific calcium recordings and manipulations using in vivo fiber photometry and chemogenetics. We find that locomotion elicits astrocyte calcium activity over a slower timescale than neurons. Acute pharmacological blockade of dopamine receptors only moderately reduced locomotion-related astrocyte activity. Yet, unilateral dopamine depletion significantly attenuated astrocyte calcium responses. Chemogenetic stimulation of Gi-coupled receptors partially improved this functional astrocyte deficit in dopamine-lesioned mice. In parallel, chemogenetic manipulation restored asymmetrical motor deficits and moderately improved open-field exploratory behavior. Together, our results establish a novel role for functional striatal astrocyte signaling in modulating motor function in PD and highlight non-neuronal targets for potential PD therapeutics.
Collapse
Affiliation(s)
- Wesley R. Evans
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNew JerseyUSA
| | - Sindhuja S. Baskar
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNew JerseyUSA
| | - Angelica Vellore
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNew JerseyUSA
| | - Ana Raquel Castro E. Costa
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNew JerseyUSA
| | - Cynthia Jacob
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNew JerseyUSA
| | - Sanya Ravoori
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNew JerseyUSA
| | - Abimbola Arigbe
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNew JerseyUSA
| | - Rafiq Huda
- WM Keck Center for Collaborative Neuroscience, Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNew JerseyUSA
| |
Collapse
|
8
|
Tsuda S, Hou J, Thompson FJ, Bose PK. Traumatic brain injury-induced anxiety: Injury and plasticity of the central noradrenergic system. Exp Neurol 2025; 388:115182. [PMID: 39929384 DOI: 10.1016/j.expneurol.2025.115182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 03/17/2025]
Abstract
Long-term anxiety is a hallmark symptom following traumatic brain injury (TBI). Although the central noradrenergic system (CNAS) is known to play a critical role in anxiety by regulating the excitability of several intricately interconnected brain structures via its projections to them, critical questions remain regarding the nature and extent of TBI-induced neuroplastic alterations in the CNAS and how these alterations relate to anxiety disorders. Knowledge relative to these questions is pivotal to development and refinement of therapies for TBI-associated anxiety disorders, including post-traumatic stress disorder. To this end, this study was designed to determine the impacts of chronic TBI on neuroplasticity of the CNAS and their significance in anxiety disorders in a clinically relevant rodent model. A standardized weight-drop model was used to produce controlled impacts of mild-to-moderate TBI in rats. Following the elevated plus maze tests to longitudinally assess anxiety-like behavior at 2 and 18 weeks post-injury of TBI animals, brain tissues of naïve and TBI rats were coronally sectioned and immunostained for a noradrenergic (NA) marker (dopamine β-hydroxylase) and neuronal nuclei in the central NA production sites and critical anxiety-regulating brain structures. We discovered that TBI caused robust losses of NA cells in the locus coeruleus and NA innervation of the central nucleus of the amygdala, an emotional processing center. Conversely, TBI caused intense gains of NA cells in the A2/A1 cell groups and NA innervation of other major anxiety-regulating regions. These changes coincided with progressively elevated anxiety-like behavior. Possibly, NA properties of A2/A1 cells were upregulated to compensate for the TBI-induced severe cell losses in the locus coeruleus. We conclude that these bi-directional vast alterations in the CNAS following chronic TBI contribute to dysregulated anxiety and, in part, the pathophysiology of human post-traumatic stress disorder.
Collapse
Affiliation(s)
- Shigeharu Tsuda
- Department of Anesthesiology, University of Florida, 1600 SW Archer Rd m509, Gainesville, FL 32610, USA; Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville, FL 32608, USA
| | - Jiamei Hou
- Department of Anesthesiology, University of Florida, 1600 SW Archer Rd m509, Gainesville, FL 32610, USA; Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville, FL 32608, USA
| | - Floyd J Thompson
- Department of Neuroscience, University of Florida, 1149 Newell Dr, Gainesville, FL 32610, USA; Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville, FL 32608, USA
| | - Prodip K Bose
- Department of Anesthesiology, University of Florida, 1600 SW Archer Rd m509, Gainesville, FL 32610, USA; Department of Neurology, University of Florida, 1149 Newell Dr, Gainesville, FL 32611, USA; Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd, Gainesville, FL 32608, USA.
| |
Collapse
|
9
|
Giofrè F, Lugaresi A, Baccari F, Lui E, Roberts S, Malpas C, Kalincik T. The topography of infratentorial lesions in depression and anxiety in multiple sclerosis. J Neurol 2025; 272:420. [PMID: 40418350 DOI: 10.1007/s00415-025-13167-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Depression and anxiety are highly prevalent in multiple sclerosis and significantly impact patient outcomes. However, their link to specific areas of demyelination remains unclear. OBJECTIVES This study examines the association between infratentorial lesions and depression or anxiety, focusing on three regions of interest: raphe nuclei, locus coeruleus, and cerebellar lobule VIIA. METHODS Patients were recruited from the cognitive neuroimmunology clinic at the Royal Melbourne Hospital. Participants were categorised as belonging to the groups 'depression'/'no depression' and 'anxiety'/'no anxiety' based on SPECTRA Indices of Psychopathology. MRI scans were examined for lesion presence in regions of interest. Association analyses were performed using multivariable logistic regression, adjusting for demographics, clinical parameters, and therapy. RESULTS Of 73 patients, 21 (29%) had clinically relevant depressive symptoms, and 18 (25%) had anxiety. Depression was significantly associated with lesions in the raphe nuclei (47.6% vs. 17.3%, OR 10.5, 95%CI 1.9-57.5, p=0.007) and locus coeruleus (38.1% vs. 15.4%, OR 20.5, 95%CI 2.3-184, p=0.007). Anxiety showed a potential association with locus coeruleus lesions (38.9% vs. 16.4%, OR 8.62, 95%CI 0.9-79.2, p=0.057). CONCLUSIONS Depression in multiple sclerosis is associated with lesions within serotoninergic and noradrenergic brainstem nuclei. No definitive anatomical substrate for anxiety was identified. These findings suggest that inflammatory structural changes may underlie mood disorders in multiple sclerosis, potentially serving as early imaging markers of susceptibility to depression.
Collapse
Affiliation(s)
- Federica Giofrè
- Dipartimento di Scienze Mediche e Chirurgiche, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Alessandra Lugaresi
- UOSI Riabilitazione Sclerosi Multipla, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Flavia Baccari
- Unità Operativa di Epidemiologia e Statistica, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Elaine Lui
- Department of Radiology, The University of Melbourne, Melbourne, Australia
- Department of Medical Imaging, The Royal Melbourne Hospital, Melbourne, Australia
| | - Stefanie Roberts
- CORe, Department of Medicine, The University of Melbourne, Melbourne, Australia
- Department of Neurology, Neuroimmunology Centre, The Royal Melbourne Hospital, Melbourne, Australia
| | - Charles Malpas
- CORe, Department of Medicine, The University of Melbourne, Melbourne, Australia
- Department of Neurology, Neuroimmunology Centre, The Royal Melbourne Hospital, Melbourne, Australia
| | - Tomas Kalincik
- CORe, Department of Medicine, The University of Melbourne, Melbourne, Australia.
- Department of Neurology, Neuroimmunology Centre, The Royal Melbourne Hospital, Melbourne, Australia.
| |
Collapse
|
10
|
Salvi V, Courtand G, de Deurwaerdère P, Cardoit L, Valerio S, Delcasso S, Georges F, Michelet T. Cingulate cortex stimulation drives distinct pupillary responses in rat via recruitment of noradrenergic neurons in the locus coeruleus. Cereb Cortex 2025; 35:bhaf085. [PMID: 40315430 DOI: 10.1093/cercor/bhaf085] [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: 01/22/2025] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 05/04/2025] Open
Abstract
The organization of the cingulate cortex has been the subject of intensive studies, concluding to its central role in motor control, cognition, and arousal. One of the key anatomical pathways through which the cingulate cortex influences behavior is its efferent connection to the locus coeruleus (LC). This brainstem region is responsible for noradrenaline (NA) release and is critical for various cognitive and behavioral functions. However, the specific impact of cingulate subregions on the LC-NA system remains unexplored. This study investigated how the different cingulate cortex areas affect LC-NA activity by measuring pupil-evoked responses (PERs) as an index of LC-NA activity. Using intra-cortical stimulation across the eight cingulate areas in rats, we found that anterior cingulate cortex and midcingulate cortex subregions evoked rare autonomic responses but significant pupil dilations whose amplitude increased along the caudo-rostral and dorso-ventral axes. By using the DSP-4, a neurotoxin-selective ablation of the LC-NA system, we suppressed PER and confirmed the role of LC-NA activity in this response. The differential influence of cortical areas on the PER demonstrates that each subregion of the rat cingulate cortex has the potential to differentially activate the LC-NA system, suggesting a clear parcellation of the rodent cingulate cortex, likely corresponding to functional specialization.
Collapse
Affiliation(s)
- Vianney Salvi
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, 2 rue Dr. Hoffmann Martinot, 33000 Bordeaux, France
| | - Gilles Courtand
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, 2 rue Dr. Hoffmann Martinot, 33000 Bordeaux, France
| | | | - Laura Cardoit
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, 2 rue Dr. Hoffmann Martinot, 33000 Bordeaux, France
| | | | | | - François Georges
- Univ. Bordeaux, CNRS, IMN, UMR 5293, 146 rue Leo Saignat - CS 61292 33076 Bordeaux cedex, France
| | - Thomas Michelet
- Univ. Bordeaux, CNRS, INCIA, UMR 5287, 2 rue Dr. Hoffmann Martinot, 33000 Bordeaux, France
| |
Collapse
|
11
|
Shen J, Li X, Zhong Y, Zhang J, Qin H, Chen F, Zhao X. Neuroendocrine characterization into schizophrenia: norepinephrine and melatonin as promising biomarkers. Front Endocrinol (Lausanne) 2025; 16:1551172. [PMID: 40375951 PMCID: PMC12078152 DOI: 10.3389/fendo.2025.1551172] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/07/2025] [Indexed: 05/18/2025] Open
Abstract
Background Although brain-derived neurotrophic factor (BDNF)has garnered extensive attention as a neuroendocrine marker in schizophrenia (SZ), its clinical utility remains limite due to inconsistent findings. Methods To address this gap, serum samples were collected from 24 female patients with SZ and 25 healthy controls. The metabolic profiling was performed using gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-mass spectrometry (LC-MS) to capture abroad range of metabolites. Results Our results revealed that BDNF is not a robust discriminatory biomarker. Marked differences in metabolic profiles were identified between patients with SZ and healthy individuals. The GC-MS analysis revealed significant differences in 79 metabolites; while the LC-MS analysis identified 419 significantly differential metabolites. Functional analysis reveals that these differential metabolites predominantly contribute to metabolic and neuro-related processes. Our findings demonstrate that norepinephrine and melatonin, two additional neuroendocrine compounds, are significantly elevated in patients with SZ compared to healthy controls. Notably, their higher areas under the curve (AUC) values compared to BDNF highlight their potential as more reliable biomarkers for SZ. Conclusion This study offers valuable insights into the altered metabolic patterns of female patients with SZ and establishes melatonin and norepinephrine as promising neuroendocrine biomarkers, underscoring their diagnostic value and role in the neuroendocrine regulation of mental disorders.
Collapse
Affiliation(s)
- Junwei Shen
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, Shanghai, China
| | - Xin Li
- Department of Physical Examination Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yinghua Zhong
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, Shanghai, China
| | - Jiechun Zhang
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, Shanghai, China
| | - Hongyun Qin
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, Shanghai, China
| | - Fazhan Chen
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, Shanghai, China
| | - Xudong Zhao
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
12
|
Koevoet D, Van Zantwijk L, Naber M, Mathôt S, van der Stigchel S, Strauch C. Effort drives saccade selection. eLife 2025; 13:RP97760. [PMID: 40193176 PMCID: PMC11975373 DOI: 10.7554/elife.97760] [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: 04/09/2025] Open
Abstract
What determines where to move the eyes? We recently showed that pupil size, a well-established marker of effort, also reflects the effort associated with making a saccade ('saccade costs'). Here, we demonstrate saccade costs to critically drive saccade selection: when choosing between any two saccade directions, the least costly direction was consistently preferred. Strikingly, this principle even held during search in natural scenes in two additional experiments. When increasing cognitive demand experimentally through an auditory counting task, participants made fewer saccades and especially cut costly directions. This suggests that the eye-movement system and other cognitive operations consume similar resources that are flexibly allocated among each other as cognitive demand changes. Together, we argue that eye-movement behavior is tuned to adaptively minimize saccade-inherent effort.
Collapse
Affiliation(s)
- Damian Koevoet
- Experimental Psychology, Helmholtz Institute, Utrecht UniversityUtrechtNetherlands
| | - Laura Van Zantwijk
- Experimental Psychology, Helmholtz Institute, Utrecht UniversityUtrechtNetherlands
| | - Marnix Naber
- Experimental Psychology, Helmholtz Institute, Utrecht UniversityUtrechtNetherlands
| | - Sebastiaan Mathôt
- Department of Psychology, University of GroningenGroningenNetherlands
| | | | - Christoph Strauch
- Experimental Psychology, Helmholtz Institute, Utrecht UniversityUtrechtNetherlands
| |
Collapse
|
13
|
Liu Y, Zhou ZX, Lv Q, Huang G, Zhang H, Wang YQ, Chen JG, Wang F. A superior colliculus-originating circuit prevents cocaine reinstatement via VR-based eye movement desensitization treatment. Natl Sci Rev 2025; 12:nwae467. [PMID: 40160681 PMCID: PMC11951104 DOI: 10.1093/nsr/nwae467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/24/2024] [Accepted: 11/26/2024] [Indexed: 04/02/2025] Open
Abstract
While Virtual Reality (VR) technology shows promise in the management of substance use disorders, the development of an effective VR-based extinction procedure remains lacking. In this study, we developed a VR-based eye movement desensitization and reprocessing extinction training program tailored for mice. We found that this VR treatment during cocaine extinction prevents reinstatement by suppressing the hyperactivation of glutamatergic excitatory neurons in the intermediate layers of the superior colliculus (SCiCaMKIIα) during exposure to environmental cues. Additionally, SCiCaMKIIα neurons innervate tyrosine hydroxylase-positive neurons in the locus coeruleus (LCTH). Environmental cues trigger stronger phasic activation of LCTH neurons through this SCiCaMKIIα→LCTH projection, leading to increased dopamine release onto the dorsal CA3 (dCA3) region, thereby facilitating reinstatement. Furthermore, we demonstrate that VR treatment effectively inhibits the neural circuitry involving SCiCaMKIIα→LCTH→dCA3 in response to environmental cues, thus preventing cocaine reinstatement. Our findings suggest that VR treatment may represent a promising strategy for achieving drug abstinence.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zi-Xiang Zhou
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiu Lv
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guan Huang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Han Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ye-Qin Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian-Guo Chen
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Shizhen Laboratory, Wuhan 430030, China
| | - Fang Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Shizhen Laboratory, Wuhan 430030, China
| |
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
|
Fiorentini G, Massé E, Ficarella SC, Torromino G. Peripheral transcutaneous electrical stimulation to improve cognition: a review of the main effects in healthy humans and in mildly cognitively impaired patient populations. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111290. [PMID: 39938732 DOI: 10.1016/j.pnpbp.2025.111290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
Peripheral nerve stimulation (PNS) is an ancient technique, up to now mainly used for pain management. The least invasive approach for PNS is transcutaneous electrical stimulation (TENS), which is performed by delivering mild electric currents through the skin and, depending on the stimulation pattern, activates the somatosensory Aβ-, Aδ- and C-fibers. In addition to its use for pain relief, accumulating data indicates that TENS can have broad-spectrum cognitive effects through the activation of neuromodulatory brain pathways. This review aims to summarize the current evidence on the cognitive effects of TENS, from healthy participants and mildly cognitively affected patients. Most studies on this topic have investigated the effects of TENS on memory, while fewer studies have explored attention, executive functions, and verbal fluency. Overall, promising evidence suggests that TENS may exert positive effects on specific cognitive functions. Further research is needed to build consensus on the most effective stimulation protocols, for both neurorehabilitation and enhancement, and to better understand the neurobiological mechanisms underlying the cognitive effects of TENS.
Collapse
Affiliation(s)
- Giulia Fiorentini
- Department of Humanistic Studies, University of Naples Federico II, Naples, Italy
| | - Eva Massé
- Information Processing and Systems, Office National d'Etudes et de Recherches Aérospatiales, Salon de Provence, France; Institut de Neurosciences de la Timone, CNRS & Aix-Marseille Université, Marseille, France
| | - Stefania C Ficarella
- Information Processing and Systems, Office National d'Etudes et de Recherches Aérospatiales, Salon de Provence, France
| | - Giulia Torromino
- Department of Humanistic Studies, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
16
|
Kamate M, Teranishi H, Umeda R, Shikano K, Kitaoka S, Hanada T, Hikida T, Kawano K, Hanada R. Dietary texture-driven masticatory activity and its impact on stress tolerance. J Oral Biosci 2025; 67:100628. [PMID: 39923995 DOI: 10.1016/j.job.2025.100628] [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: 02/06/2025] [Accepted: 01/26/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVES Although previous studies suggest that dietary texture-driven masticatory activity is correlated with stress tolerance, the underlying mechanisms, including neurotransmitter dynamics, remain unclear. This study investigated the effects of dietary texture-driven masticatory activity on stress tolerance in mice. METHODS Behavioral responses to stress were assessed using the repeated social defeat stress (R-SDS) and social interaction test (SIT) model. Neurotransmitter levels in stress-related brain regions were analyzed in mice fed a solid diet (promoting masticatory activity) or a powdered diet (decreasing masticatory activity). RESULTS Mice fed the powdered diet exhibited reduced stress tolerance compared with those fed the solid diet. Following the R-SDS, the powdered diet group displayed elevated gamma-aminobutyric acid (GABA) and norepinephrine levels in the prefrontal cortex. Before stress treatment, glutamic acid levels increased and those of choline decreased in the amygdala, whereas dopamine levels decreased in the powdered diet group after the R-SDS. In the locus coeruleus, mice on the powdered diet showed decreased glutamic acid and adenosine levels, alongside increased GABA levels. Serotonin levels decreased in the powdered diet group after the R-SDS, with no changes observed after the SIT. In the ventral hippocampus, GABA levels increased in the powdered diet group but decreased after the SIT. CONCLUSIONS This study demonstrates a correlation between masticatory activity and stress tolerance, evidenced by both behavioral and neurotransmitter changes. These findings suggest that reduced masticatory activity due to dietary texture contributes to decreased stress resilience.
Collapse
Affiliation(s)
- Mie Kamate
- Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan; Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Hitoshi Teranishi
- Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Ryohei Umeda
- Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan; Department of Advanced Medical Science, Faculty of Medicine, Oita University, Oita, Japan
| | - Kenshiro Shikano
- Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Shiho Kitaoka
- Department of Pharmacology, School of Medicine, Hyogo Medical University, Nishinomiya, Japan
| | - Toshikatsu Hanada
- Department of Cell Biology, Faculty of Medicine, Oita University, Oita, Japan
| | - Takatoshi Hikida
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Kenji Kawano
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Reiko Hanada
- Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan.
| |
Collapse
|
17
|
Galgani A, Scotto M, Faraguna U, Giorgi FS. Fading Blue: Exploring the Causes of Locus Coeruleus Damage Across the Lifespan. Antioxidants (Basel) 2025; 14:255. [PMID: 40227216 PMCID: PMC11939699 DOI: 10.3390/antiox14030255] [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: 01/31/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 04/15/2025] Open
Abstract
Locus Coeruleus (LC) is a brain nucleus that is involved in a variety of key functions (ranging from attention modulation to sleep-wake cycle regulation, to memory encoding); its proper function is necessary both during brain development and for brain integrity maintenance, and both at the microscale and macroscale level. Due to their specific intrinsic and extrinsic features, LC cells are considered particularly susceptible to damage concerning a variety of insults. This explains LC involvement in degenerative diseases not only in adults (in the context of neurodegenerative disease, mainly), but also in children (in relation to early hypoxic damage and Down's Syndrome, among others). In this narrative review, we dissect the potential mechanisms through which LC is affected in different diseases, with a special emphasis on the high rate of activity it is subjected to and the oxidative stress associated with it. Further research aimed at deepening our understanding of these mechanisms is needed to enable the development of potential strategies in the future that could slow down LC degeneration in subjects predisposed to specific brain disorders.
Collapse
Affiliation(s)
- Alessandro Galgani
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56125 Pisa, Italy
| | - Marco Scotto
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56125 Pisa, Italy
- Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Ugo Faraguna
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56125 Pisa, Italy
- I.R.C.C.S. Stella Maris, Calambrone, 56128 Pisa, Italy
| | - Filippo S. Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56125 Pisa, Italy
- I.R.C.C.S. Stella Maris, Calambrone, 56128 Pisa, Italy
| |
Collapse
|
18
|
Varga AG, Reid BT, Maletz SN, Dossat AM, Levitt ES. Opposing control of the respiratory brainstem on multiple timescales achieved by transmitter co-release from the locus coeruleus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.21.639476. [PMID: 40027822 PMCID: PMC11870594 DOI: 10.1101/2025.02.21.639476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The locus coeruleus (LC) provides widespread noradrenergic (NAergic) modulation throughout the brain to influence a wide range of functions, including breathing. Although both anatomical and physiological evidence supports the involvement of the LC in both the upstream integration and the downstream modulation of breathing, the circuitry behind the latter is unknown. Here, we show that NAergic LC neurons send projections to the Kӧlliker-Fuse nucleus (KF), a critical site in the control of breathing. Long duration activation of NAergic LC neuron terminals in pontine slices induces persistent inhibitory and excitatory NA currents or increases firing rate in postsynaptic KF neurons. Short stimulation on the other hand leads to the VGluT2-dependent release of glutamate that may be co-released with NA in a monosynaptic circuit. Together these results demonstrate that LC neurons can exert flexible, opposing effects on different timescales via glutamatergic and NAergic signaling onto a key respiratory brainstem nucleus.
Collapse
|
19
|
Carrasco RA, Breen KM. Brainstem Noradrenergic Neuronal Populations: Dual Effects on Regulating GnRH and LH Secretion. Endocrinology 2025; 166:bqaf021. [PMID: 39891672 PMCID: PMC11815497 DOI: 10.1210/endocr/bqaf021] [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: 10/31/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/03/2025]
Abstract
Noradrenergic neurons are a brain network that integrate viscero-sensorial signals to modulate neural and neuroendocrine function. Although it has been known for decades that noradrenergic neural circuits influence neuroendocrine and reproductive function, the cellular and molecular players involved remain largely unknown. The objective of this review is to summarize past and current knowledge regarding the influence of brainstem noradrenergic systems on GnRH and gonadotrophin secretion. The main noradrenergic cell groups A1, A2, and A6, known as the ventrolateral medulla, nucleus of the solitary tract, and locus coeruleus, respectively, are involved in the control of reproductive neuroendocrine secretion. Current evidence suggests that brainstem noradrenergic circuits promote the generation and maintenance of the LH surge in both spontaneous (rats, sheep) and induced (rabbit, ferret) ovulators. In contrast, recent studies have established that LH pulsatile secretion is suppressed by specific activation of brainstem noradrenergic cell groups. The duality of the GnRH/LH response to noradrenaline reflects the inherent complexity of hindbrain noradrenaline neurons, which are responsive to stressors and gonadal steroids (ie, estradiol) and coexpress a variety of neurotransmitters and neuropeptides. Therefore, elucidating the organization and functionality of brainstem noradrenergic systems will provide targets for controlling reproduction and understanding the interconnection with stress.
Collapse
Affiliation(s)
- Rodrigo A Carrasco
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093-0674, USA
| | - Kellie M Breen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093-0674, USA
| |
Collapse
|
20
|
Nigro M, Tortorelli LS, Garad M, Zlebnik NE, Yang H. Locus coeruleus modulation of single-cell representation and population dynamics in the mouse prefrontal cortex during attentional switching. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.13.571356. [PMID: 38168151 PMCID: PMC10760137 DOI: 10.1101/2023.12.13.571356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Behavioral flexibility, the ability to adjust behavioral strategies in response to changing environmental contingencies and internal demands, is fundamental to cognitive functions. Despite a large body of pharmacology and lesion studies, the precise neurophysiological mechanisms that underlie behavioral flexibility are still under active investigations. This work is aimed to determine the role of a brainstem-to-prefrontal cortex circuit in flexible rule switching. We trained mice to perform a set-shifting task, in which they learned to switch attention to distinguish complex sensory cues. Using chemogenetic inhibition, we selectively targeted genetically-defined locus coeruleus (LC) neurons or their input to the medial prefrontal cortex (mPFC). We revealed that suppressing either the LC or its mPFC projections severely impaired switching behavior, establishing the critical role of the LC-mPFC circuit in supporting attentional switching. To uncover the neurophysiological substrates of the behavioral deficits, we paired endoscopic calcium imaging of the mPFC with chemogenetic inhibition of the LC in task-performing mice. We found that mPFC prominently responded to attentional switching and that LC inhibition not only enhanced the engagement of mPFC neurons but also broadened single-neuron tuning in the task. At the population level, LC inhibition disrupted mPFC dynamic changes and impaired the encoding capacity for switching. Our results highlight the profound impact of the ascending LC input on modulating prefrontal dynamics and provide new insights into the cellular and circuit-level mechanisms that support behavioral flexibility.
Collapse
Affiliation(s)
- Marco Nigro
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA 92521, USA
| | - Lucas Silva Tortorelli
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA 92521, USA
| | - Machhindra Garad
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA 92521, USA
| | - Natalie E Zlebnik
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
- Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA
| | - Hongdian Yang
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA 92521, USA
- Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA
| |
Collapse
|
21
|
Akyuz E, Arulsamy A, Aslan FS, Sarisözen B, Guney B, Hekimoglu A, Yilmaz BN, Retinasamy T, Shaikh MF. An Expanded Narrative Review of Neurotransmitters on Alzheimer's Disease: The Role of Therapeutic Interventions on Neurotransmission. Mol Neurobiol 2025; 62:1631-1674. [PMID: 39012443 PMCID: PMC11772559 DOI: 10.1007/s12035-024-04333-y] [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: 07/06/2023] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. The accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles are the key players responsible for the pathogenesis of the disease. The accumulation of Aβ plaques and tau affect the balance in chemical neurotransmitters in the brain. Thus, the current review examined the role of neurotransmitters in the pathogenesis of Alzheimer's disease and discusses the alterations in the neurochemical activity and cross talk with their receptors and transporters. In the presence of Aβ plaques and neurofibrillary tangles, changes may occur in the expression of neuronal receptors which in turn triggers excessive release of glutamate into the synaptic cleft contributing to cell death and neuronal damage. The GABAergic system may also be affected by AD pathology in a similar way. In addition, decreased receptors in the cholinergic system and dysfunction in the dopamine neurotransmission of AD pathology may also contribute to the damage to cognitive function. Moreover, the presence of deficiencies in noradrenergic neurons within the locus coeruleus in AD suggests that noradrenergic stimulation could be useful in addressing its pathophysiology. The regulation of melatonin, known for its effectiveness in enhancing cognitive function and preventing Aβ accumulation, along with the involvement of the serotonergic system and histaminergic system in cognition and memory, becomes remarkable for promoting neurotransmission in AD. Additionally, nitric oxide and adenosine-based therapeutic approaches play a protective role in AD by preventing neuroinflammation. Overall, neurotransmitter-based therapeutic strategies emerge as pivotal for addressing neurotransmitter homeostasis and neurotransmission in the context of AD. This review discussed the potential for neurotransmitter-based drugs to be effective in slowing and correcting the neurodegenerative processes in AD by targeting the neurochemical imbalance in the brain. Therefore, neurotransmitter-based drugs could serve as a future therapeutic strategy to tackle AD.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, International School of Medicine, University of Health Sciences, Istanbul, Turkey
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
| | | | - Bugra Sarisözen
- School of Medicine, Tekirdağ Namık Kemal University, Tekirdağ, Turkey
| | - Beyzanur Guney
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | | | - Beyza Nur Yilmaz
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, New South Wales, 2800, Australia.
| |
Collapse
|
22
|
Yan S, Lu J, Duan B, Zhu H, Tian T, Qin Y, Li Y, Zhu W. Genetic and neurochemical profiles underlying cortical morphometric vulnerability to Parkinson's disease. Brain Res Bull 2025; 221:111222. [PMID: 39855312 DOI: 10.1016/j.brainresbull.2025.111222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Increasing evidence has documented cortical involvement at all stages of PD. The local vulnerabilities within certain brain regions in PD have been previously demonstrated, whereas its underlying genetic and neurochemical factors remain unclear. This study aims to investigate the spatial spectrum of cortical atrophy in Parkinson's disease (PD) and link these variances in gray matter properties and curvature respectively to putative molecular pathways and neurotransmitter factors. METHODS We recruited 141 clinically diagnosed PD patients and 70 healthy controls. Cortical morphological abnormalities of PD were obtained by intergroup comparisons in gray matter properties metrics and curvature measurements. Then we performed gene-category enrichment and spatial correlation analyses to evaluate the specific correspondence between cortical alteration in PD and genetic expression from the Allen Human Brain Atlas and normative neurotransmitter atlases from Neuromaps. RESULTS We found decreased gray matter properties in temporal, somatomotor, cingulate and occipital cortices, decreased curvature measures in occipital, temporal and orbitofrontal cortices, and increased curvature measures in somatomotor, prefrontal and posterior parietal cortices for PD patients. The related genes were enriched for the glucose metabolism, mitochondrial function, and post-translational histone modifications processes. In addition, the serotonin and norepinephrine transporter devoted more to gray matter properties alterations while the dopamine, gamma-aminobutyric acid receptors, and norepinephrine transporter were strong contributors of curvature abnormalities in PD. CONCLUSIONS Collectively, the present study offered interpretation of cortical morphological alterations and the cortical pathogenic theory in PD from genetic and neurochemical perspectives, which inspire further research on new pharmacotherapeutic approaches.
Collapse
Affiliation(s)
- Su Yan
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Lu
- Department of CT & MRI, The First Affiliated Hospital, College of Medicine, Shihezi University, 107 North Second Road, Shihezi, China
| | - Bingfang Duan
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongquan Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tian Tian
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Qin
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanhao Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
23
|
Yang Y, Tao Y. Regenerating Locus Coeruleus-Norepinephrine (LC-NE) Function: A Novel Approach for Neurodegenerative Diseases. Cell Prolif 2025:e13807. [PMID: 39876531 DOI: 10.1111/cpr.13807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/16/2024] [Accepted: 01/03/2025] [Indexed: 01/30/2025] Open
Abstract
Pathological changes in the locus coeruleus-norepinephrine (LC-NE) neurons, the major source of norepinephrine (NE, also known as noradrenaline) in the brain, are evident during the early stages of neurodegenerative diseases (ND). Research on both human and animal models have highlighted the therapeutic potential of targeting the LC-NE system to mitigate the progression of ND and alleviate associated psychiatric symptoms. However, the early and widespread degeneration of the LC-NE system presents a significant challenge for direct intervention in ND. Recent advances in regenerative cell therapy offer promising new strategies for ND treatment. The regeneration of LC-NE from pluripotent stem cells (PSCs) could significantly broaden the scope of LC-NE-based therapies for ND. In this review, we delve into the fundamental background and physiological functions of LC-NE. Additionally, we systematically examine the evidence and role of the LC-NE system in the neuropathology of ND and psychiatric diseases over recent years. Notably, we focus on the significance of PSCs-derived LC-NE and its potential impact on ND therapy. A deeper understanding and further investigation into the regeneration of LC-NE function could pave the way for practical and effective treatments for ND.
Collapse
Affiliation(s)
- Yana Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Yunlong Tao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| |
Collapse
|
24
|
Singh A, Shim P, Naeem S, Rahman S, Lutfy K. Pituitary adenylyl cyclase-activating polypeptide modulates the stress response: the involvement of different brain areas and microglia. Front Psychiatry 2025; 15:1495598. [PMID: 39931196 PMCID: PMC11807976 DOI: 10.3389/fpsyt.2024.1495598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/06/2024] [Indexed: 02/13/2025] Open
Abstract
Stress is necessary for survival. However, chronic unnecessary stress exposure leads to cardiovascular, gastrointestinal and neuropsychiatric disorders. Thus, understanding the mechanisms involved in the initiation and maintenance of the stress response is essential since it may reveal the underpinning pathophysiology of these disorders and may aid in the development of medication to treat stress-mediated diseases. Pituitary adenylyl cyclase activating polypeptide (PACAP) and its receptors (PAC1, VPAC1 and VPAC2) are expressed in the hypothalamus and other brain areas as well as in the adrenal gland. Previous research has shown that this peptide/receptor system serves as a modulator of the stress response. In addition to modulating the stress response, this system may also be connected to its emerging role as neuroprotective against hypoxia, ischemia, and neurodegeneration. This article aims to review the literature regarding the role of PACAP and its receptors in the stress response, the involvement of different brain regions and microglia in PACAP-mediated modulation of the stress response, and the long-term adaptation to stress recognizable clinically as survival with resilience while manifested in anxiety, depression and other neurobehavioral disorders.
Collapse
Affiliation(s)
- Anika Singh
- College of Pharmacy, The University of Rhode Island, Kingston, RI, United States
| | - Paul Shim
- Department of Biological Sciences, California State Polytechnic University, Pomona, CA, United States
| | - Sadaf Naeem
- Institute of Pharmaceutical Sciences, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD, United States
| | - Kabirullah Lutfy
- College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
25
|
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
|
26
|
Geng C, Li R, Li S, Liu P, Peng Y, Liu C, Wang Z, Zhang H, Li A. Noradrenergic inputs from the locus coeruleus to anterior piriform cortex and the olfactory bulb modulate olfactory outputs. Nat Commun 2025; 16:260. [PMID: 39747920 PMCID: PMC11697270 DOI: 10.1038/s41467-024-55609-9] [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: 07/27/2023] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Norepinephrine (NE) released from locus coeruleus (LC) noradrenergic (NAergic) neurons plays a pivotal role in the regulation of olfactory behaviors. However, the precise circuits and receptor mechanisms underlying this function are not well understood. Here, in DBH-Cre mice model, we show that LC NAergic neurons project directly to both anterior piriform cortex (aPC) and the olfactory bulb (OB). By using pharmacological and optogenetic manipulations in vitro and in vivo, we found that NE reduces the excitability of aPC pyramidal neurons directly via α2 receptors and that it bidirectionally regulates the activity of OB mitral cells via modulation of inhibitory inputs. Activation of the NAergic projection reduced both spontaneous and odor-evoked activity in the aPC/OB in awake mice, enhanced the odor-decoding ability of the aPC, and decreased the odor-decoding ability of the OB. Furthermore, activation of LC-aPC/OB NAergic projections accelerated odor discrimination and specific inactivation of the LC-aPC/OB NAergic pathway impaired olfactory detection and discrimination. These findings identify the mechanism underlying NAergic modulation of the aPC/OB and elucidate its role in odor processing and olfactory behaviors.
Collapse
Affiliation(s)
- Chi Geng
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ruochen Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shan Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Penglai Liu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuxin Peng
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Changyu Liu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhen Wang
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Anan Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
27
|
Cheng C, Xue X, Jiao Y, Du M, Zhang M, Zeng X, Sun JB, Qin W, Deng H, Yang XJ. Can earlobe stimulation serve as a sham for transcutaneous auricular vagus stimulation? Evidence from an alertness study following sleep deprivation. Psychophysiology 2025; 62:e14744. [PMID: 39727264 DOI: 10.1111/psyp.14744] [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: 11/17/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024]
Abstract
Transcutaneous auricular vagus nerve stimulation (taVNS) has garnered increasing attention as a safe and effective peripheral neuromodulation technique in various clinical and cognitive neuroscience fields. However, there is ongoing debate about whether the commonly used earlobe control interferes with the objective assessment of taVNS regulatory effects. This study aims to further explore the regulatory effects of taVNS and earlobe stimulation (ES) on alertness levels and physiological indicators following 24 h of sleep deprivation (SD), based on previous findings that both taVNS and ES showed significant positive effects. The goal is to evaluate whether ES can serve as a neutral sham condition. Using a within-subject randomized experimental design involving 56 participants, we assessed alertness, heart rate variability (HRV), and salivary alpha-amylase (sAA) levels in the morning of the first day. After 24 h of SD and 30 min of either taVNS or ES intervention, these indicators were re-evaluated, and the changes in both groups were analyzed. The results indicated that both taVNS and ES improved alertness levels following SD. However, taVNS significantly increased sAA levels, indicating activation of the LC-NE system, whereas ES significantly increased HR and reduced HRV, promoting sympathetic nervous activity. Additionally, the regulatory effect of taVNS on the alertness showed a higher correlation with SD impairment. Although taVNS and ES may involve different and separable neuromodulation mechanisms, both can enhance alertness following SD. Future studies should carefully consider the potential regulatory effects of ES when using it as a sham condition in taVNS research.
Collapse
Affiliation(s)
- Chen Cheng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
| | - Xinxin Xue
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
| | - Yunyun Jiao
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
| | - Mengyu Du
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
| | - Mengkai Zhang
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
| | - Xiao Zeng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
- Guangzhou Institute of Technology, Xidian University, Xi'an, China
| | - Jin-Bo Sun
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
- Guangzhou Institute of Technology, Xidian University, Xi'an, China
| | - Wei Qin
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
- Guangzhou Institute of Technology, Xidian University, Xi'an, China
| | - Hui Deng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Center of Journal Publication, Xidian University, Xi'an, China
| | - Xue-Juan Yang
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
- Guangzhou Institute of Technology, Xidian University, Xi'an, China
| |
Collapse
|
28
|
Xie S, Miao X, Li G, Zheng Y, Li M, Ji E, Wang J, Li S, Cai R, Geng L, Feng J, Wei C, Li Y. Red-shifted GRAB acetylcholine sensors for multiplex imaging in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.22.627112. [PMID: 39763957 PMCID: PMC11703214 DOI: 10.1101/2024.12.22.627112] [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: 01/14/2025]
Abstract
The neurotransmitter acetylcholine (ACh) is essential in both the central and peripheral nervous systems. Recent studies highlight the significance of interactions between ACh and various neuromodulators in regulating complex behaviors. The ability to simultaneously image ACh and other neuromodulators can provide valuable information regarding the mechanisms underlying these behaviors. Here, we developed a series of red fluorescent G protein-coupled receptor activation-based (GRAB) ACh sensors, with a wide detection range and expanded spectral profile. The high-affinity sensor, rACh1h, reliably detects ACh release in various brain regions, including the nucleus accumbens, amygdala, hippocampus, and cortex. Moreover, rACh1h can be co-expressed with green fluorescent sensors in order to record ACh release together with other neurochemicals in various behavioral contexts using fiber photometry and two-photon imaging, with high spatiotemporal resolution. These new ACh sensors can therefore provide valuable new insights regarding the functional role of the cholinergic system under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Shu Xie
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- These authors contributed equally
| | - Xiaolei Miao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
- These authors contributed equally
| | - Guochuan Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- These authors contributed equally
| | - Yu Zheng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Mengyao Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - En Ji
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jinxu Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Shaochuang Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Ruyi Cai
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Lan Geng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing 102206, China
- National Biomedical Imaging Center, Peking University, Beijing 100871, China
| |
Collapse
|
29
|
Slavova D, Ortiz V, Blaise M, Bairachnaya M, Giros B, Isingrini E. Role of the locus coeruleus-noradrenergic system in stress-related psychopathology and resilience: Clinical and pre-clinical evidences. Neurosci Biobehav Rev 2024; 167:105925. [PMID: 39427811 DOI: 10.1016/j.neubiorev.2024.105925] [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: 07/05/2024] [Revised: 08/28/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Stressful events, from daily stressors to traumatic experiences, are common and occur at any age. Despite the high prevalence of trauma, not everyone develops stress-related disorders like major depressive disorder (MDD) and post-traumatic stress disorder (PTSD), a variation attributed to resilience, the ability to adapt and avoid negative consequences of significant stress. This review examines the locus coeruleus-norepinephrine (LC-NE) system, a critical component in the brain's stress response. It discusses the LC-NE system's anatomical and functional complexity and its role in individual variability in stress responses. How different etiological factors and stress modalities affect the LC-NE system, influencing both adaptive stress responses and psychopathologies, are discussed and supported by evidence from human and animal studies. It also explores molecular and cellular adaptations in the LC that contribute to resilience, including roles of neuropeptide, inflammatory cytokines, and genetic modulation, and addresses developmental and sex differences in stress vulnerability. The need for a multifaceted approach to understand stress-induced psychopathologies is emphasized and pave the way for more personalized interventions for stress-related disorders.
Collapse
Affiliation(s)
- Déa Slavova
- Université Paris Cité, INCC UMR 8002, CNRS, Paris F-75006, France
| | - Vanesa Ortiz
- Université Paris Cité, INCC UMR 8002, CNRS, Paris F-75006, France
| | - Maud Blaise
- Université Paris Cité, INCC UMR 8002, CNRS, Paris F-75006, France
| | - Marya Bairachnaya
- Douglas Research Center Institute, McGill University, Montreal, Canada
| | - Bruno Giros
- Université Paris Cité, INCC UMR 8002, CNRS, Paris F-75006, France; Douglas Research Center Institute, McGill University, Montreal, Canada
| | - Elsa Isingrini
- Université Paris Cité, INCC UMR 8002, CNRS, Paris F-75006, France.
| |
Collapse
|
30
|
Tod P, Varga A, Román V, Lendvai B, Pálkovács R, Sperlágh B, Vizi ES. Tetrabenazine, a vesicular monoamine transporter 2 inhibitor, inhibits vesicular storage capacity and release of monoamine transmitters in mouse brain tissue. Br J Pharmacol 2024; 181:5094-5109. [PMID: 39304979 DOI: 10.1111/bph.17348] [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: 08/04/2023] [Revised: 07/10/2024] [Accepted: 08/23/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND AND PURPOSE Tetrabenazine (TBZ), used for treating hyperkinetic disorders, inhibits vesicular monoamine transporter-2 (VMAT-2), which sequesters monoamines into vesicles for exocytosis. However, our knowledge of the effect of TBZ on monoaminergic transmission is limited. Herein, we provide neurochemical evidence regarding the effect of VMAT-2 inhibition on vesicular neurotransmitter release from the prefrontal cortex (PFC) and striatum (STR) (brain regions involved in characteristic TBZ treatment side effects). The interaction between TBZ and MDMA was also assessed regarding motor behaviour in mice. EXPERIMENTAL APPROACH Vesicular storage capacity and release of [3H]-noradrenaline ([3H]-NA), [3H]-dopamine ([3H]-DA), [3H]-serotonin ([3H]-5-HT), and [3H]-acetylcholine ([3H]-ACh) was studied in mouse PFC and STR ex vivo slice preparations using electrical field stimulation. Additionally, locomotor activity was assessed in vehicle-treated mice and compared with that of MDMA, TBZ, and co-administered animals (n = 6) using the LABORAS system. KEY RESULTS TBZ lowered the storage capacity and inhibited the vesicular release of [3H]-NA and [3H]-DA from the PFC, and [3H]-DA and [3H]-5-HT from the STR in a concentration-dependent manner. Unlike vesamicol (vesicular ACh uptake inhibitor), TBZ failed to inhibit the vesicular release of [3H]-ACh from the PFC. When the vesicular storage of the investigated monoamines was inhibited by TBZ in the PFC and STR, MDMA induced the release of transmitters through transporter reversal; MDMA dose dependently increased locomotor activity in vivo. CONCLUSION AND IMPLICATIONS Our observations provide neurochemical evidence explaining the mechanism of VMAT-2 inhibitors in the brain and support the involvement of dopaminergic and noradrenergic transmission in hyperkinetic movement disorders.
Collapse
Affiliation(s)
- Pál Tod
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Anita Varga
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Viktor Román
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Balázs Lendvai
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Roland Pálkovács
- Pharmacology and Drug Safety Research, Gedeon Richter Plc, Budapest, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - E Sylvester Vizi
- Laboratory of Molecular Pharmacology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| |
Collapse
|
31
|
Sudo M, Kitajima D, Takagi Y, Mochizuki K, Fujibayashi M, Costello JT, Ando S. Effects of voluntary exercise and electrical muscle stimulation on reaction time in the Go/No-Go task. Eur J Appl Physiol 2024; 124:3571-3581. [PMID: 39044028 DOI: 10.1007/s00421-024-05562-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Acute exercise improves cognitive performance. However, it remains unclear what triggers cognitive improvement. Electrical muscle stimulation (EMS) facilitates the examination of physiological changes derived from peripheral muscle contraction during exercise. Thus, we compared the effects of EMS and voluntary exercise at low- or moderate-intensity on reaction time (RT) in a cognitive task to understand the contribution of central and peripheral physiological factors to RT improvement. METHODS Twenty-four young, healthy male participants performed a Go/No-Go task before and after EMS/exercise. In the EMS condition, EMS was applied to the lower limb muscles. In the low-intensity exercise condition, the participants cycled an ergometer while maintaining their heart rate (HR) at the similar level during EMS. In the moderate-intensity exercise condition, exercise intensity corresponded to ratings of perceived exertion of 13/20. The natural log-transformed root mean square of successive differences between adjacent inter-beat (R-R) intervals (LnRMSSD), which predominantly reflects parasympathetic HR modulation, was calculated before and during EMS/exercise. RESULTS RT improved following moderate-intensity exercise (p = 0.002, Cohen' d = 0.694), but not following EMS (p = 0.107, Cohen' d = 0.342) and low-intensity exercise (p = 0.076, Cohen' d = 0.380). Repeated measures correlation analysis revealed that RT was correlated with LnRMSSD (Rrm(23) = 0.599, p = 0.002) in the moderate-intensity exercise condition. CONCLUSION These findings suggest that the amount of central neural activity and exercise pressor reflex may be crucial for RT improvement. RT improvement following moderate-intensity exercise may, at least partly, be associated with enhanced sympathetic nervous system activity.
Collapse
Affiliation(s)
- Mizuki Sudo
- Physical Fitness Research Institute, Meiji Yasuda Life Foundation of Health and Welfare, 150 Tobuki, Hachioji, Tokyo, 192-0001, Japan
| | - Daisuke Kitajima
- Faculty of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu, Tokyo, 182-8585, Japan
| | - Yoko Takagi
- Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu, Tokyo, 182-8585, Japan
| | - Kodai Mochizuki
- Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu, Tokyo, 182-8585, Japan
| | - Mami Fujibayashi
- Faculty of Agriculture, Setsunan University, 45-1 Nagaotoge-Cho, Hirakata, Osaka, 573-0101, Japan
| | - Joseph T Costello
- School of Psychology, Sport and Health Sciences, University of Portsmouth, Portsmouth, UK
| | - Soichi Ando
- Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu, Tokyo, 182-8585, Japan.
| |
Collapse
|
32
|
Zhou J, Lin M, Xu W. Individual differences in baseline eye movement indices: Examining the relationships between baseline pupil size, inhibitory control, and fixation stability. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2024; 24:1084-1095. [PMID: 39198300 DOI: 10.3758/s13415-024-01213-9] [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] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
The relationship among baseline pupil size, fixation stability, and inhibitory control were examined in this study. Participants performed a baseline eye measure in which they were instructed to stare at a fixation dot on screen for 2 min. Following the baseline eye measure, participants completed an antisaccade task to measure inhibitory control ability. We found a correlation between baseline pupil size variability and inhibitory control, as well as between fixation stability and inhibitory control. We showed that participants with better inhibitory control exhibited larger variability in pupil size, and those with better fixation stability showed superior inhibitory control ability. Overall, our results indicate that there are significant correlations between inhibitory control and baseline pupil size, as well as between inhibitory control and fixation stability.
Collapse
Affiliation(s)
- Junyi Zhou
- School of Physical Education and Sport Science, Fujian Normal University, 1 Keji Rd., Minhou District, Fuzhou, 350117, Fujian, China
| | - Min Lin
- School of Physical Education and Sport Science, Fujian Normal University, 1 Keji Rd., Minhou District, Fuzhou, 350117, Fujian, China
- Nanxing Middle School, 18 Binxi Rd., Shuitou Town, Nan'an City, Quanzhou, 362342, Fujian, China
| | - Wenxin Xu
- School of Physical Education and Sport Science, Fujian Normal University, 1 Keji Rd., Minhou District, Fuzhou, 350117, Fujian, China.
| |
Collapse
|
33
|
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
|
34
|
Asim M, Wang H, Waris A, Qianqian G, Chen X. Cholecystokinin neurotransmission in the central nervous system: Insights into its role in health and disease. Biofactors 2024; 50:1060-1075. [PMID: 38777339 PMCID: PMC11627476 DOI: 10.1002/biof.2081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
Cholecystokinin (CCK) plays a key role in various brain functions, including both health and disease states. Despite the extensive research conducted on CCK, there remain several important questions regarding its specific role in the brain. As a result, the existing body of literature on the subject is complex and sometimes conflicting. The primary objective of this review article is to provide a comprehensive overview of recent advancements in understanding the central nervous system role of CCK, with a specific emphasis on elucidating CCK's mechanisms for neuroplasticity, exploring its interactions with other neurotransmitters, and discussing its significant involvement in neurological disorders. Studies demonstrate that CCK mediates both inhibitory long-term potentiation (iLTP) and excitatory long-term potentiation (eLTP) in the brain. Activation of the GPR173 receptor could facilitate iLTP, while the Cholecystokinin B receptor (CCKBR) facilitates eLTP. CCK receptors' expression on different neurons regulates activity, neurotransmitter release, and plasticity, emphasizing CCK's role in modulating brain function. Furthermore, CCK plays a pivotal role in modulating emotional states, Alzheimer's disease, addiction, schizophrenia, and epileptic conditions. Targeting CCK cell types and circuits holds promise as a therapeutic strategy for alleviating these brain disorders.
Collapse
Affiliation(s)
- Muhammad Asim
- Department of NeuroscienceCity University of Hong KongKowloon TongHong Kong
- Department of Biomedical ScienceCity University of Hong KongKowloon TongHong Kong
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of SciencesPak Shek KokHong Kong
| | - Huajie Wang
- Department of NeuroscienceCity University of Hong KongKowloon TongHong Kong
| | - Abdul Waris
- Department of Biomedical ScienceCity University of Hong KongKowloon TongHong Kong
| | - Gao Qianqian
- Department of NeuroscienceCity University of Hong KongKowloon TongHong Kong
| | - Xi Chen
- Department of NeuroscienceCity University of Hong KongKowloon TongHong Kong
- Department of Biomedical ScienceCity University of Hong KongKowloon TongHong Kong
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of SciencesPak Shek KokHong Kong
| |
Collapse
|
35
|
Wahis J, Akkaya C, Kirunda AM, Mak A, Zeise K, Verhaert J, Gasparyan H, Hovhannisyan S, Holt MG. The astrocyte α1A-adrenoreceptor is a key component of the neuromodulatory system in mouse visual cortex. Glia 2024; 72:1955-1973. [PMID: 39001577 DOI: 10.1002/glia.24591] [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: 05/04/2023] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 11/15/2024]
Abstract
Noradrenaline (norepinephrine) is known to modulate many physiological functions and behaviors. In this study, we tested to what extent astrocytes, a type of glial cell, participate in noradrenergic signaling in mouse primary visual cortex (V1). Astrocytes are essential partners of neurons in the central nervous system. They are central to brain homeostasis, but also dynamically regulate neuronal activity, notably by relaying and regulating neuromodulator signaling. Indeed, astrocytes express receptors for multiple neuromodulators, including noradrenaline, but the extent to which astrocytes are involved in noradrenergic signaling remains unclear. To test whether astrocytes are involved in noradrenergic neuromodulation in mice, we employed both short hairpin RNA mediated knockdown as well as pharmacological manipulation of the major noradrenaline receptor in astrocytes, the α1A-adrenoreceptor. Using acute brain slices, we found that the astrocytic α1A-adrenoreceptor subtype contributes to the generation of large intracellular Ca2+ signals in visual cortex astrocytes, which are generally thought to underlie astrocyte function. To test if reduced α1A-adrenoreceptor signaling in astrocytes affected the function of neuronal circuits in V1, we used both patch-clamp and field potential recordings. These revealed that noradrenergic signaling through the astrocyte α1A-adrenoreceptor is important to not only modulate synaptic activity but also to regulate plasticity in V1, through the potentiation of synaptic responses in circuits involved in visual information processing.
Collapse
Affiliation(s)
- Jérôme Wahis
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
| | - Cansu Akkaya
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Andre M Kirunda
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Aline Mak
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Karen Zeise
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Jens Verhaert
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Hayk Gasparyan
- Department of Mathematics and Mechanics, Yerevan State University, Yerevan, Armenia
- Armenian Bioinformatics institute, Yerevan, Armenia
| | - Sargis Hovhannisyan
- Department of Mathematics and Mechanics, Yerevan State University, Yerevan, Armenia
- Armenian Bioinformatics institute, Yerevan, Armenia
| | - Matthew G Holt
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| |
Collapse
|
36
|
Verduzco-Mendoza A, Mota-Rojas D, Olmos-Hernández A, Avila-Luna A, García-García K, Gálvez-Rosas A, Hidalgo-Bravo A, Ríos C, Parra-Cid C, Montes S, García-López J, Ramos-Languren LE, Pérez-Severiano F, González-Piña R, Bueno-Nava A. Changes in Noradrenergic Synthesis and Dopamine Beta-Hydroxylase Activity in Response to Oxidative Stress after Iron-induced Brain Injury. Neurochem Res 2024; 49:3043-3059. [PMID: 39105899 DOI: 10.1007/s11064-024-04222-9] [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: 06/10/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/07/2024]
Abstract
Noradrenaline (NA) levels are altered during the first hours and several days after cortical injury. NA modulates motor functional recovery. The present study investigated whether iron-induced cortical injury modulated noradrenergic synthesis and dopamine beta-hydroxylase (DBH) activity in response to oxidative stress in the brain cortex, pons and cerebellum of the rat. Seventy-eight rats were divided into two groups: (a) the sham group, which received an intracortical injection of a vehicle solution; and (b) the injured group, which received an intracortical injection of ferrous chloride. Motor deficits were evaluated for 20 days post-injury. On the 3rd and 20th days, the rats were euthanized to measure oxidative stress indicators (reactive oxygen species (ROS), reduced glutathione (GSH) and oxidized glutathione (GSSG)) and catecholamines (NA, dopamine (DA)), plus DBH mRNA and protein levels. Our results showed that iron-induced brain cortex injury increased noradrenergic synthesis and DBH activity in the brain cortex, pons and cerebellum at 3 days post-injury, predominantly on the ipsilateral side to the injury, in response to oxidative stress. A compensatory increase in contralateral noradrenergic activity was observed, but without changes in the DBH mRNA and protein levels in the cerebellum and pons. In conclusion, iron-induced cortical injury increased the noradrenergic response in the brain cortex, pons and cerebellum, particularly on the ipsilateral side, accompanied by a compensatory response on the contralateral side. The oxidative stress was countered by antioxidant activity, which favored functional recovery following motor deficits.
Collapse
Affiliation(s)
- Antonio Verduzco-Mendoza
- Programa de Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México, Mexico
- Dirección de Investigación, Arenal de Guadalupe, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, Mexico
| | - Daniel Mota-Rojas
- División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana CBS, Unidad Xochimilco, Ciudad de México, Mexico
| | - Adriana Olmos-Hernández
- Dirección de Investigación, Arenal de Guadalupe, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, Mexico
| | - Alberto Avila-Luna
- Dirección de Investigación, Arenal de Guadalupe, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, Mexico
| | - Karla García-García
- Dirección de Investigación, Arenal de Guadalupe, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, Mexico
| | - Arturo Gálvez-Rosas
- Dirección de Investigación, Arenal de Guadalupe, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, Mexico
| | - Alberto Hidalgo-Bravo
- Dirección de Investigación, Arenal de Guadalupe, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, Mexico
| | - Camilo Ríos
- Dirección de Investigación, Arenal de Guadalupe, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, Mexico
- División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana CBS, Unidad Xochimilco, Ciudad de México, Mexico
| | - Carmen Parra-Cid
- Dirección de Investigación, Arenal de Guadalupe, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, Mexico
| | - Sergio Montes
- Unidad Académica Multidisciplinaria Reynosa-Aztlán, Universidad Autónoma de Tamaulipas, calle 16 y lago de Chapala, Aztlán, Tamaulipas, Mexico
| | - Julieta García-López
- Dirección de Investigación, Arenal de Guadalupe, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, Mexico
| | - Laura E Ramos-Languren
- Facultad de Psicología, División de Estudios Profesionales, Universidad Nacional Autónoma de Mexico, Av. Universidad 3040, Col, Copilco Universidad Alcaldía Coyoacán, Ciudad de México, Mexico
| | - Francisca Pérez-Severiano
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, SSa, Insurgentes Sur 3877, Ciudad de México, Mexico
| | - Rigoberto González-Piña
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Salvador Díaz Mirón esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, 11340, Ciudad de México, Mexico
- Clínica de Rehabilitación del Daño Cerebral CLIREDACE "Dr. Hugo Iván González Gutiérrez", Monterrey 243, Col. Roma Sur, Alcaldía Cuauhtémoc, Ciudad de México, Mexico
| | - Antonio Bueno-Nava
- Dirección de Investigación, Arenal de Guadalupe, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calzada México-Xochimilco 289, Arenal de Guadalupe, Ciudad de México, Mexico.
- Laboratorio de Neurofisiología Química de la Discapacidad, Coordinación de Neurociencias Básica, Arenal de Guadalupe, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, SSa, Calz. México-Xochimilco 289, Ciudad de México, 14389, Mexico.
| |
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
|
Özçete ÖD, Banerjee A, Kaeser PS. Mechanisms of neuromodulatory volume transmission. Mol Psychiatry 2024; 29:3680-3693. [PMID: 38789677 PMCID: PMC11540752 DOI: 10.1038/s41380-024-02608-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024]
Abstract
A wealth of neuromodulatory transmitters regulate synaptic circuits in the brain. Their mode of signaling, often called volume transmission, differs from classical synaptic transmission in important ways. In synaptic transmission, vesicles rapidly fuse in response to action potentials and release their transmitter content. The transmitters are then sensed by nearby receptors on select target cells with minimal delay. Signal transmission is restricted to synaptic contacts and typically occurs within ~1 ms. Volume transmission doesn't rely on synaptic contact sites and is the main mode of monoamines and neuropeptides, important neuromodulators in the brain. It is less precise than synaptic transmission, and the underlying molecular mechanisms and spatiotemporal scales are often not well understood. Here, we review literature on mechanisms of volume transmission and raise scientific questions that should be addressed in the years ahead. We define five domains by which volume transmission systems can differ from synaptic transmission and from one another. These domains are (1) innervation patterns and firing properties, (2) transmitter synthesis and loading into different types of vesicles, (3) architecture and distribution of release sites, (4) transmitter diffusion, degradation, and reuptake, and (5) receptor types and their positioning on target cells. We discuss these five domains for dopamine, a well-studied monoamine, and then compare the literature on dopamine with that on norepinephrine and serotonin. We include assessments of neuropeptide signaling and of central acetylcholine transmission. Through this review, we provide a molecular and cellular framework for volume transmission. This mechanistic knowledge is essential to define how neuromodulatory systems control behavior in health and disease and to understand how they are modulated by medical treatments and by drugs of abuse.
Collapse
Affiliation(s)
- Özge D Özçete
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
39
|
Majdi A, Asamoah B, Mc Laughlin M. Understanding novel neuromodulation pathways in tDCS: brain stem recordings in rats during trigeminal nerve direct current stimulation. Transl Psychiatry 2024; 14:456. [PMID: 39468008 PMCID: PMC11519445 DOI: 10.1038/s41398-024-03158-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
tDCS is widely assumed to cause neuromodulation via the electric field in the cortex acting directly on cortical neurons. However, recent evidence suggests that tDCS may indirectly influence brain activity through cranial nerve pathways, notably the trigeminal nerve, but these neuromodulatory pathways remain unexplored. To investigate the first stages in this potential pathway we developed an animal model to study the effect of trigeminal nerve direct current stimulation (TN-DCS) on neuronal activity in the principal sensory nucleus (NVsnpr) and the mesencephalic nucleus of the trigeminal nerve (MeV). We conducted experiments on twenty-four male Sprague Dawley rats (n = 10 NVsnpr, n = 10 MeV during anodic stimulation, and n = 4 MeV during cathodic stimulation). DC stimulation, ranging from 0.5 to 3 mA, targeted the trigeminal nerve's marginal branch. Concurrently, single-unit electrophysiological recordings were obtained using a 32-channel silicon probe, encompassing three 1-min intervals: pre, during, and post-stimulation. Xylocaine trigeminal nerve blockage served as a control. TN-DCS increased neuronal spiking activity in both NVsnpr and MeV, returning to baseline during the post-stimulation phase. The 3 mA DC stimulation of the blocked trigeminal nerve failed to induce increased spiking activity in the trigeminal nuclei. These findings provide empirical support for trigeminal nuclei modulation via TN-DCS, suggesting the cranial nerve pathways could play a role in mediating the tDCS effects in humans.
Collapse
Affiliation(s)
- Alireza Majdi
- Research Group Experimental Oto-rhino-laryngology, Department of Neuroscience, Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium
| | - Boateng Asamoah
- Research Group Experimental Oto-rhino-laryngology, Department of Neuroscience, Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium
| | - Myles Mc Laughlin
- Research Group Experimental Oto-rhino-laryngology, Department of Neuroscience, Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium.
- Leuven Brain Institute, KU Leuven, Leuven, 3000, Belgium.
| |
Collapse
|
40
|
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
|
41
|
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
|
42
|
Tanner J, Orthlieb G, Helms Tillery S. Effect of touch on proprioception: non-invasive trigeminal nerve stimulation suggests general arousal rather than tactile-proprioceptive integration. Front Hum Neurosci 2024; 18:1429843. [PMID: 39469503 PMCID: PMC11513270 DOI: 10.3389/fnhum.2024.1429843] [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: 05/08/2024] [Accepted: 09/09/2024] [Indexed: 10/30/2024] Open
Abstract
Introduction Proprioceptive error of estimated fingertip position in two-dimensional space is reduced with the addition of tactile stimulation applied at the fingertip. Tactile input does not disrupt the participants' estimation strategy, as the individual error vector maps maintain their overall structure. This relationship suggests integration of proprioception and tactile information improves proprioceptive estimation, which can also be improved with trained mental focus and attention. Task attention and arousal are physiologically regulated by the reticular activating system (RAS), a brainstem circuit including the locus coeruleus (LC). There is direct and indirect evidence that these structures can be modulated by non-invasive trigeminal nerve stimulation (nTNS), providing an opportunity to examine nTNS effect on the integrative relationship of proprioceptive and tactile information. Methods Fifteen right-handed participants performed a simple right-handed proprioceptive estimation task with tactile feedback (touch) and no tactile (hover) feedback. Participants repeated the task after nTNS administration. Stimulation was delivered for 10 min, and stimulation parameters were 3,000 Hz, 50 μs pulse width, with a mean of 7 mA. Error maps across the workspace are generated using polynomial models of the participants' target responses. Results Error maps did not demonstrate significant vector direction changes between conditions for any participant, indicating that nTNS does not disrupt spatial proprioception estimation strategies. A linear mixed model regression with nTNS epoch, tactile condition, and the interaction as factors demonstrated that nTNS reduced proprioceptive error under the hover condition only. Discussion We argue that nTNS does not disrupt spatial proprioceptive error maps but can improve proprioceptive estimation in the absence of tactile feedback. However, we observe no evidence that nTNS enhances tactile-proprioceptive integration as the touch condition does not exhibit significantly reduced error after nTNS.
Collapse
Affiliation(s)
- Justin Tanner
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| | | | | |
Collapse
|
43
|
Parent JH, Cassady K, Jagust WJ, Berry AS. Pathological and neurochemical correlates of locus coeruleus functional network activity. Biol Psychol 2024; 192:108847. [PMID: 39038634 PMCID: PMC11464174 DOI: 10.1016/j.biopsycho.2024.108847] [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: 03/19/2024] [Revised: 06/25/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024]
Abstract
The locus coeruleus (LC) produces the neuromodulators norepinephrine and dopamine, and projects widely to subcortical and cortical brain regions. The LC has been a focus of neuroimaging biomarker development for the early detection of Alzheimer's disease (AD) since it was identified as one of the earliest brain regions to develop tau pathology. Our recent research established the use of positron emission tomography (PET) to measure LC catecholamine synthesis capacity in cognitively unimpaired older adults. We extend this work by investigating the possible influence of pathology and LC neurochemical function on LC network activity using functional magnetic resonance imaging (fMRI). In separate sessions, participants underwent PET imaging to measure LC catecholamine synthesis capacity ([18F]Fluoro-m-tyrosine), tau pathology ([18F]Flortaucipir), and amyloid-β pathology ([11C]Pittsburgh compound B), and fMRI imaging to measure LC functional network activity at rest. Consistent with a growing body of research in aging and preclinical AD, we find that higher functional network activity is associated with higher tau burden in individuals at risk of developing AD (amyloid-β positive). Critically, relationships between higher LC network activity and higher pathology (amyloid-β and tau) were moderated by LC catecholamine synthesis capacity. High levels of LC catecholamine synthesis capacity reduced relationships between higher network activity and pathology. Broadly, these findings support the view that individual differences in functional network activity are shaped by interactions between pathology and neuromodulator function, and point to catecholamine systems as potential therapeutic targets.
Collapse
Affiliation(s)
- Jourdan H Parent
- Department of Psychology, Brandeis University, Waltham, MA 02453, USA.
| | - Kaitlin Cassady
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Anne S Berry
- Department of Psychology, Brandeis University, Waltham, MA 02453, USA; Volen Center for Complex Systems, Brandeis University, Waltham, MA 02453, USA
| |
Collapse
|
44
|
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
|
45
|
Manto M, Adamaszek M, Apps R, Carlson E, Guarque-Chabrera J, Heleven E, Kakei S, Khodakhah K, Kuo SH, Lin CYR, Joshua M, Miquel M, Mitoma H, Larry N, Péron JA, Pickford J, Schutter DJLG, Singh MK, Tan T, Tanaka H, Tsai P, Van Overwalle F, Yamashiro K. Consensus Paper: Cerebellum and Reward. CEREBELLUM (LONDON, ENGLAND) 2024; 23:2169-2192. [PMID: 38769243 DOI: 10.1007/s12311-024-01702-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/06/2024] [Indexed: 05/22/2024]
Abstract
Cerebellum is a key-structure for the modulation of motor, cognitive, social and affective functions, contributing to automatic behaviours through interactions with the cerebral cortex, basal ganglia and spinal cord. The predictive mechanisms used by the cerebellum cover not only sensorimotor functions but also reward-related tasks. Cerebellar circuits appear to encode temporal difference error and reward prediction error. From a chemical standpoint, cerebellar catecholamines modulate the rate of cerebellar-based cognitive learning, and mediate cerebellar contributions during complex behaviours. Reward processing and its associated emotions are tuned by the cerebellum which operates as a controller of adaptive homeostatic processes based on interoceptive and exteroceptive inputs. Lobules VI-VII/areas of the vermis are candidate regions for the cortico-subcortical signaling pathways associated with loss aversion and reward sensitivity, together with other nodes of the limbic circuitry. There is growing evidence that the cerebellum works as a hub of regional dysconnectivity across all mood states and that mental disorders involve the cerebellar circuitry, including mood and addiction disorders, and impaired eating behaviors where the cerebellum might be involved in longer time scales of prediction as compared to motor operations. Cerebellar patients exhibit aberrant social behaviour, showing aberrant impulsivity/compulsivity. The cerebellum is a master-piece of reward mechanisms, together with the striatum, ventral tegmental area (VTA) and prefrontal cortex (PFC). Critically, studies on reward processing reinforce our view that a fundamental role of the cerebellum is to construct internal models, perform predictions on the impact of future behaviour and compare what is predicted and what actually occurs.
Collapse
Affiliation(s)
- Mario Manto
- Service de Neurologie, Médiathèque Jean Jacquy, CHU-Charleroi, 6000, Charleroi, Belgium.
- Service Des Neurosciences, Université de Mons, 7000, Mons, Belgium.
- Unité Des Ataxies Cérébelleuses, CHU-Charleroi, Service Des Neurosciences, University of Mons, 7000, Mons, Belgium.
| | - Michael Adamaszek
- Department of Clinical and Cognitive Neurorehabilitation, Klinik Bavaria Kreischa, 01731, Kreischa, Germany
| | - Richard Apps
- School of Physiology, Pharmacology & Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Erik Carlson
- Department of Psychiatry and Behavioural Sciences, University of Washington, Seattle, WA, 98108, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA
| | - Julian Guarque-Chabrera
- Área de Psicobiología, Facultat de Ciències de La Salut, Universitat Jaume I, 12071, Castellón de La Plana, Spain
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, 10461, USA
| | - Elien Heleven
- Faculty of Psychology and Center for Neuroscience, Vrije Universiteit Brussel, 1050, Brussels, Belgium
| | - Shinji Kakei
- Department of Anatomy and Physiology, Jissen Women's University, Tokyo, 191-8510, Japan
| | - Kamran Khodakhah
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, 10461, USA
| | - Sheng-Han Kuo
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
- Initiative of Columbia Ataxia and Tremor, Columbia University Medical Center, New York, NY, 10032, USA
| | - Chi-Ying R Lin
- Alzheimer's Disease and Memory Disorders Center, Department of Neurology, Baylor College of Medicine, Houston, 77030 TX, USA
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, 77030 TX, USA
| | - Mati Joshua
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem, Israel
| | - Marta Miquel
- Área de Psicobiología, Facultat de Ciències de La Salut, Universitat Jaume I, 12071, Castellón de La Plana, Spain
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, 10461, USA
| | - Hiroshi Mitoma
- Department of Medical Education, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Noga Larry
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem, Israel
| | - Julie Anne Péron
- Clinical and Experimental Neuropsychology Laboratory, Department of Psychology and Educational Sciences, University of Geneva, 1205, Geneva, Switzerland
| | - Jasmine Pickford
- School of Physiology, Pharmacology & Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Dennis J L G Schutter
- Department of Experimental Psychology, Helmholtz Institute, Utrecht University, Heidelberglaan 1, 3584 CS, Utrecht, The Netherlands
| | - Manpreet K Singh
- Psychiatry and Behavioral Sciences, University of California Davis, 2230 Stockton Blvd, Sacramento, CA, 95817, USA
| | - Tommy Tan
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Hirokazu Tanaka
- Faculty of Information Technology, Tokyo City University, Tokyo, 158-8557, Japan
| | - Peter Tsai
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, 75235, USA
- Departments of Neuroscience, Pediatrics, Psychiatry, UT Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Frank Van Overwalle
- Faculty of Psychology and Center for Neuroscience, Vrije Universiteit Brussel, 1050, Brussels, Belgium
| | - Kunihiko Yamashiro
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, 75235, USA
| |
Collapse
|
46
|
Yu J, Ji S, Tao H, Shan X, Yan Y, Sun X, Tu X, Li L, Deng C. Cell-specific expression of Cre recombinase in rat noradrenergic neurons via CRISPR-Cas9 system. Neuroscience 2024; 556:31-41. [PMID: 39067682 DOI: 10.1016/j.neuroscience.2024.07.032] [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: 01/18/2024] [Revised: 05/01/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Noradrenergic neurons play a crucial role in the functioning of the nervous system. They formed compact small clusters in the central nervous system. To target noradrenergic neurons in combination with viral tracing and achieve cell-type specific functional manipulation using chemogenetic or optogenetic tools, new transgenic animal lines are needed, especially rat models for their advantages in large body size with facilitating easy operation, physiological parameter monitoring, and accommodating complex behavioral and cognitive studies. In this study, we successfully generated a transgenic rat strain capable of expressing Cre recombinase under the control of the dopamine beta-hydroxylase (DBH) gene promoter using the CRISPR-Cas9 system. Our validation process included co-immunostaining with Cre and DBH antibodies, confirming the specific expression of Cre recombinase. Furthermore, stereotaxic injection of a fluorescence-labeled AAV-DIO virus illustrated the precise Cre-loxP-mediated recombination activity in noradrenergic neurons within the locus coeruleus (LC). Through crossbreeding with the LSL-fluorescence reporter rat line, DBH-Cre rats proved instrumental in delineating the position and structure of noradrenergic neuron clusters A1, A2, A6 (LC), and A7 in rats. Additionally, our specific activation of the LC noradrenergic neurons showed effective behavioral readout using chemogenetics of this rat line. Our results underscore the effectiveness and specificity of Cre recombinase in noradrenergic neurons, serving as a robust tool for cell-type specific targeting of small-sized noradrenergic nuclei. This approach enhances our understanding of their anatomical, physiological, and pathological roles, contributing to a more profound comprehension of noradrenergic neuron function in the nervous system.
Collapse
Affiliation(s)
- Jingwei Yu
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Shuqin Ji
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Huixin Tao
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaochun Shan
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yu Yan
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xiangzhou Sun
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xiang'an Tu
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Lei Li
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 101408, China; CAS Key Laboratory of Brain Connectome and Manipulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Chunhua Deng
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
47
|
Ricou C, Rabadan V, Mofid Y, Aguillon-Hernandez N, Wardak C. Pupil dilation reflects the social and motion content of faces. Soc Cogn Affect Neurosci 2024; 19:nsae055. [PMID: 39167473 PMCID: PMC11403811 DOI: 10.1093/scan/nsae055] [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: 02/07/2024] [Revised: 07/15/2024] [Accepted: 08/19/2024] [Indexed: 08/23/2024] Open
Abstract
Human facial features (eyes, nose, and mouth) allow us to communicate with others. Observing faces triggers physiological responses, including pupil dilation. Still, the relative influence of social and motion content of a visual stimulus on pupillary reactivity has never been elucidated. A total of 30 adults aged 18-33 years old were recorded with an eye tracker. We analysed the event-related pupil dilation in response to stimuli distributed along a gradient of social salience (non-social to social, going from objects to avatars to real faces) and dynamism (static to micro- to macro-motion). Pupil dilation was larger in response to social (faces and avatars) compared to non-social stimuli (objects), with surprisingly a larger response for avatars. Pupil dilation was also larger in response to macro-motion compared to static. After quantifying each stimulus' real quantity of motion, we found that the higher the quantity of motion, the larger the pupil dilated. However, the slope of this relationship was not higher for social stimuli. Overall, pupil dilation was more sensitive to the real quantity of motion than to the social component of motion, highlighting the relevance of ecological stimulations. Physiological response to faces results from specific contributions of both motion and social processing.
Collapse
Affiliation(s)
- Camille Ricou
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, Tours 37032, France
| | - Vivien Rabadan
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, Tours 37032, France
| | - Yassine Mofid
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, Tours 37032, France
| | | | - Claire Wardak
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, Tours 37032, France
| |
Collapse
|
48
|
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
|
49
|
Tao Y, Li X, Dong Q, Kong L, Petersen AJ, Yan Y, Xu K, Zima S, Li Y, Schmidt DK, Ayala M, Mathivanan S, Sousa AMM, Chang Q, Zhang SC. Generation of locus coeruleus norepinephrine neurons from human pluripotent stem cells. Nat Biotechnol 2024; 42:1404-1416. [PMID: 37974010 PMCID: PMC11392812 DOI: 10.1038/s41587-023-01977-4] [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: 12/15/2020] [Accepted: 08/30/2023] [Indexed: 11/19/2023]
Abstract
Central norepinephrine (NE) neurons, located mainly in the locus coeruleus (LC), are implicated in diverse psychiatric and neurodegenerative diseases and are an emerging target for drug discovery. To facilitate their study, we developed a method to generate 40-60% human LC-NE neurons from human pluripotent stem cells. The approach depends on our identification of ACTIVIN A in regulating LC-NE transcription factors in dorsal rhombomere 1 (r1) progenitors. In vitro generated human LC-NE neurons display extensive axonal arborization; release and uptake NE; and exhibit pacemaker activity, calcium oscillation and chemoreceptor activity in response to CO2. Single-nucleus RNA sequencing (snRNA-seq) analysis at multiple timepoints confirmed NE cell identity and revealed the differentiation trajectory from hindbrain progenitors to NE neurons via an ASCL1-expressing precursor stage. LC-NE neurons engineered with an NE sensor reliably reported extracellular levels of NE. The availability of functional human LC-NE neurons enables investigation of their roles in psychiatric and neurodegenerative diseases and provides a tool for therapeutics development.
Collapse
Affiliation(s)
- Yunlong Tao
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China.
| | - Xueyan Li
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiping Dong
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Linghai Kong
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Yuanwei Yan
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Ke Xu
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Seth Zima
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Yanru Li
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Melvin Ayala
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Andre M M Sousa
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, Department of Neurology, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, Department of Neurology, University of Wisconsin-Madison, Madison, WI, USA
| | - Su-Chun Zhang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Neuroscience, Department of Neurology, University of Wisconsin-Madison, Madison, WI, USA.
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
50
|
Wang J, Zhang Y, Yang H, Tian E, Guo Z, Chen J, Qiao C, Jiang H, Guo J, Zhou Z, Luo Q, Shi S, Yao H, Lu Y, Zhang S. Advanced progress of vestibular compensation in vestibular neural networks. CNS Neurosci Ther 2024; 30:e70037. [PMID: 39268632 PMCID: PMC11393560 DOI: 10.1111/cns.70037] [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: 07/16/2024] [Revised: 08/12/2024] [Accepted: 08/27/2024] [Indexed: 09/17/2024] Open
Abstract
Vestibular compensation is the natural process of recovery that occurs with acute peripheral vestibular lesion. Here, we summarize the current understanding of the mechanisms underlying vestibular compensation, focusing on the role of the medial vestibular nucleus (MVN), the central hub of the vestibular system, and its associated neural networks. The disruption of neural activity balance between the bilateral MVNs underlies the vestibular symptoms after unilateral vestibular damage, and this balance disruption can be partially reversed by the mutual inhibitory projections between the bilateral MVNs, and their top-down regulation by other brain regions via different neurotransmitters. However, the detailed mechanism of how MVN is involved in vestibular compensation and regulated remains largely unknown. A deeper understanding of the vestibular neural network and the neurotransmitter systems involved in vestibular compensation holds promise for improving treatment outcomes and developing more effective interventions for vestibular disorders.
Collapse
Affiliation(s)
- Jun Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- National Clinical Research Center for Otolaryngologic Diseases, Jiangxi Branch Center, Nanchang, China
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuejin Zhang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Huajing Yang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - E Tian
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaoqi Guo
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyu Chen
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Caijuan Qiao
- Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Hongqun Jiang
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- National Clinical Research Center for Otolaryngologic Diseases, Jiangxi Branch Center, Nanchang, China
| | - Jiaqi Guo
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhanghong Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Luo
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- National Clinical Research Center for Otolaryngologic Diseases, Jiangxi Branch Center, Nanchang, China
| | - Shiyu Shi
- Department of Rehabilitation, Liyuan Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyi Yao
- Department of Rehabilitation, Liyuan Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yisheng Lu
- Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Sulin Zhang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|