1
|
Brudzynski SM, Burgdorf JS, Moskal JR. From emotional arousal to executive action. Role of the prefrontal cortex. Brain Struct Funct 2024; 229:2327-2338. [PMID: 39096390 PMCID: PMC11611949 DOI: 10.1007/s00429-024-02837-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 07/12/2024] [Indexed: 08/05/2024]
Abstract
Emotional arousal is caused by the activity of two parallel ascending systems targeting mostly the subcortical limbic regions and the prefrontal cortex. The aversive, negative arousal system is initiated by the activity of the mesolimbic cholinergic system and the hedonic, appetitive, arousal is initiated by the activity of the mesolimbic dopaminergic system. Both ascending projections have a diffused nature and arise from the rostral, tegmental part of the brain reticular activating system. The mesolimbic cholinergic system originates in the laterodorsal tegmental nucleus and the mesolimbic dopaminergic system in the ventral tegmental area. Cholinergic and dopaminergic arousal systems have converging input to the medial prefrontal cortex. The arousal system can modulate cortical EEG with alpha rhythms, which enhance synaptic strength as shown by an increase in long-term potentiation (LTP), whereas delta frequencies are associated with decreased arousal and a decrease in synaptic strength as shown by an increase in long-term depotentiation (LTD). It is postulated that the medial prefrontal cortex is an adaptable node with decision making capability and may control the switch between positive and negative affect and is responsible for modifying or changing emotional state and its expression.
Collapse
Affiliation(s)
| | - Jeffrey S Burgdorf
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Joseph R Moskal
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| |
Collapse
|
2
|
Mao T, Guo B, Quan P, Deng Y, Chai Y, Xu J, Jiang C, Zhang Q, Lu Y, Goel N, Basner M, Dinges DF, Rao H. Morning resting hypothalamus-dorsal striatum connectivity predicts individual differences in diurnal sleepiness accumulation. Neuroimage 2024; 299:120833. [PMID: 39233125 DOI: 10.1016/j.neuroimage.2024.120833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/06/2024] Open
Abstract
While the significance of obtaining restful sleep at night and maintaining daytime alertness is well recognized for human performance and overall well-being, substantial variations exist in the development of sleepiness during diurnal waking periods. Despite the established roles of the hypothalamus and striatum in sleep-wake regulation, the specific contributions of this neural circuit in regulating individual sleep homeostasis remain elusive. This study utilized resting-state functional magnetic resonance imaging (fMRI) and mathematical modeling to investigate the role of hypothalamus-striatum connectivity in subjective sleepiness variation in a cohort of 71 healthy adults under strictly controlled in-laboratory conditions. Mathematical modeling results revealed remarkable individual differences in subjective sleepiness accumulation patterns measured by the Karolinska Sleepiness Scale (KSS). Brain imaging data demonstrated that morning hypothalamic connectivity to the dorsal striatum significantly predicts the individual accumulation of subjective sleepiness from morning to evening, while no such correlation was observed for the hypothalamus-ventral striatum connectivity. These findings underscore the distinct roles of hypothalamic connectivity to the dorsal and ventral striatum in individual sleep homeostasis, suggesting that hypothalamus-dorsal striatum circuit may be a promising target for interventions mitigating excessive sleepiness and promoting alertness.
Collapse
Affiliation(s)
- Tianxin Mao
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain-Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and Management, Shanghai International Studies University, Shanghai, China
| | - Bowen Guo
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain-Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and Management, Shanghai International Studies University, Shanghai, China
| | - Peng Quan
- Center for Functional Neuroimaging, Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA; Research Center for Quality of Life and Applied Psychology, Guangdong Medical University, Dongguan, China
| | - Yao Deng
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain-Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and Management, Shanghai International Studies University, Shanghai, China; Center for Functional Neuroimaging, Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ya Chai
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain-Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and Management, Shanghai International Studies University, Shanghai, China; Center for Functional Neuroimaging, Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jing Xu
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain-Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and Management, Shanghai International Studies University, Shanghai, China; Center for Functional Neuroimaging, Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Caihong Jiang
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain-Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and Management, Shanghai International Studies University, Shanghai, China
| | - Qingyun Zhang
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain-Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and Management, Shanghai International Studies University, Shanghai, China
| | - Yingjie Lu
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain-Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and Management, Shanghai International Studies University, Shanghai, China
| | - Namni Goel
- Biological Rhythms Research Laboratory, Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Mathias Basner
- Unit for Experimental Psychiatry, Division of Sleep and Chronobiology, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - David F Dinges
- Unit for Experimental Psychiatry, Division of Sleep and Chronobiology, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Hengyi Rao
- Center for Magnetic Resonance Imaging Research & Key Laboratory of Brain-Machine Intelligence for Information Behavior (Ministry of Education and Shanghai), School of Business and Management, Shanghai International Studies University, Shanghai, China; Center for Functional Neuroimaging, Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA; Unit for Experimental Psychiatry, Division of Sleep and Chronobiology, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Stewart AF, Fulton SL, Durand-de Cuttoli R, Thompson RE, Chen PJ, Brindley E, Cetin B, Farrelly LA, Futamura R, Claypool S, Bastle RM, Di Salvo G, Peralta C, Molina H, Baljinnyam E, Marro SG, Russo SJ, DeVita RJ, Muir TW, Maze I. Hippocampal γCaMKII dopaminylation promotes synaptic-to-nuclear signaling and memory formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613951. [PMID: 39345578 PMCID: PMC11430047 DOI: 10.1101/2024.09.19.613951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Protein monoaminylation is a class of posttranslational modification (PTM) that contributes to transcription, physiology and behavior. While recent analyses have focused on histones as critical substrates of monoaminylation, the broader repertoire of monoaminylated proteins in brain remains unclear. Here, we report the development/implementation of a chemical probe for the bioorthogonal labeling, enrichment and proteomics-based detection of dopaminylated proteins in brain. We identified 1,557 dopaminylated proteins - many synaptic - including γCaMKII, which mediates Ca2+-dependent cellular signaling and hippocampal-dependent memory. We found that γCaMKII dopaminylation is largely synaptic and mediates synaptic-to-nuclear signaling, neuronal gene expression and intrinsic excitability, and contextual memory. These results indicate a critical role for synaptic dopaminylation in adaptive brain plasticity, and may suggest roles for these phenomena in pathologies associated with altered monoaminergic signaling.
Collapse
Affiliation(s)
- Andrew F. Stewart
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Sasha L. Fulton
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Romain Durand-de Cuttoli
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | | | - Peng-Jen Chen
- Department of Pharmacological Sciences and Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Elizabeth Brindley
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Bulent Cetin
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Lorna A. Farrelly
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Rita Futamura
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Sarah Claypool
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Ryan M. Bastle
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Giuseppina Di Salvo
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Christopher Peralta
- The Rockefeller University Proteomics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Henrik Molina
- The Rockefeller University Proteomics Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Erdene Baljinnyam
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Samuele G. Marro
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Scott J. Russo
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Robert J. DeVita
- Department of Pharmacological Sciences and Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Tom W. Muir
- Department of Chemistry, Princeton, New Jersey 08544, USA
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Department of Pharmacological Sciences and Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
4
|
Cohen JS, Radhakrishnan H, Olm CA, Das SR, Cook PA, Wolk DA, Weintraub D, Irwin DJ, McMillan CT. Microstructural changes in the inferior tuberal hypothalamus correlate with daytime sleepiness in Lewy body disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.16.24312102. [PMID: 39185524 PMCID: PMC11343243 DOI: 10.1101/2024.08.16.24312102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Background Excessive daytime sleepiness (EDS) is a disabling symptom of Lewy body disorders (LBD). The hypothalamus is a key sleep-wake regulator, but its contribution to EDS in LBD remains unclear. Objectives Use diffusion MRI to evaluate the relationship of hypothalamic microstructure to EDS symptoms in LBD. Methods We studied 102 patients with clinically-defined LBD (Parkinson's disease, n=93; Parkinson's disease dementia, n=4; and dementia with Lewy bodies, n=5) and Epworth Sleepiness Scale (ESS) within 2 years of MRI. Mean diffusivity (MD) was compared between EDS+ (ESS≥10, n=37) and EDS- (ESS<10, n=65) groups in the whole hypothalamus and three subregions, covarying for age and sex. Secondary analyses tested correlations between subregion MD and continuous ESS, global cognition, and motor scores; and between subregion volume and continuous ESS. Results MD was increased in EDS+ compared to EDS- only in the inferior tuberal subregion (Cohen's d=0.43, p=0.043, β=0.117±0.057), with trend level differences in the whole hypothalamus (Cohen's d=0.39, p=0.064, β=0.070±0.037) and superior tuberal subregion (Cohen's d=0.38, p=0.073, β=0.063±0.035). No difference was seen in the posterior subregion (Cohen's d=0.1, p=0.628, β=0.019±0.038). Significant correlations with continuous ESS were seen in MD of whole hypothalamus (r2=0.074, p=0.0057), superior tuberal (r2=0.081, p=0.0038), and inferior tuberal (r2=0.073, p=0.0059) subregions. There was no correlation of hypothalamic MD with global cognition or motor scores, and no correlation of whole/subregional hypothalamic volumes with ESS. Conclusions Daytime sleepiness associates with increased MD in the inferior tuberal hypothalamus in an LBD cohort. This suggests degeneration within this region could contribute to EDS symptoms.
Collapse
|
5
|
Benoy A, Ramaswamy S. Histamine in the neocortex: Towards integrating multiscale effectors. Eur J Neurosci 2024; 60:4597-4623. [PMID: 39032115 DOI: 10.1111/ejn.16447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/10/2024] [Accepted: 06/05/2024] [Indexed: 07/22/2024]
Abstract
Histamine is a modulatory neurotransmitter, which has received relatively less attention in the central nervous system than other neurotransmitters. The functional role of histamine in the neocortex, the brain region that controls higher-order cognitive functions such as attention, learning and memory, remains largely unknown. This article focuses on the emerging roles and mechanisms of histamine release in the neocortex. We describe gaps in current knowledge and propose the application of interdisciplinary tools to dissect the detailed multiscale functional logic of histaminergic action in the neocortex ranging from sub-cellular, cellular, dendritic and synaptic levels to microcircuits and mesoscale effects.
Collapse
Affiliation(s)
- Amrita Benoy
- Neural Circuits Laboratory, Biosciences Institute, Newcastle University, Newcastle, UK
| | - Srikanth Ramaswamy
- Neural Circuits Laboratory, Biosciences Institute, Newcastle University, Newcastle, UK
- Theoretical Sciences Visiting Program (TSVP), Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| |
Collapse
|
6
|
Szabadi E. Three paradoxes related to the mode of action of pramipexole: The path from D2/D3 dopamine receptor stimulation to modification of dopamine-modulated functions. J Psychopharmacol 2024; 38:581-596. [PMID: 39041250 DOI: 10.1177/02698811241261022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Pramipexole, a D2/D3 dopamine receptor agonist, is used to treat the motor symptoms of Parkinson's disease, caused by degeneration of the dopaminergic nigrostriatal pathway. There are three paradoxes associated with its mode of action. Firstly, stimulation of D2/D3 receptors leads to neuronal inhibition, although pramipexole does not inhibit but promotes some dopamine-modulated functions, such as locomotion and reinforcement. Secondly, another dopamine-modulated function, arousal, is not promoted but inhibited by pramipexole, leading to sedation. Thirdly, pramipexole-evoked sedation is associated with an increase in pupil diameter, although sedation is expected to cause pupil constriction. To resolve these paradoxes, the path from stimulation of D2/D3 receptors to the modification of dopamine-modulated functions has been tracked. The functions considered are modulated by midbrain dopaminergic nuclei: locomotion - substantia nigra pars compacta (SNc), reinforcement/motivation - ventral tegmental area (VTA), sympathetic activity (as reflected in pupil function) - VTA; arousal - ventral periaqueductal grey (vPAG), with contributions from VTA and SNc. The application of genetics-based molecular techniques (optogenetics and chemogenetics) has enabled tracing the chains of neurones from the dopaminergic nuclei to their final targets executing the functions. The functional neuronal circuits linked to the D2/D3 receptors in the dorsal and ventral striata, stimulated by inputs from SNc and VTA, respectively, may explain how neuronal inhibition induced by pramipexole is translated into the promotion of locomotion, reinforcement/motivation and sympathetic activity. As the vPAG may increase arousal mainly by stimulating cortical D1 dopamine receptors, pramipexole would stimulate only presynaptic D2/D3 receptors on vPAG neurones, curtailing their activity and leading to sedation.
Collapse
Affiliation(s)
- Elemer Szabadi
- Developmental Psychiatry, University of Nottingham, Nottingham, UK
| |
Collapse
|
7
|
Fornaro M, Caiazza C, De Simone G, Rossano F, de Bartolomeis A. Insomnia and related mental health conditions: Essential neurobiological underpinnings towards reduced polypharmacy utilization rates. Sleep Med 2024; 113:198-214. [PMID: 38043331 DOI: 10.1016/j.sleep.2023.11.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/05/2023] [Accepted: 11/21/2023] [Indexed: 12/05/2023]
Abstract
Insomnia represents a significant public health burden, with a 10% prevalence in the general population. Reduced sleep affects social and working functioning, productivity, and patient's quality of life, leading to a total of $100 billion per year in direct and indirect healthcare costs. Primary insomnia is unrelated to any other mental or medical illness; secondary insomnia co-occurs with other underlying medical, iatrogenic, or mental conditions. Epidemiological studies found a 40-50% comorbidity prevalence between insomnia and psychiatric disorders, suggesting a high relevance of mental health in insomniacs. Sleep disturbances also worsen the outcomes of several psychiatric disorders, leading to more severe psychopathology and incomplete remission, plausibly contributing to treatment-resistant conditions. Insomnia and psychiatric disorder coexistence can lead to polypharmacy, namely, the concurrent use of two or more medications in the same patient, regardless of their purpose or rationale. Polypharmacy increases the risk of using unnecessary drugs, the likelihood of drug interactions and adverse events, and reduces the patient's compliance due to regimen complexity. The workup of insomnia must consider the patient's sleep habits and inquire about any medical and mental concurrent conditions that must be handled to allow insomnia to be remitted adequately. Monotherapy or limited polypharmacy should be preferred, especially in case of multiple comorbidities, promoting multipurpose molecules with sedative properties and with bedtime administration. Also, non-pharmacological interventions for insomnia, such as sleep hygiene, relaxation training and Cognitive Behavioral Therapy may be useful in secondary insomnia to confront behaviors and thoughts contributing to insomnia and help optimizing the pharmacotherapy. However, insomnia therapy should always be patient-tailored, considering drug indications, contraindications, and pharmacokinetics, besides insomnia phenotype, clinical picture, patient preferences, and side effect profile.
Collapse
Affiliation(s)
- Michele Fornaro
- Clinical Section of Psychiatry and Psychology, Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy
| | - Claudio Caiazza
- Clinical Section of Psychiatry and Psychology, Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy.
| | - Giuseppe De Simone
- Clinical Section of Psychiatry and Psychology, Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy; Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy
| | - Flavia Rossano
- Clinical Section of Psychiatry and Psychology, Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy
| | - Andrea de Bartolomeis
- Clinical Section of Psychiatry and Psychology, Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy; Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy
| |
Collapse
|
8
|
Ferretti A, Gatto M, Velardi M, Di Nardo G, Foiadelli T, Terrin G, Cecili M, Raucci U, Valeriani M, Parisi P. Migraine, Allergy, and Histamine: Is There a Link? J Clin Med 2023; 12:jcm12103566. [PMID: 37240671 DOI: 10.3390/jcm12103566] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/14/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
The relationship between migraines and allergies is controversial. Though they are epidemiologically linked, the underlying pathophysiological connection between them remains unclear. Migraines and allergic disorders have various underlying genetic and biological causes. As per the literature, these conditions are epidemiologically linked, and some common pathophysiological pathways have been hypothesized. The histaminergic system may be the clue to understanding the correlation among these diseases. As a neurotransmitter in the central nervous system with a vasodilatory effect, histamine has a well-documented influence on the allergic response and could be involved in the pathophysiology of migraines. Histamine may influence hypothalamic activity, which may play a major role in migraines or may simply influence their severity. In both cases, antihistamine drugs could prove useful. This review examines whether the histaminergic system, particularly H3 and H4 receptors, may provide a mechanistic link between the pathophysiology of migraines and allergic disorders, two common and debilitating conditions. Identifying their connection could help identify novel therapeutic strategies.
Collapse
Affiliation(s)
- Alessandro Ferretti
- Pediatrics Unit, Neuroscience, Mental Health and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Mattia Gatto
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Margherita Velardi
- General and Emergency Department, Bambino Gesù Children's Hospital, Istituto di Ricerca e Cura a Carattere Scientifico, 00165 Rome, Italy
| | - Giovanni Di Nardo
- Pediatrics Unit, Neuroscience, Mental Health and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Thomas Foiadelli
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Gianluca Terrin
- Department of Mother and Child, Gynecological and Urological Sciences, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00185 Rome, Italy
| | - Manuela Cecili
- Pediatrics Unit, Neuroscience, Mental Health and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Umberto Raucci
- General and Emergency Department, Bambino Gesù Children's Hospital, Istituto di Ricerca e Cura a Carattere Scientifico, 00165 Rome, Italy
| | - Massimiliano Valeriani
- Developmental Neurology Unit, Bambino Gesù Children's Hospital, Istituto di Ricerca e Cura a Carattere Scientifico, 00165 Rome, Italy
| | - Pasquale Parisi
- Pediatrics Unit, Neuroscience, Mental Health and Sense Organs (NESMOS) Department, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
9
|
Water Drinking Behavior Associated with Aversive Arousal in Rats: An Integrative Approach. Brain Sci 2022; 13:brainsci13010060. [PMID: 36672042 PMCID: PMC9857118 DOI: 10.3390/brainsci13010060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 12/30/2022] Open
Abstract
Cholinergic muscarinic stimulation of vast areas of the limbic brain induced a well-documented polydipsia in laboratory rats. This excessive water-drinking behavior has not received any convincing biological and physiological interpretation for the last 50 years. This review offers such an interpretation and suggests that cholinergically induced drinking response, mostly by carbachol, is associated with activation of the ascending mesolimbic cholinergic system that serves for initiation of emotional aversive arousal of the organism. The ascending cholinergic system originates from the laterodorsal tegmental nucleus, has a diffuse nature, and affects numerous subcortical limbic structures. It is proposed that the carbachol-induced drinking response is related to the state of anxiety and does not serve the regulation of thirst. Instead, the response is anxiety-induced polydipsia that might occur as a soothing procedure that decreases the aversiveness of the negative emotional state induced by carbachol. It is concluded that carbachol-induced water-drinking behavior is a rewarding process that contributes to alleviating the feeling of anxiety by bringing some relief from the cholinergically induced aversive state, and it is a homologue to anxiety-driven polydipsia in humans.
Collapse
|
10
|
Eshtiaghi A, Eapen-John D, Zaslavsky K, Vosoughi R, Murray BJ, Margolin E. Sleep Quality in Neuromyelitis Optica Spectrum Disorder: A Systematic Review. Int J MS Care 2022; 24:124-131. [PMID: 35645625 PMCID: PMC9135364 DOI: 10.7224/1537-2073.2021-019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
BACKGROUND This review summarizes the literature on sleep quality in neuromyelitis optica spectrum disorder (NMOSD) and discusses these findings in the context of current knowledge of sleep physiology. METHODS A literature search was performed using Ovid MEDLINE, Embase, and Scopus from inception to September 3, 2020. All included studies reported at least 1 measure of sleep quality in individuals with NMOSD. Pittsburgh Sleep Quality Index (PSQI) scores of individuals from 4 studies were compared with those from a data set of controls. RESULTS Thirteen studies (1041 individuals with NMOSD) were included in the review. Disturbed sleep was demonstrated across subjective metrics based on patient surveys and objective metrics such as polysomnography. An estimated 70% of individuals with NMOSD can be classified as poor sleepers. Standardized mean difference between PSQI scores of 183 individuals with NMOSD and those of 9284 controls was 0.72 (95% CI, 0.57-0.86; P < .001). Decreased sleep quality was significantly associated with decreased quality of life and increased anxiety, depression, and disability status. Sleep disturbances in NMOSD were similar in severity to those in multiple sclerosis. CONCLUSIONS Sleep disturbances are a major contributor to NMOSD disease burden and may arise from the disruption of sleep circuitry, in addition to physical and psychological complications. Multiple processes involved in sleep regulation may be affected, such as, but not limited to, neural circadian circuit disruption, direct effects of inflammation, aminergic projecting system abnormalities, glymphatic system impairment, and development of sleep disorders such as restless legs syndrome/sleep apnea. A better understanding of these mechanisms is necessary for developing effective therapies for NMOSD-associated sleep disturbances.
Collapse
Affiliation(s)
- Arshia Eshtiaghi
- From the Faculty of Medicine (AE, DE-J), University of Toronto, Toronto, ON, Canada
| | - David Eapen-John
- From the Faculty of Medicine (AE, DE-J), University of Toronto, Toronto, ON, Canada
| | - Kirill Zaslavsky
- From the Department of Ophthalmology and Vision Sciences (KZ, EM), University of Toronto, Toronto, ON, Canada
| | - Reza Vosoughi
- From the Division of Neurology, Department of Medicine (RV, BJM), University of Toronto, Toronto, ON, Canada
- From the St Michael’s Hospital, Toronto, ON, Canada (RV)
| | - Brian J. Murray
- From the Division of Neurology, Department of Medicine (RV, BJM), University of Toronto, Toronto, ON, Canada
- From the Sunnybrook Health Science Centre, Toronto, ON, Canada (BJM)
| | - Edward Margolin
- From the Department of Ophthalmology and Vision Sciences (KZ, EM), University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Sleep Apnea and Hypertension. High Blood Press Cardiovasc Prev 2021; 29:23-31. [PMID: 34739711 DOI: 10.1007/s40292-021-00484-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/30/2021] [Indexed: 12/20/2022] Open
Abstract
Obstructive sleep apnea is a frequent finding in clinical practice especially with the obesity epidemic and the growing awareness of sleep-disordered breathing as a potential and treatable risk factor for cardiovascular diseases. It frequently coexists undiagnosed activating pathophysiological mechanisms known to participate in development and progression of cardiovascular diseases and resistance to therapeutical strategies. The sympathetic activation and the baroreflex and chemoreflex impairment appear to be the main pathophysiological factors that activating several mechanisms elicit cardiac and vascular damage. Data from cross-sectional population-based studies, prospective studies and meta-analysis have clearly shown the implication of OSA in the development of the hypertensive state and the benefits obtained by continuous positive airway pressure on daytime blood pressure and cardiovascular risk.
Collapse
|
12
|
Carthy E, Ellender T. Histamine, Neuroinflammation and Neurodevelopment: A Review. Front Neurosci 2021; 15:680214. [PMID: 34335160 PMCID: PMC8317266 DOI: 10.3389/fnins.2021.680214] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/18/2021] [Indexed: 12/16/2022] Open
Abstract
The biogenic amine, histamine, has been shown to critically modulate inflammatory processes as well as the properties of neurons and synapses in the brain, and is also implicated in the emergence of neurodevelopmental disorders. Indeed, a reduction in the synthesis of this neuromodulator has been associated with the disorders Tourette's syndrome and obsessive-compulsive disorder, with evidence that this may be through the disruption of the corticostriatal circuitry during development. Furthermore, neuroinflammation has been associated with alterations in brain development, e.g., impacting synaptic plasticity and synaptogenesis, and there are suggestions that histamine deficiency may leave the developing brain more vulnerable to proinflammatory insults. While most studies have focused on neuronal sources of histamine it remains unclear to what extent other (non-neuronal) sources of histamine, e.g., from mast cells and other sources, can impact brain development. The few studies that have started exploring this in vitro, and more limited in vivo, would indicate that non-neuronal released histamine and other preformed mediators can influence microglial-mediated neuroinflammation which can impact brain development. In this Review we will summarize the state of the field with regard to non-neuronal sources of histamine and its impact on both neuroinflammation and brain development in key neural circuits that underpin neurodevelopmental disorders. We will also discuss whether histamine receptor modulators have been efficacious in the treatment of neurodevelopmental disorders in both preclinical and clinical studies. This could represent an important area of future research as early modulation of histamine from neuronal as well as non-neuronal sources may provide novel therapeutic targets in these disorders.
Collapse
Affiliation(s)
- Elliott Carthy
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Tommas Ellender
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
13
|
Dupont D, Lin JS, Peyron F, Akaoka H, Wallon M. Chronic Toxoplasma gondii infection and sleep-wake alterations in mice. CNS Neurosci Ther 2021; 27:895-907. [PMID: 34085752 PMCID: PMC8265947 DOI: 10.1111/cns.13650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/21/2021] [Accepted: 03/24/2021] [Indexed: 11/29/2022] Open
Abstract
AIM Toxoplasma gondii (Tg) is an intracellular parasite infecting more than a third of the human population. Yet, the impact of Tg infection on sleep, a highly sensitive index of brain functions, remains unknown. We designed an experimental mouse model of chronic Tg infection to assess the effects on sleep-wake states. METHODS Mice were infected using cysts of the type II Prugniaud strain. We performed chronic sleep-wake recordings and monitoring as well as EEG power spectral density analysis in order to assess the quantitative and qualitative changes of sleep-wake states. Pharmacological approach was combined to evaluate the direct impact of the infection and inflammation caused by Tg. RESULTS Infected mouse exhibited chronic sleep-wake alterations over months, characterized by a marked increase (>20%) in time spent awake and in cortical EEG θ power density of all sleep-wake states. Meanwhile, slow-wave sleep decreased significantly. These effects were alleviated by an anti-inflammatory treatment using corticosteroid dexamethasone. CONCLUSION We demonstrated for the first time the direct consequences of Tg infection on sleep-wake states. The persistently increased wakefulness and reduced sleep fit with the parasite's strategy to enhance dissemination through host predation and are of significance in understanding the neurodegenerative and neuropsychiatric disorders reported in infected patients.
Collapse
Affiliation(s)
- Damien Dupont
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.,Physiologie intégrée du système d'éveil, Faculté de Médecine, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Université Claude Bernard Lyon 1, Lyon, France
| | - Jian-Sheng Lin
- Physiologie intégrée du système d'éveil, Faculté de Médecine, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Université Claude Bernard Lyon 1, Lyon, France
| | - François Peyron
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Hideo Akaoka
- Physiologie intégrée du système d'éveil, Faculté de Médecine, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Université Claude Bernard Lyon 1, Lyon, France
| | - Martine Wallon
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.,Physiologie intégrée du système d'éveil, Faculté de Médecine, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
14
|
Shimada M, Miyagawa T, Kodama T, Toyoda H, Tokunaga K, Honda M. Metabolome analysis using cerebrospinal fluid from narcolepsy type 1 patients. Sleep 2021; 43:5837570. [PMID: 32412602 DOI: 10.1093/sleep/zsaa095] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/25/2020] [Indexed: 12/14/2022] Open
Abstract
Narcolepsy type 1 (NT1) is a hypersomnia characterized by excessive daytime sleepiness and cataplexy. Inappropriate regulation of fatty acid metabolism has been suggested to be involved in the pathophysiology of NT1, but the detailed mechanisms remain uncertain. Here we performed a metabolomic analysis of cerebrospinal fluid samples from 14 NT1 and 17 control subjects using a novel capillary electrophoresis coupled with Fourier transform mass spectrometry. A total of 268 metabolites were identified and the amount of histidine was the most significantly increased in NT1 patients (p = 4.0 × 10-4). Validation analysis using high-performance liquid chromatography (HPLC) including independent replication samples also identified the association of histidine (p = 2.02 × 10-3). Further, levels of histamine, which is synthesized from histidine, were also examined using HPLC and were found to be significantly decreased in NT1 patients (p = 6.12 × 10-4). Pathway analysis with nominally significant metabolites identified several pathways related to the metabolism of glycogenic amino acids, suggesting that glycogenesis is enhanced in NT1 as a compensatory mechanism for fatty acid metabolism. We performed further exploratory analysis, searching for metabolites associated with sleep variables from polysomnography and the multiple sleep latency test. As a result, 5'-deoxy-5'-methylthioadenosine showed a significant association with apnea-hypopnea index (p = 2.66 ×10-6). Moreover, gamma aminobutyric acid displayed a negative correlation with rapid eye movement sleep latency (REML), and thus might represent an intriguing target for future studies to elucidate how the controlling circuit of REM sleep is associated with abnormally short REML in NT1.
Collapse
Affiliation(s)
- Mihoko Shimada
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taku Miyagawa
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tohru Kodama
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hiromi Toyoda
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsushi Tokunaga
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Genome Medical Science Project (Toyama), National Center for Global Health and Medicine (NCGM), Tokyo, Japan
| | - Makoto Honda
- Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Seiwa Hospital, Institute of Neuropsychiatry, Tokyo, Japan
| |
Collapse
|
15
|
Wilbraham D, Biglan KM, Svensson KA, Tsai M, Kielbasa W. Safety, Tolerability, and Pharmacokinetics of Mevidalen (LY3154207), a Centrally Acting Dopamine D1 Receptor-Positive Allosteric Modulator (D1PAM), in Healthy Subjects. Clin Pharmacol Drug Dev 2021; 10:393-403. [PMID: 33029934 PMCID: PMC8048550 DOI: 10.1002/cpdd.874] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/02/2020] [Indexed: 12/27/2022]
Abstract
Activation of the brain dopamine D1 receptor has attracted attention because of its promising role in neuropsychiatric diseases. Although efforts to develop D1 agonists have been challenging, a positive allosteric modulator (PAM), represents an attractive approach with potential better drug-like properties. Phase 1 single-ascending-dose (SAD; NCT03616795) and multiple-ascending-dose (MAD; NCT02562768) studies with the D1PAM mevidalen (LY3154207) were conducted with healthy subjects. There were no treatment-related serious adverse events (AEs) in these studies. In the SAD study, 25-200 mg administered orally showed dose-proportional pharmacokinetics (PK) and acute dose-related increases in systolic blood pressure (SBP) and diastolic blood pressure DBP) and pulse rate at doses ≥ 75 mg. AE related to central activation were seen at doses ≥ 75 mg. At 25 and 75 mg, central penetration of mevidalen was confirmed by measurement of mevidalen in cerebrospinal fluid. In the MAD study, once-daily doses of mevidalen at 15-150 mg for 14 days showed dose-proportional PK. Acute dose-dependent increases in SBP, DBP, and PR were observed on initial administration, but with repeated dosing the effects diminished and returned toward baseline levels. Overall, these findings support further investigation of mevidalen as a potential treatment for a range of neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | | | - Max Tsai
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | | |
Collapse
|
16
|
Zhang X, Lin JS, Spruyt K. Sleep problems in Rett syndrome animal models: A systematic review. J Neurosci Res 2020; 99:529-544. [PMID: 32985711 DOI: 10.1002/jnr.24730] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/27/2020] [Accepted: 08/30/2020] [Indexed: 02/01/2023]
Abstract
Due to the discovery of Rett Syndrome (RTT) genetic mutations, animal models have been developed. Sleep research in RTT animal models may unravel novel neural mechanisms for this severe neurodevelopmental heritable rare disease. In this systematic literature review we summarize the findings on sleep research of 13 studies in animal models of RTT. We found disturbed efficacy and continuity of sleep in all genetically mutated models of mice, cynomolgus monkeys, and Drosophila. Models presented highly fragmented sleep with distinct differences in 24-hr sleep/wake cyclicity and circadian arrhythmicity. Overall, animal models mimic sleep complaints reported in individuals with RTT. However, contrary to human studies, in mutant mice, attenuated sleep delta waves, and sleep apneas in non-rapid eye movement sleep were reported. Future studies may focus on sleep structure and EEG alterations, potential central mechanisms involved in sleep fragmentation and the occurrence of sleep apnea across different sleep stages. Given that locomotor dysfunction is characteristic of individuals with RTT, studies may consider to integrate its potential impact on the behavioral analysis of sleep.
Collapse
Affiliation(s)
- Xinyan Zhang
- INSERM - School of Medicine, University Claude Bernard, Lyon, France
| | - Jian-Sheng Lin
- INSERM - School of Medicine, University Claude Bernard, Lyon, France
| | - Karen Spruyt
- INSERM - School of Medicine, University Claude Bernard, Lyon, France
| |
Collapse
|
17
|
Pedrazzoli M, Mazzotti DR, Ribeiro AO, Mendes JV, Bittencourt LRA, Tufik S. A single nucleotide polymorphism in the HOMER1 gene is associated with sleep latency and theta power in sleep electroencephalogram. PLoS One 2020; 15:e0223632. [PMID: 32645048 PMCID: PMC7347117 DOI: 10.1371/journal.pone.0223632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 06/16/2020] [Indexed: 12/14/2022] Open
Abstract
Glutamate is the most excitatory neurotransmitter in the central nervous system and it is involved in the initiation and maintaining of waking and rapid-eye-movement (REM) sleep. Homer proteins act in the trafficking and/or clustering of metabotropic glutamate receptors, and polymorphisms in the HOMER1 gene have been associated with phenotypes related to glutamate signaling dysregulation. In this study, we report the association of a single nucleotide polymorphism (SNP) in the HOMER1 gene (rs3822568) with specific aspects of sleep in a sample of the Brazilian population. To accomplish this, 1,042 individuals were subjected to a full-night polysomnography, and a subset of 983 subjects had rs3822568 genotyping data available. When compared with the A allele carriers, GG genotyped individuals showed higher sleep latency, lower sleep efficiency, reduced number of arousals per hour, lower apnea-hypopnea index (AHI) and lower theta spectral power. In summary, the present findings suggest that the rs3822568 polymorphism in the HOMER1 gene is associated with sleep EEG profiles and might have an impact on sleep quality and sleep structure, with potential to explain inter-individual variation in sleep homeostasis.
Collapse
Affiliation(s)
- Mario Pedrazzoli
- School of Arts, Sciences and Humanities, University of São Paulo (USP), São Paulo, Brazil
| | - Diego Robles Mazzotti
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | | | - Juliana Viana Mendes
- School of Arts, Sciences and Humanities, University of São Paulo (USP), São Paulo, Brazil
| | | | - Sergio Tufik
- Department of Psychobiology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
18
|
Jang SH, Kwon HG. The Neural Tract Between the Hypothalamus and Basal Forebrain in the Ascending Reticular Activating System: A Diffusion Tensor Tractography Study. Curr Med Imaging 2020; 15:369-372. [PMID: 31989905 DOI: 10.2174/1573405614666180522081034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 04/26/2018] [Accepted: 05/10/2018] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Ascending Reticular Activating System (ARAS) has a key role in consciousness. The ARAS is a complex network consisting of a portion of the brainstem reticular formation, nonspecific thalamic nuclei, hypothalamus, Basal Forebrain (BF), and cerebral cortex. We examined the reconstruction method and features of the neural tract between the hypothalamus and the BF in normal subjects, using Diffusion Tensor Tractography (DTT). METHODS Twenty-three healthy subjects were recruited. The ARAS between the hypothalamus and the BF was reconstructed by two Regions of Interest (ROIs): 1) seed ROI - the isolated green portion for the BF on the color map, 2) target ROI - the hypothalamus on the axial image. DTT parameters of the ARAS between the hypothalamus and the BF were examined. RESULTS Among 46 hemispheres in 23 normal subjects, 24 hemispheres (52.2 %) were identified in the ARAS between the hypothalamus and the BF. The reconstructed ARAS between the hypothalamus and the BF connected from the hypothalamus to the commissural level and anteriorly through the anterior commissure and then reached the BF. CONCLUSION Using DTT, the ARAS between the hypothalamus and the BF was identified in normal subjects. Because the hypothalamus and BF are related to the regulation of wakefulness and sleep, our reconstruction method and results would be useful in the research on sleep and wakefulness aspects of consciousness.
Collapse
Affiliation(s)
- Sung Ho Jang
- Department of Physical Medicine and Rehabilitation, College of Medicine, Yeungnam University, Gyeongsan, Korea
| | - Hyeok Gyu Kwon
- Department of Physical Therapy, College of Health Sciences, Eulji University, Gyeonggi, Korea
| |
Collapse
|
19
|
Abstract
The neural mechanisms of sleep, a fundamental biological behavior from invertebrates to humans, have been a long-standing mystery and present an enormous challenge. Gradually, perspectives on the neurobiology of sleep have been more various with the technical innovations over the recent decades, and studies have now identified many specific neural circuits that selectively regulate the initiation and maintenance of wake, rapid eye movement (REM) sleep, and non-REM (NREM) sleep. The cholinergic system in basal forebrain (BF) that fire maximally during waking and REM sleep is one of the key neuromodulation systems related to waking and REM sleep. Here we outline the recent progress of the BF cholinergic system in sleep-wake cycle. The intricate local connectivity and multiple projections to other cortical and subcortical regions of the BF cholinergic system elaborately presented here form a conceptual framework for understanding the coordinating effects with the dissecting regions. This framework also provides evidences regarding the relationships between the general anesthesia and wakefulness/sleep cycle focusing on the neural circuitry of unconsciousness induced by anesthetic drugs.
Collapse
|
20
|
Sergeeva OA, Chepkova AN, Görg B, Rodrigues Almeida F, Bidmon HJ, Haas HL, Häussinger D. Histamine-induced plasticity and gene expression in corticostriatal pathway under hyperammonemia. CNS Neurosci Ther 2019; 26:355-366. [PMID: 31571389 PMCID: PMC7052803 DOI: 10.1111/cns.13223] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 08/10/2019] [Accepted: 09/04/2019] [Indexed: 01/13/2023] Open
Abstract
Aims Histamine H3 receptor (H3R) antagonists/inverse agonists increase vigilance. We studied brain histaminergic pathways under hyperammonemia and the transcriptome of receptors and their signaling cascades to provide a rationale for wake‐promoting therapies. Methods We analyzed histamine‐induced long‐lasting depression of corticostriatal synaptic transmission (LLDhist). As the expression of dopamine 1 receptors (D1R) is upregulated in LGS‐KO striatum where D1R‐H3R dimers may exist, we investigated actions of H3R and D1R agonists and antagonists. We analyzed transcription of selected genes in cortex and dorsal striatum in a mouse model of inborn hyperammonemia (liver‐specific glutamine synthetase knockout: LGS‐KO) and compared it with human hepatic encephalopathy. Results LGS‐KO mice showed significant reduction of the direct depression (DD) but not the long‐lasting depression (LLD) by histamine. Neither pharmacological activation nor inhibition of D1R significantly affected DDhist and LLDhist in WT striatum, while in LGS‐KO mice D1R activation suppressed LLDhist. Histaminergic signaling was found unchanged at the transcriptional level except for the H2R. A study of cAMP‐regulated genes indicated a significant reduction in the molecular signature of wakefulness in the diseased cortex. Conclusions Our findings provide a rationale for the development of aminergic wake‐promoting therapeutics in hyperammonemic disorders.
Collapse
Affiliation(s)
- Olga A Sergeeva
- Molecular Neurophysiology, Medical Faculty, Institute of Neural and Sensory Physiology, Heinrich-Heine University, Duesseldorf, Germany.,Medical Faculty, Institute of Clinical Neurosciences and Medical Psychology, Heinrich-Heine University, Duesseldorf, Germany
| | - Aisa N Chepkova
- Molecular Neurophysiology, Medical Faculty, Institute of Neural and Sensory Physiology, Heinrich-Heine University, Duesseldorf, Germany.,Clinic of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| | - Boris Görg
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| | - Filipe Rodrigues Almeida
- Medical Faculty, Institute of Clinical Neurosciences and Medical Psychology, Heinrich-Heine University, Duesseldorf, Germany
| | - Hans-Jürgen Bidmon
- Medical Faculty, C.&O. Vogt Institute for Brain Research, Heinrich-Heine University, Duesseldorf, Germany
| | - Helmut L Haas
- Molecular Neurophysiology, Medical Faculty, Institute of Neural and Sensory Physiology, Heinrich-Heine University, Duesseldorf, Germany
| | - Dieter Häussinger
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| |
Collapse
|
21
|
Boes AD, Fischer D, Geerling JC, Bruss J, Saper CB, Fox MD. Connectivity of sleep- and wake-promoting regions of the human hypothalamus observed during resting wakefulness. Sleep 2019; 41:5021065. [PMID: 29850898 DOI: 10.1093/sleep/zsy108] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Indexed: 11/13/2022] Open
Abstract
The hypothalamus is a central hub for regulating sleep-wake patterns, the circuitry of which has been investigated extensively in experimental animals. This work has identified a wake-promoting region in the posterior hypothalamus, with connections to other wake-promoting regions, and a sleep-promoting region in the anterior hypothalamus, with inhibitory projections to the posterior hypothalamus. It is unclear whether a similar organization exists in humans. Here, we use anatomical landmarks to identify homologous sleep- and wake-promoting regions of the human hypothalamus and investigate their functional relationships using resting-state functional connectivity magnetic resonance imaging in healthy awake participants. First, we identify a negative correlation (anticorrelation) between the anterior and posterior hypothalamus, two regions with opposing roles in sleep-wake regulation. Next, we show that hypothalamic connectivity predicts a pattern of regional sleep-wake changes previously observed in humans. Specifically, regions that are more positively correlated with the posterior hypothalamus and more negatively correlated with the anterior hypothalamus correspond to regions with the greatest change in cerebral blood flow between sleep-wake states. Taken together, these findings provide preliminary evidence relating a hypothalamic circuit investigated in animals to sleep-wake neuroimaging results in humans, with implications for our understanding of human sleep-wake regulation and the functional significance of anticorrelations.
Collapse
Affiliation(s)
- Aaron D Boes
- Department of Pediatrics, Iowa Neuroimaging and Noninvasive Brain Stimulation Program, University of Iowa Hospitals and Clinics, Iowa City, IA.,Department of Neurology, Iowa Neuroimaging and Noninvasive Brain Stimulation Program, University of Iowa Hospitals and Clinics, Iowa City, IA.,Department of Psychiatry, Iowa Neuroimaging and Noninvasive Brain Stimulation Program, University of Iowa Hospitals and Clinics, Iowa City, IA
| | - David Fischer
- Department of Neurology, Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, MA
| | - Joel C Geerling
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA
| | - Joel Bruss
- Department of Neurology, Iowa Neuroimaging and Noninvasive Brain Stimulation Program, University of Iowa Hospitals and Clinics, Iowa City, IA
| | - Clifford B Saper
- Department of Neurology, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, MA
| | - Michael D Fox
- Department of Neurology, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, MA.,Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
22
|
Abstract
Over the past decade, basic sleep research investigating the circuitry controlling sleep and wakefulness has been boosted by pharmacosynthetic approaches, including chemogenetic techniques using designed receptors exclusively activated by designer drugs (DREADD). DREADD offers a series of tools that selectively control neuronal activity as a way to probe causal relationship between neuronal sub-populations and the regulation of the sleep-wake cycle. Following the path opened by optogenetics, DREADD tools applied to discrete neuronal sub-populations in numerous brain areas quickly made their contribution to the discovery and the expansion of our understanding of critical brain structures involved in a wide variety of behaviors and in the control of vigilance state architecture.
Collapse
|
23
|
Yin D, Dong H, Wang TX, Hu ZZ, Cheng NN, Qu WM, Huang ZL. Glutamate Activates the Histaminergic Tuberomammillary Nucleus and Increases Wakefulness in Rats. Neuroscience 2019; 413:86-98. [DOI: 10.1016/j.neuroscience.2019.05.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 05/10/2019] [Accepted: 05/17/2019] [Indexed: 01/23/2023]
|
24
|
Chazot PL, Johnston L, Mcauley E, Bonner S. Histamine and Delirium: Current Opinion. Front Pharmacol 2019; 10:299. [PMID: 31024298 PMCID: PMC6467166 DOI: 10.3389/fphar.2019.00299] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/11/2019] [Indexed: 12/14/2022] Open
Abstract
Delirium is a very common, but refractory clinical state, notably present in intensive care and in the growing aging community. It is characterized by fluctuating disturbances in a number of key behavioral features, namely cognition, mood, attention, arousal, and self-awareness. Histamine is arguably the most pleotropic neurotransmitter in the human brain, and this review provides a rationale, and proposes that this neuroactive amine plays a role in modulating the characteristic features of delirium. While centrally permeable H1 and H2 histamine receptor antagonists have pro-delirium potential, we propose that centrally permeable H3 histamine receptor antagonists may provide an exciting new strategy to combat delirium. The Histamine H4 receptor may also have an indirect inflammatory neuroglial role which requires further exploration.
Collapse
Affiliation(s)
- Paul L. Chazot
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - Laura Johnston
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - Edel Mcauley
- Intensive Care, South Tees Hospitals NHS Foundation Trust, The James Cook University Hospital, Middlesbrough, United Kingdom
| | - Stephen Bonner
- Intensive Care, South Tees Hospitals NHS Foundation Trust, The James Cook University Hospital, Middlesbrough, United Kingdom
| |
Collapse
|
25
|
Shao YF, Lin JS, Hou YP. Paraventricular Thalamus as A Major Thalamic Structure for Wake Control. Neurosci Bull 2019; 35:946-948. [PMID: 30879175 DOI: 10.1007/s12264-019-00364-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 12/30/2018] [Indexed: 11/30/2022] Open
Affiliation(s)
- Yu-Feng Shao
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jian-Sheng Lin
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, Lyon, 69373, France
| | - Yi-Ping Hou
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
26
|
Spruyt K, Herbillon V, Putois B, Franco P, Lachaux JP. Mind-wandering, or the allocation of attentional resources, is sleep-driven across childhood. Sci Rep 2019; 9:1269. [PMID: 30718835 PMCID: PMC6362223 DOI: 10.1038/s41598-018-37434-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 11/22/2018] [Indexed: 11/09/2022] Open
Abstract
Mind-wandering or the spontaneous, uncontrolled changes in the allocation of attention resources (lapses) may cause variability in performance. In childhood, the relationship between the activation state of the brain, such as in attentional performance, and sleep has not been explored in detail. We investigated the role of sleep in attentional performance, and explored the most important parameters of their relationship. We objectively measured momentary lapses of attention of 522 children and correlated them with sleep schedules. In the subgroup of young children (age 7.1 ± 0.6 years; 60.8% girls), increasing age, long sleep duration and assessment closer to the previous night’s sleep period was associated with impaired performance speed and consistency. From pre-adolescence (age 9.4 ± 0.8 years; 50.5% girls) onwards somno-typologies may develop. As a result, in adolescence (age 13.4 ± 1.2 years; 51.3% girls) not only sleep duration but also sleep midpoint and sleep regularity influence the individual speed and stability of attention. Across development, regularity of sleep, individual sleep midpoint and bedtime become increasingly important for optimal performance throughout the day. Attentional performance and sleep shared almost half of their variance, and performance was sleep-driven across childhood. Future studies should focus on intra- and inter-individual differences in sleep-wake behavior to improve performance or decrease mind-wandering in youth by targeting sleep habits.
Collapse
Affiliation(s)
- Karen Spruyt
- Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292 - Waking Team, University Claude Bernard, School of Medicine, Lyon, France.
| | - Vania Herbillon
- Epilepsy, Sleep and Pediatric Neurophysiology Department, University Hospitals of Lyon, Lyon, France
| | - Benjamin Putois
- Epilepsy, Sleep and Pediatric Neurophysiology Department, University Hospitals of Lyon, Lyon, France
| | - Patricia Franco
- Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292 - Waking Team, University Claude Bernard, School of Medicine, Lyon, France.,Epilepsy, Sleep and Pediatric Neurophysiology Department, University Hospitals of Lyon, Lyon, France
| | - Jean-Philippe Lachaux
- Lyon Neuroscience Research Center, INSERM U1028-CNRS5292 - Brain Dynamics and Cognition Team, University Claude Bernard, School of Medicine, Lyon, France
| |
Collapse
|
27
|
Zhong YH, Wu HY, He RH, Zheng BE, Fan JZ. Sex Differences in Sex Hormone Profiles and Prediction of Consciousness Recovery After Severe Traumatic Brain Injury. Front Endocrinol (Lausanne) 2019; 10:261. [PMID: 31080439 PMCID: PMC6497747 DOI: 10.3389/fendo.2019.00261] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 04/08/2019] [Indexed: 12/16/2022] Open
Abstract
Objective: The clinical course of unconsciousness after traumatic brain injury (TBI) is commonly unpredictable and it remains a challenge with limited therapeutic options. The aim of this study was to evaluate the early changes in serum sex hormone levels after severe TBI (sTBI) and the use of these hormones to predict recovery from unconsciousness with regard to sex. Methods: We performed a retrospective study including patients with sTBI. A statistical of analysis of serum sex hormone levels and recovery of consciousness at 6 months was made to identify the effective prognostic indicators. Results: Fifty-five male patients gained recovery of consciousness, and 37 did not. Of the female patients, 22 out of 32 patients regained consciousness. Male patients (n = 92) with sTBI, compared with healthy subjects (n = 60), had significantly lower levels of follicular stimulating hormone (FSH), testosterone and progesterone and higher levels of prolactin. Female patients (n = 32) with sTBI, compared with controls (n = 60), had significantly lower levels of estradiol, progesterone, and testosterone and significantly higher levels of FSH and prolactin. Testosterone significantly predicted consciousness recovery in male patients. Normal or elevated testosterone levels in the serum were associated with a reduced risk of the unconscious state in male patients with sTBI. For women patients with sTBI, sex hormone levels did not contribute to the prediction of consciousness recovery. Conclusion: These findings indicate that TBI differentially affects the levels of sex-steroid hormones in men and women patients. Plasma levels of testosterone could be a good candidate blood marker to predict recovery from unconsciousness after sTBI for male patients.
Collapse
|
28
|
De Luca R, Mazur K, Kernder A, Suvorava T, Kojda G, Haas HL, Sergeeva OA. Mechanisms of N-oleoyldopamine activation of central histaminergic neurons. Neuropharmacology 2018; 143:327-338. [DOI: 10.1016/j.neuropharm.2018.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/30/2018] [Accepted: 09/07/2018] [Indexed: 12/18/2022]
|
29
|
Jang SH, Yeo SS. Restoration of an injured lower dorsal ascending reticular activating system in a patient with intraventricular hemorrhage. Neural Regen Res 2018; 13:2022-2024. [PMID: 30233078 PMCID: PMC6183035 DOI: 10.4103/1673-5374.238719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Sung Ho Jang
- Department of Physical Medicine and Rehabilitation, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Sang Seok Yeo
- Department of Physical Therapy, College of Health Sciences, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
30
|
Franco P, Dauvilliers Y, Inocente CO, Guyon A, Villanueva C, Raverot V, Plancoulaine S, Lin JS. Impaired histaminergic neurotransmission in children with narcolepsy type 1. CNS Neurosci Ther 2018; 25:386-395. [PMID: 30225986 DOI: 10.1111/cns.13057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 08/03/2018] [Accepted: 08/04/2018] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE Narcolepsy is a sleep disorder characterized in humans by excessive daytime sleepiness and cataplexy. Greater than fifty percent of narcoleptic patients have an onset of symptoms prior to the age of 18. Current general agreement considers the loss of hypothalamic hypocretin (orexin) neurons as the direct cause of narcolepsy notably cataplexy. To assess whether brain histamine (HA) is also involved, we quantified the cerebrospinal fluid (CSF) levels of HA and tele-methylhistamine (t-MeHA), the direct metabolite of HA between children with orexin-deficient narcolepsy type 1 (NT1) and controls. METHODS We included 24 children with NT1 (12.3 ± 3.6 years, 11 boys, 83% cataplexy, 100% HLA DQB1*06:02) and 21 control children (11.2 ± 4.2 years, 10 boys). CSF HA and t-MeHA were measured in all subjects using a highly sensitive liquid chromatographic-electrospray/tandem mass spectrometric assay. CSF hypocretin-1 values were determined in the narcoleptic patients. RESULTS Compared with the controls, NT1 children had higher CSF HA levels (771 vs 234 pmol/L, P < 0.001), lower t-MeHA levels (879 vs 1924 pmol/L, P < 0.001), and lower t-MeHA/HA ratios (1.1 vs 8.2, P < 0.001). NT1 patients had higher BMI z-scores (2.7 ± 1.6 vs 1.0 ± 2.3, P = 0.006) and were more often obese (58% vs 29%, P = 0.05) than the controls. Multivariable analyses including age, gender, and BMI z-score showed a significant decrease in CSF HA levels when the BMI z-score increased in patients (P = 0.007) but not in the controls. No association was found between CSF HA, t-MeHA, disease duration, age at disease onset, the presence of cataplexy, lumbar puncture timing, and CSF hypocretin levels. CONCLUSIONS Narcolepsy type 1 children had a higher CSF HA level together with a lower t-MeHA level leading to a significant decrease in the t-MeHA/HA ratios. These results suggest a decreased HA turnover and an impairment of histaminergic neurotransmission in narcoleptic children and support the use of a histaminergic therapy in the treatment against narcolepsy.
Collapse
Affiliation(s)
- Patricia Franco
- Integrative Physiology of the Brain Arousal System, CRNL, INSERM-U1028, CNRS, UMR5292, University Lyon1, Lyon, France.,National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic hypersomnia and Kleine-Levin Syndrome (CNR narcolepsie-hypersomnie), Bron, France.,Pediatric Sleep Unit, Mother- Children Hospital, Hospices Civils de Lyon, University Lyon1, Lyon, France
| | - Yves Dauvilliers
- National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic hypersomnia and Kleine-Levin Syndrome (CNR narcolepsie-hypersomnie), Bron, France.,Sleep Unit, Department of Neurology, Gui de Chauliac Hospital, CHU Montpellier, Montpellier, France.,Inserm, U1061, Univ Montpellier 1, Montpellier, France
| | - Clara Odilia Inocente
- Integrative Physiology of the Brain Arousal System, CRNL, INSERM-U1028, CNRS, UMR5292, University Lyon1, Lyon, France
| | - Aurore Guyon
- Integrative Physiology of the Brain Arousal System, CRNL, INSERM-U1028, CNRS, UMR5292, University Lyon1, Lyon, France.,National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic hypersomnia and Kleine-Levin Syndrome (CNR narcolepsie-hypersomnie), Bron, France.,Pediatric Sleep Unit, Mother- Children Hospital, Hospices Civils de Lyon, University Lyon1, Lyon, France
| | - Carine Villanueva
- Department of Endocrinology, Mother- Children Hospital, Hospices Civils de Lyon, University Lyon1, France
| | - Veronique Raverot
- Laboratoire de Hormonologie, Groupement Est, Hospices Civils de Lyon, University Lyon1, Lyon, France
| | - Sabine Plancoulaine
- INSERM, UMR1153, Centre of Research in Epidemiology and Statistics Sorbonne Paris Cité (CRESS), Villejuif, Paris-Descartes University, Paris, France
| | - Jian-Sheng Lin
- Integrative Physiology of the Brain Arousal System, CRNL, INSERM-U1028, CNRS, UMR5292, University Lyon1, Lyon, France
| |
Collapse
|
31
|
Lin JS, Roussel B, Gaspar A, Zhao Y, Hou Y, Schmidt M, Jouvet A, Jouvet M. The unfinished journey with modafinil and discovery of a novel population of modafinil-immunoreactive neurons. Sleep Med 2018; 49:40-52. [DOI: 10.1016/j.sleep.2018.06.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
32
|
Vodovar D, Duchêne A, Wimberley C, Leroy C, Pottier G, Dauvilliers Y, Giaume C, Lin JS, Mouthon F, Tournier N, Charvériat M. Cortico-Amygdala-Striatal Activation by Modafinil/Flecainide Combination. Int J Neuropsychopharmacol 2018; 21:687-696. [PMID: 29635319 PMCID: PMC6031015 DOI: 10.1093/ijnp/pyy027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/21/2018] [Accepted: 03/14/2018] [Indexed: 02/07/2023] Open
Abstract
Background Modafinil, a nonamphetaminic wake-promoting compound, is prescribed as first line therapy in narcolepsy, an invalidating disorder characterized by excessive daytime sleepiness and cataplexy. Although its mode of action remains incompletely known, recent studies indicated that modafinil modulates astroglial connexin-based gap junctional communication as administration of a low dose of flecainide, an astroglial connexin inhibitor, enhanced the wake-promoting and procognitive activity of modafinil in rodents and healthy volunteers. The aim of this study is to investigate changes in glucose cerebral metabolism in rodents, induced by the combination of modafinil+flecainide low dose (called THN102). Methods The impact of THN102 on brain glucose metabolism was noninvasively investigated using 18F-2-fluoro-2-deoxy-D-glucose Positron Emission Tomography imaging in Sprague-Dawley male rats. Animals were injected with vehicle, flecainide, modafinil, or THN102 and further injected with 18F-2-fluoro-2-deoxy-D-glucose followed by 60-minute Positron Emission Tomography acquisition. 18F-2-fluoro-2-deoxy-D-glucose Positron Emission Tomography images were coregistered to a rat brain template and normalized from the total brain Positron Emission Tomography signal. Voxel-to-voxel analysis was performed using SPM8 software. Comparison of brain glucose metabolism between groups was then performed. Results THN102 significantly increased regional brain glucose metabolism as it resulted in large clusters of 18F-2-fluoro-2-deoxy-D-glucose uptake localized in the cortex, striatum, and amygdala compared with control or drugs administered alone. These regions, highly involved in the regulation of sleep-wake cycle, emotions, and cognitive functions were hence quantitatively modulated by THN102. Conclusion Data presented here provide the first evidence of a regional brain activation induced by THN102, currently being tested in a phase II clinical trial in narcoleptic patients.
Collapse
Affiliation(s)
- Dominique Vodovar
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Univ. Paris Saclay, CEA-SHFJ, Orsay, France
| | | | - Catriona Wimberley
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Univ. Paris Saclay, CEA-SHFJ, Orsay, France
| | - Claire Leroy
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Univ. Paris Saclay, CEA-SHFJ, Orsay, France
| | - Géraldine Pottier
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Univ. Paris Saclay, CEA-SHFJ, Orsay, France
| | - Yves Dauvilliers
- National Reference Centre for Narcolepsy, CHU Montpellier, INSERM, France
| | - Christian Giaume
- Collège de France, Centre for Interdisciplinary Research in Biology, Paris, France
| | - Jian-Sheng Lin
- Laboratory WAKING, CRNL-INSERM U1028-CNRS UMR 5292-UCBL, Lyon, France
| | | | - Nicolas Tournier
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Univ. Paris Saclay, CEA-SHFJ, Orsay, France
| | | |
Collapse
|
33
|
Histamine N-methyltransferase regulates aggression and the sleep-wake cycle. Sci Rep 2017; 7:15899. [PMID: 29162912 PMCID: PMC5698467 DOI: 10.1038/s41598-017-16019-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 11/06/2017] [Indexed: 01/16/2023] Open
Abstract
Histamine is a neurotransmitter that regulates diverse physiological functions including the sleep-wake cycle. Recent studies have reported that histaminergic dysfunction in the brain is associated with neuropsychiatric disorders. Histamine N-methyltransferase (HNMT) is an enzyme expressed in the central nervous system that specifically metabolises histamine; yet, the exact physiological roles of HNMT are unknown. Accordingly, we phenotyped Hnmt knockout mice (KO) to determine the relevance of HNMT to various brain functions. First, we showed that HNMT deficiency enhanced brain histamine concentrations, confirming a role for HNMT in histamine inactivation. Next, we performed comprehensive behavioural testing and determined that KO mice exhibited high aggressive behaviours in the resident-intruder and aggressive biting behaviour tests. High aggression in KO mice was suppressed by treatment with zolantidine, a histamine H2 receptor (H2R) antagonist, indicating that abnormal H2R activation promoted aggression in KO mice. A sleep analysis revealed that KO mice exhibited prolonged bouts of awakening during the light (inactive) period and compensatory sleep during the dark (active) period. Abnormal sleep behaviour was suppressed by treatment with pyrilamine, a H1R antagonist, prior to light period, suggesting that excessive H1R activation led to the dysregulation of sleep-wake cycles in KO mice. These observations inform the physiological roles of HNMT.
Collapse
|
34
|
Chen Q, Luo F, Yue F, Xia J, Xiao Q, Liao X, Jiang J, Zhang J, Hu B, Gao D, He C, Hu Z. Histamine Enhances Theta-Coupled Spiking and Gamma Oscillations in the Medial Entorhinal Cortex Consistent With Successful Spatial Recognition. Cereb Cortex 2017; 28:2439-2457. [DOI: 10.1093/cercor/bhx145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Quanhui Chen
- Department of Physiology, Collaborative Innovation Center for Brain Science, Third Military Medical University, Chongqing, China
| | - Fenlan Luo
- Department of Physiology, Collaborative Innovation Center for Brain Science, Third Military Medical University, Chongqing, China
| | - Faguo Yue
- Department of Physiology, Collaborative Innovation Center for Brain Science, Third Military Medical University, Chongqing, China
| | - Jianxia Xia
- Department of Physiology, Collaborative Innovation Center for Brain Science, Third Military Medical University, Chongqing, China
| | - Qin Xiao
- Department of Physiology, Collaborative Innovation Center for Brain Science, Third Military Medical University, Chongqing, China
| | - Xiang Liao
- Brain Research Center, Third Military Medical University, Chongqing, China
| | - Jun Jiang
- Department of Basic Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Jun Zhang
- Department of Physiology, Collaborative Innovation Center for Brain Science, Third Military Medical University, Chongqing, China
| | - Bo Hu
- Department of Physiology, Collaborative Innovation Center for Brain Science, Third Military Medical University, Chongqing, China
| | - Dong Gao
- Department of Sleep and Psychology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Chao He
- Department of Physiology, Collaborative Innovation Center for Brain Science, Third Military Medical University, Chongqing, China
| | - Zhian Hu
- Department of Physiology, Collaborative Innovation Center for Brain Science, Third Military Medical University, Chongqing, China
| |
Collapse
|
35
|
Jurič DM, Kržan M, Lipnik-Stangelj M. Histamine and astrocyte function. Pharmacol Res 2016; 111:774-783. [DOI: 10.1016/j.phrs.2016.07.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/11/2016] [Accepted: 07/24/2016] [Indexed: 12/31/2022]
|
36
|
Bolam JP, Ellender TJ. Histamine and the striatum. Neuropharmacology 2016; 106:74-84. [PMID: 26275849 PMCID: PMC4917894 DOI: 10.1016/j.neuropharm.2015.08.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/30/2015] [Accepted: 08/06/2015] [Indexed: 12/25/2022]
Abstract
The neuromodulator histamine is released throughout the brain during periods of wakefulness. Combined with an abundant expression of histamine receptors, this suggests potential widespread histaminergic control of neural circuit activity. However, the effect of histamine on many of these circuits is unknown. In this review we will discuss recent evidence for histaminergic modulation of the basal ganglia circuitry, and specifically its main input nucleus; the striatum. Furthermore, we will discuss recent findings of histaminergic dysfunction in several basal ganglia disorders, including in Parkinson's disease and most prominently, in Tourette's syndrome, which has led to a resurgence of interest in this neuromodulator. Combined, these recent observations not only suggest a central role for histamine in modulating basal ganglia activity and behaviour, but also as a possible target in treating basal ganglia disorders. This article is part of the Special Issue entitled 'Histamine Receptors'.
Collapse
Affiliation(s)
- J Paul Bolam
- Department of Pharmacology, MRC Brain Network Dynamics Unit, Mansfield Road, OX1 3TH Oxford, United Kingdom
| | - Tommas J Ellender
- Department of Pharmacology, MRC Brain Network Dynamics Unit, Mansfield Road, OX1 3TH Oxford, United Kingdom.
| |
Collapse
|
37
|
Panula P, Chazot PL, Cowart M, Gutzmer R, Leurs R, Liu WLS, Stark H, Thurmond RL, Haas HL. International Union of Basic and Clinical Pharmacology. XCVIII. Histamine Receptors. Pharmacol Rev 2016; 67:601-55. [PMID: 26084539 DOI: 10.1124/pr.114.010249] [Citation(s) in RCA: 390] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Histamine is a developmentally highly conserved autacoid found in most vertebrate tissues. Its physiological functions are mediated by four 7-transmembrane G protein-coupled receptors (H1R, H2R, H3R, H4R) that are all targets of pharmacological intervention. The receptors display molecular heterogeneity and constitutive activity. H1R antagonists are long known antiallergic and sedating drugs, whereas the H2R was identified in the 1970s and led to the development of H2R-antagonists that revolutionized stomach ulcer treatment. The crystal structure of ligand-bound H1R has rendered it possible to design new ligands with novel properties. The H3R is an autoreceptor and heteroreceptor providing negative feedback on histaminergic and inhibition on other neurons. A block of these actions promotes waking. The H4R occurs on immuncompetent cells and the development of anti-inflammatory drugs is anticipated.
Collapse
Affiliation(s)
- Pertti Panula
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Paul L Chazot
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Marlon Cowart
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Ralf Gutzmer
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Rob Leurs
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Wai L S Liu
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Holger Stark
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Robin L Thurmond
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| | - Helmut L Haas
- Department of Anatomy, and Neuroscience Center, University of Helsinki, Finland (P.P.); School of Biological and Biomedical Sciences, University of Durham, United Kingdom (P.L.C.); AbbVie, Inc. North Chicago, Illinois (M.C.); Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany (R.G.); Department of Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines and Systems, VU University Amsterdam, The Netherlands (R.L.); Ziarco Pharma Limited, Canterbury, United Kingdom (W.L.S.L.); Institute of Pharmaceutical and Medical Chemistry and Institute of Neurophysiology, Medical Faculty, Westfalische-Wilhelms-University, Muenster, Germany (H.L.H.); Heinrich-Heine-University Duesseldorf, Germany (H.S.); and Janssen Research & Development, LLC, San Diego, California (R.L.T.)
| |
Collapse
|
38
|
|
39
|
Parmentier R, Zhao Y, Perier M, Akaoka H, Lintunen M, Hou Y, Panula P, Watanabe T, Franco P, Lin JS. Role of histamine H1-receptor on behavioral states and wake maintenance during deficiency of a brain activating system: A study using a knockout mouse model. Neuropharmacology 2015; 106:20-34. [PMID: 26723880 DOI: 10.1016/j.neuropharm.2015.12.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/10/2015] [Accepted: 12/14/2015] [Indexed: 10/22/2022]
Abstract
Using knockout (KO) mice lacking the histamine (HA)-synthesizing enzyme (histidine decarboxylase, HDC), we have previously shown the importance of histaminergic neurons in maintaining wakefulness (W) under behavioral challenges. Since the central actions of HA are mediated by several receptor subtypes, it remains to be determined which one(s) could be responsible for such a role. We have therefore compared the cortical-EEG, sleep and W under baseline conditions or behavioral/pharmacological stimuli in littermate wild-type (WT) and H1-receptor KO (H1-/-) mice. We found that H1-/- mice shared several characteristics with HDC KO mice, i.e. 1) a decrease in W after lights-off despite its normal baseline daily amount; 2) a decreased EEG slow wave sleep (SWS)/W power ratio; 3) inability to maintain W in response to behavioral challenges demonstrated by a decreased sleep latency when facing various stimuli. These effects were mediated by central H1-receptors. Indeed, in WT mice, injection of triprolidine, a brain-penetrating H1-receptor antagonist increased SWS, whereas ciproxifan (H3-receptor antagonist/inverse agonist) elicited W; all these injections had no effect in H1-/- mice. Finally, H1-/- mice showed markedly greater changes in EEG power (notably in the 0.8-5 Hz band) and sleep-wake cycle than in WT mice after application of a cholinergic antagonist or an indirect agonist, i.e., scopolamine or physostigmine. Hence, the role of HA in wake-promotion is largely ensured by H1-receptors. An upregulated cholinergic system may account for a quasi-normal daily amount of W in HDC or H1-receptor KO mice and likely constitutes a major compensatory mechanism when the brain is facing deficiency of an activating system. This article is part of the Special Issue entitled 'Histamine Receptors'.
Collapse
Affiliation(s)
- Régis Parmentier
- Waking Team, Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM-U1028, CNRS UMR5292, School of Medicine, Claude Bernard University, Lyon, France
| | - Yan Zhao
- Waking Team, Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM-U1028, CNRS UMR5292, School of Medicine, Claude Bernard University, Lyon, France; Department of Physiology, Zhongshan Medical College, Sun Yat-Sen University, Guangzhou, China
| | - Magali Perier
- Waking Team, Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM-U1028, CNRS UMR5292, School of Medicine, Claude Bernard University, Lyon, France
| | - Hideo Akaoka
- Waking Team, Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM-U1028, CNRS UMR5292, School of Medicine, Claude Bernard University, Lyon, France
| | - Minnamaija Lintunen
- Department of Anatomy and Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Yiping Hou
- Department of Neuroscience, Anatomy, Histology & Embryology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Pertti Panula
- Department of Anatomy and Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Takeshi Watanabe
- Unit for Immune Surveillance Research, Research Center for Allergy and Immunology, RIKEN Institute, Tsurumi-ku, Yokohama, Japan
| | - Patricia Franco
- Waking Team, Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM-U1028, CNRS UMR5292, School of Medicine, Claude Bernard University, Lyon, France
| | - Jian-Sheng Lin
- Waking Team, Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM-U1028, CNRS UMR5292, School of Medicine, Claude Bernard University, Lyon, France.
| |
Collapse
|
40
|
Fell MJ, Flik G, Dijkman U, Folgering JH, Perry KW, Johnson BJ, Westerink BH, Svensson KA. Glutamatergic regulation of brain histamine neurons: In vivo microdialysis and electrophysiology studies in the rat. Neuropharmacology 2015; 99:1-8. [DOI: 10.1016/j.neuropharm.2015.05.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 04/17/2015] [Accepted: 05/27/2015] [Indexed: 10/23/2022]
|
41
|
Yu L, Zhang XY, Cao SL, Peng SY, Ji DY, Zhu JN, Wang JJ. Na(+) -Ca(2+) Exchanger, Leak K(+) Channel and Hyperpolarization-Activated Cyclic Nucleotide-Gated Channel Comediate the Histamine-Induced Excitation on Rat Inferior Vestibular Nucleus Neurons. CNS Neurosci Ther 2015; 22:184-93. [PMID: 26387685 DOI: 10.1111/cns.12451] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 08/11/2015] [Accepted: 08/14/2015] [Indexed: 12/11/2022] Open
Abstract
AIMS Antihistaminergic drugs have traditionally been used to treat vestibular disorders in the clinic. As a potential central target for antihistaminergic drugs, the inferior vestibular nucleus (IVN) is the largest subnucleus of the central vestibular nuclear complex and is considered responsible for vestibular-autonomic responses and integration of vestibular, cerebellar, and multisensory signals. However, the role of histamine on the IVN, particularly the underlying mechanisms, is still not clear. METHODS Using whole-cell patch-clamp recordings on rat brain slices, histamine-induced effect on IVN neurons and the underlying receptor and ionic mechanisms were investigated. RESULTS We found that histamine remarkably depolarized both spontaneous firing neurons and silent neurons in IVN via both histamine H1 and histamine H2 receptors. Furthermore, Na(+) -Ca(2+) exchangers (NCXs) and background leak K(+) channels linked to H1 receptors and hyperpolarization-activated cyclic nucleotide-gated (HCN) channels coupled to H2 receptors comediate the histamine-induced depolarization on IVN neurons. CONCLUSION These results demonstrate the multiple ionic mechanisms underlying the excitatory modulation of histamine/central histaminergic system on IVN neurons and the related vestibular reflexes and functions. The findings also suggest potential targets for the treatment of vestibular disorders in the clinic, at the level of ionic channels in central vestibular nuclei.
Collapse
Affiliation(s)
- Lei Yu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biological Science and Technology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xiao-Yang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biological Science and Technology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Shu-Liang Cao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biological Science and Technology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Shi-Yu Peng
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biological Science and Technology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Deng-Yu Ji
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biological Science and Technology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Jing-Ning Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biological Science and Technology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Jian-Jun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biological Science and Technology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
42
|
De Luca R, Suvorava T, Yang D, Baumgärtel W, Kojda G, Haas HL, Sergeeva OA. Identification of histaminergic neurons through histamine 3 receptor-mediated autoinhibition. Neuropharmacology 2015; 106:102-15. [PMID: 26297536 DOI: 10.1016/j.neuropharm.2015.08.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/30/2015] [Accepted: 08/16/2015] [Indexed: 10/23/2022]
Abstract
Using a reporter mouse model with expression of the tomato fluorescent protein under the dopamine transporter promoter (Tmt-DAT) we discovered a new group of neurons in the histaminergic tuberomamillary nucleus (TMN), which, in contrast to tuberoinfundibular dopaminergic neurons of the dorsomedial arcuate nucleus, do not express tyrosine hydroxylase but can synthesize and store dopamine. Tmt-DAT neurons located within TMN share electrophysiological properties with histaminergic neurons: spontaneous firing at a membrane potential around -50 mV and presence of hyperpolarization-activated cyclic nucleotide-gated ion channels. Histamine (30 μM) depolarizes and excites Tmt-DAT neurons through H1R activation but inhibits histaminergic neurons through H3R activation thus allowing a pharmacological identification of the different neurons. Single-cell RT-PCR revealed that all histaminergic neurons expressing histidine decarboxylase (HDC) also express H3R. This includes neurons retrogradely traced from the striatum whose inhibition by a selective H3R agonist was indistinguishable from the whole population. Prolonged depolarization reduces the autoinhibition. The potency of histamine at H3R depends on membrane potential and on extracellular and intracellular calcium. Autoinhibition can be impaired by preincubation with capsaicin, a ligand of the calcium-permeable TRPV1 channel or by blockade of Ca(2+)-ATPase with thapsigargin. The pharmacology of autoinhibition is revisited and physiological conditions for its functionality are determined. Usage of reporter mouse models for the safe identification of aminergic neurons under pathophysiological conditions is recommended. This article is part of the Special Issue entitled 'Histamine Receptors'.
Collapse
Affiliation(s)
- Roberto De Luca
- Department of Neurophysiology, Heinrich-Heine-Universität, Medical Faculty, D-40225 Düsseldorf, Germany
| | - Tatsiana Suvorava
- Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-Universität, Medical Faculty, D-40225 Düsseldorf, Germany
| | - Danqing Yang
- Department of Neurophysiology, Heinrich-Heine-Universität, Medical Faculty, D-40225 Düsseldorf, Germany
| | - Wilhelm Baumgärtel
- Department of Neurophysiology, Heinrich-Heine-Universität, Medical Faculty, D-40225 Düsseldorf, Germany
| | - Georg Kojda
- Institute for Pharmacology and Clinical Pharmacology, Heinrich-Heine-Universität, Medical Faculty, D-40225 Düsseldorf, Germany
| | - Helmut L Haas
- Department of Neurophysiology, Heinrich-Heine-Universität, Medical Faculty, D-40225 Düsseldorf, Germany
| | - Olga A Sergeeva
- Department of Neurophysiology, Heinrich-Heine-Universität, Medical Faculty, D-40225 Düsseldorf, Germany.
| |
Collapse
|
43
|
Benarroch EE, Schmeichel AM, Parisi JE, Low PA. Histaminergic tuberomammillary neuron loss in multiple system atrophy and dementia with Lewy bodies. Mov Disord 2015; 30:1133-9. [DOI: 10.1002/mds.26287] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/06/2015] [Accepted: 05/11/2015] [Indexed: 12/11/2022] Open
Affiliation(s)
| | | | - Joseph E. Parisi
- Department of Neurology; Mayo Clinic; Rochester Minnesota USA
- Division of Anatomical Pathology; Mayo Clinic; Rochester Minnesota USA
| | - Phillip A. Low
- Department of Neurology; Mayo Clinic; Rochester Minnesota USA
| |
Collapse
|
44
|
Lim MM, Szymusiak R. Neurobiology of Arousal and Sleep: Updates and Insights Into Neurological Disorders. CURRENT SLEEP MEDICINE REPORTS 2015. [DOI: 10.1007/s40675-015-0013-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
45
|
Song Y, Pan L, Fu Y, Sun N, Li YJ, Cai H, Su L, Shen Y, Cui L, Shi FD. Sleep abnormality in neuromyelitis optica spectrum disorder. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e94. [PMID: 25918736 PMCID: PMC4405292 DOI: 10.1212/nxi.0000000000000094] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 01/29/2015] [Indexed: 12/20/2022]
Abstract
Objectives: We investigated the sleep structure of patients with neuromyelitis optica spectrum disorder (NMOSD) and the association of abnormalities with brain lesions. Methods: This was a prospective cross-sectional study. Thirty-three patients with NMOSD and 20 matched healthy individuals were enrolled. Demographic and clinical characteristics of patients were collected. Questionnaires were used to assess quality of sleep, daytime sleepiness, fatigue, and depression. Nocturnal polysomnography was performed. Results: Compared with healthy controls, patients with NMOSD had decreases in sleep efficiency (7%; p = 0.0341), non-REM sleep N3 (12%; p < 0.0001), and arousal index (6; p = 0.0138). REM sleep increased by 4% (p = 0.0423). There were correlations between arousal index and REM% or Epworth Sleepiness Scale (r = −0.0145; p = 0.0386, respectively). Six patients with NMOSD (18%, 5 without infratentorial lesions and 1 with infratentorial lesions) had a hypopnea index >5, and all of those with sleep apnea had predominantly the peripheral type. The periodic leg movement (PLM) index was higher in patients with NMOSD than in healthy controls (20 vs 2, p = 0.0457). Surprisingly, 77% of the patients with PLM manifested infratentorial lesions. Conclusions: Sleep architecture was markedly disrupted in patients with NMOSD. Surveillance of nocturnal symptoms and adequate symptomatic control are expected to improve the quality of life of patients with NMOSD.
Collapse
Affiliation(s)
- Yijun Song
- Departments of Neurology (Y. Song, L.P., Y.F., N.S., Y.-J.L., H.C., L.S., Y. Shen, L.C., F.-D.S.) and Immunology (F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and Department of Neurology (F.D.-S.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Liping Pan
- Departments of Neurology (Y. Song, L.P., Y.F., N.S., Y.-J.L., H.C., L.S., Y. Shen, L.C., F.-D.S.) and Immunology (F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and Department of Neurology (F.D.-S.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Ying Fu
- Departments of Neurology (Y. Song, L.P., Y.F., N.S., Y.-J.L., H.C., L.S., Y. Shen, L.C., F.-D.S.) and Immunology (F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and Department of Neurology (F.D.-S.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Na Sun
- Departments of Neurology (Y. Song, L.P., Y.F., N.S., Y.-J.L., H.C., L.S., Y. Shen, L.C., F.-D.S.) and Immunology (F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and Department of Neurology (F.D.-S.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Yu-Jing Li
- Departments of Neurology (Y. Song, L.P., Y.F., N.S., Y.-J.L., H.C., L.S., Y. Shen, L.C., F.-D.S.) and Immunology (F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and Department of Neurology (F.D.-S.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Hao Cai
- Departments of Neurology (Y. Song, L.P., Y.F., N.S., Y.-J.L., H.C., L.S., Y. Shen, L.C., F.-D.S.) and Immunology (F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and Department of Neurology (F.D.-S.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Lei Su
- Departments of Neurology (Y. Song, L.P., Y.F., N.S., Y.-J.L., H.C., L.S., Y. Shen, L.C., F.-D.S.) and Immunology (F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and Department of Neurology (F.D.-S.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Yi Shen
- Departments of Neurology (Y. Song, L.P., Y.F., N.S., Y.-J.L., H.C., L.S., Y. Shen, L.C., F.-D.S.) and Immunology (F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and Department of Neurology (F.D.-S.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Linyang Cui
- Departments of Neurology (Y. Song, L.P., Y.F., N.S., Y.-J.L., H.C., L.S., Y. Shen, L.C., F.-D.S.) and Immunology (F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and Department of Neurology (F.D.-S.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Fu-Dong Shi
- Departments of Neurology (Y. Song, L.P., Y.F., N.S., Y.-J.L., H.C., L.S., Y. Shen, L.C., F.-D.S.) and Immunology (F.-D.S.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and Department of Neurology (F.D.-S.), Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| |
Collapse
|
46
|
Yang YC, Hu CC, Lai YC. Non-additive modulation of synaptic transmission by serotonin, adenosine, and cholinergic modulators in the sensory thalamus. Front Cell Neurosci 2015; 9:60. [PMID: 25852468 PMCID: PMC4360759 DOI: 10.3389/fncel.2015.00060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 02/09/2015] [Indexed: 11/13/2022] Open
Abstract
The thalamus relays sensory information to the cortex. Oscillatory activities of the thalamocortical network are modulated by monoamines, acetylcholine, and adenosine, and could be the key features characteristic of different vigilance states. Although the thalamus is almost always subject to the actions of more than just one neuromodulators, reports on the modulatory effect of coexisting neuromodulators on thalamic synaptic transmission are unexpectedly scarce. We found that, if present alone, monoamine or adenosine decreases retinothalamic synaptic strength and short-term depression, whereas cholinergic modulators generally enhance postsynaptic response to presynaptic activity. However, coexistence of different modulators tends to produce non-additive effect, not predictable based on the action of individual modulators. Acetylcholine, acting via nicotinic receptors, can interact with either serotonin or adenosine to abolish most short-term synaptic depression. Moreover, the coexistence of adenosine and monoamine, with or without acetylcholine, results in robustly decreased synaptic strength and transforms short-term synaptic depression to facilitation. These findings are consistent with a view that acetylcholine is essential for an "enriched" sensory flow through the thalamus, and the flow is trimmed down by concomitant monoamine or adenosine (presumably for the wakefulness and rapid-eye movement, or REM, sleep states, respectively). In contrast, concomitant adenosine and monoamine would lead to a markedly "deprived" (and high-pass filtered) sensory flow, and thus the dramatic decrease of monoamine may constitute the basic demarcation between non-REM and REM sleep. The collective actions of different neuromodulators on thalamic synaptic transmission thus could be indispensable for the understanding of network responsiveness in different vigilance states.
Collapse
Affiliation(s)
- Ya-Chin Yang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University Tao-Yuan, Taiwan ; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Chun-Chang Hu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University Tao-Yuan, Taiwan ; Department of Neurosurgery, Chang-Gung Memorial Hospital Linkou, Taiwan
| | - Yi-Chen Lai
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| |
Collapse
|
47
|
Abstract
Sleep and wake are fundamental behavioral states whose molecular regulation remains mysterious. Brain states and body functions change dramatically between sleep and wake, are regulated by circadian and homeostatic processes, and depend on the nutritional and emotional condition of the animal. Sleep-wake transitions require the coordination of several brain regions and engage multiple neurochemical systems, including neuropeptides. Neuropeptides serve two main functions in sleep-wake regulation. First, they represent physiological states such as energy level or stress in response to environmental and internal stimuli. Second, neuropeptides excite or inhibit their target neurons to induce, stabilize, or switch between sleep-wake states. Thus, neuropeptides integrate physiological subsystems such as circadian time, previous neuron usage, energy homeostasis, and stress and growth status to generate appropriate sleep-wake behaviors. We review the roles of more than 20 neuropeptides in sleep and wake to lay the foundation for future studies uncovering the mechanisms that underlie the initiation, maintenance, and exit of sleep and wake states.
Collapse
Affiliation(s)
- Constance Richter
- Department of Molecular and Cellular Biology, Center for Brain Science, Division of Sleep Biology, Harvard University, Cambridge, Massachusetts 02138; ,
| | | | | |
Collapse
|
48
|
Jang SH, Kwon HG. The ascending reticular activating system from pontine reticular formation to the hypothalamus in the human brain: A diffusion tensor imaging study. Neurosci Lett 2015; 590:58-61. [DOI: 10.1016/j.neulet.2015.01.071] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 01/02/2015] [Accepted: 01/28/2015] [Indexed: 01/03/2023]
|
49
|
He C, Luo F, Chen X, Chen F, Li C, Ren S, Qiao Q, Zhang J, de Lecea L, Gao D, Hu Z. Superficial Layer-Specific Histaminergic Modulation of Medial Entorhinal Cortex Required for Spatial Learning. Cereb Cortex 2015; 26:1590-1608. [PMID: 25595181 DOI: 10.1093/cercor/bhu322] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The medial entorhinal cortex (MEC) plays a crucial role in spatial learning and memory. Whereas the MEC receives a dense histaminergic innervation from the tuberomamillary nucleus of the hypothalamus, the functions of histamine in this brain region remain unclear. Here, we show that histamine acts via H1Rs to directly depolarize the principal neurons in the superficial, but not deep, layers of the MEC when recording at somata. Moreover, histamine decreases the spontaneous GABA, but not glutamate, release onto principal neurons in the superficial layers by acting at presynaptic H3Rs without effect on synaptic release in the deep layers. Histamine-induced depolarization is mediated via inhibition of Kir channels and requires the activation of protein kinase C, whereas the inhibition of spontaneous GABA release by histamine depends on voltage-gated Ca(2+) channels and extracellular Ca(2+). Furthermore, microinjection of the H1R or H3R, but not H2R, antagonist respectively into the superficial, but not deep, layers of MEC impairs rat spatial learning as assessed by water maze tasks but does not affect the motor function and exploratory activity in an open field. Together, our study indicates that histamine plays an essential role in spatial learning by selectively regulating neuronal excitability and synaptic transmission in the superficial layers of the MEC.
Collapse
Affiliation(s)
- Chao He
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Fenlan Luo
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Xingshu Chen
- Department of Histology and Embryology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Fang Chen
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Chao Li
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Shuancheng Ren
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Qicheng Qiao
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Jun Zhang
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA 94304, USA
| | - Dong Gao
- Department of Sleep and Psychology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Zhian Hu
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| |
Collapse
|
50
|
Yu X, Zecharia A, Zhang Z, Yang Q, Yustos R, Jager P, Vyssotski AL, Maywood ES, Chesham JE, Ma Y, Brickley SG, Hastings MH, Franks NP, Wisden W. Circadian factor BMAL1 in histaminergic neurons regulates sleep architecture. Curr Biol 2014; 24:2838-44. [PMID: 25454592 PMCID: PMC4252164 DOI: 10.1016/j.cub.2014.10.019] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/29/2014] [Accepted: 10/08/2014] [Indexed: 12/24/2022]
Abstract
Circadian clocks allow anticipation of daily environmental changes. The suprachiasmatic nucleus (SCN) houses the master clock, but clocks are also widely expressed elsewhere in the body. Although some peripheral clocks have established roles, it is unclear what local brain clocks do. We tested the contribution of one putative local clock in mouse histaminergic neurons in the tuberomamillary nucleus to the regulation of the sleep-wake cycle. Histaminergic neurons are silent during sleep, and start firing after wake onset; the released histamine, made by the enzyme histidine decarboxylase (HDC), enhances wakefulness. We found that hdc gene expression varies with time of day. Selectively deleting the Bmal1 (also known as Arntl or Mop3) clock gene from histaminergic cells removes this variation, producing higher HDC expression and brain histamine levels during the day. The consequences include more fragmented sleep, prolonged wake at night, shallower sleep depth (lower nonrapid eye movement [NREM] δ power), increased NREM-to-REM transitions, hindered recovery sleep after sleep deprivation, and impaired memory. Removing BMAL1 from histaminergic neurons does not, however, affect circadian rhythms. We propose that for mammals with polyphasic/nonwake consolidating sleep, the local BMAL1-dependent clock directs appropriately timed declines and increases in histamine biosynthesis to produce an appropriate balance of wake and sleep within the overall daily cycle of rest and activity specified by the SCN.
Collapse
Affiliation(s)
- Xiao Yu
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, Exhibition Road, London SW7 2AZ, UK
| | - Anna Zecharia
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, Exhibition Road, London SW7 2AZ, UK
| | - Zhe Zhang
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, Exhibition Road, London SW7 2AZ, UK
| | - Qianzi Yang
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, Exhibition Road, London SW7 2AZ, UK
| | - Raquel Yustos
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, Exhibition Road, London SW7 2AZ, UK
| | - Polona Jager
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, Exhibition Road, London SW7 2AZ, UK
| | - Alexei L Vyssotski
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Winterhurerstrasse 190, Zurich 8057, Switzerland
| | - Elizabeth S Maywood
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Johanna E Chesham
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Ying Ma
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, Exhibition Road, London SW7 2AZ, UK
| | - Stephen G Brickley
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, Exhibition Road, London SW7 2AZ, UK
| | - Michael H Hastings
- Neurobiology Division, Medical Research Council Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Nicholas P Franks
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, Exhibition Road, London SW7 2AZ, UK.
| | - William Wisden
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, Exhibition Road, London SW7 2AZ, UK.
| |
Collapse
|