1
|
Khalaf A, Lopez E, Li J, Horn A, Edlow BL, Blumenfeld H. Shared subcortical arousal systems across sensory modalities during transient modulation of attention. Neuroimage 2025; 312:121224. [PMID: 40250641 DOI: 10.1016/j.neuroimage.2025.121224] [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: 02/03/2025] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025] Open
Abstract
Subcortical arousal systems are known to play a key role in controlling sustained changes in attention and conscious awareness. Recent studies indicate that these systems have a major influence on short-term dynamic modulation of visual attention, but their role across sensory modalities is not fully understood. In this study, we investigated shared subcortical arousal systems across sensory modalities during transient changes in attention using block and event-related fMRI paradigms. We analyzed massive publicly available fMRI datasets collected while 1561 participants performed visual, auditory, tactile, and taste perception tasks. Our analyses revealed a shared circuit of subcortical arousal systems exhibiting early transient increases in activity in midbrain reticular formation and central thalamus across perceptual modalities, as well as less consistent increases in pons, hypothalamus, basal forebrain, and basal ganglia. Identifying these networks is critical for understanding mechanisms of normal attention and consciousness and may help facilitate subcortical targeting for therapeutic neuromodulation.
Collapse
Affiliation(s)
- Aya Khalaf
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Erick Lopez
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Jian Li
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Andreas Horn
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Neurology, Center for Brain Circuit Therapeutics, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Movement Disorders & Neuromodulation Section, Department of Neurology, Charité - Universitätsmedizin, Berlin, Germany
| | - Brian L Edlow
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Hal Blumenfeld
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA; Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
2
|
Yu J, Liu H, Gao R, Wang TV, Li C, Liu Y, Yang L, Xu Y, Cui Y, Jia C, Huang J, Chen PR, Rao Y. Calcineurin: An essential regulator of sleep revealed by biochemical, chemical biological, and genetic approaches. Cell Chem Biol 2025; 32:157-173.e7. [PMID: 39740665 DOI: 10.1016/j.chembiol.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/29/2024] [Accepted: 12/09/2024] [Indexed: 01/02/2025]
Abstract
Research into mechanisms underlying sleep traditionally relies on electrophysiology and genetics. Because sleep can only be measured on whole animals by behavioral observations and physical means, no sleep research was initiated by biochemical and chemical biological approaches. We used phosphorylation sites of kinases important for sleep as targets for biochemical and chemical biological approaches. Sleep was increased in mice carrying a threonine-to-alanine substitution at residue T469 of salt-inducible kinase 3 (SIK3). Our biochemical purification and photo-crosslinking revealed calcineurin (CaN) dephosphorylation, both in vitro and in vivo, of SIK3 at T469 and S551, but not T221. Knocking down CaN regulatory subunit reduced daily sleep by more than 5 h, exceeding all known mouse mutants. Our work uncovered a critical physiological role for CaN in sleep and pioneered biochemical purification and chemical biology as effective approaches to study sleep.
Collapse
Affiliation(s)
- Jianjun Yu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Huijie Liu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Rui Gao
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Tao V Wang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Chenggang Li
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Yuxiang Liu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Lu Yang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Ying Xu
- National Center for Protein Sciences Phoenix, Beijing, China
| | - Yunfeng Cui
- Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Chenxi Jia
- National Center for Protein Sciences Phoenix, Beijing, China
| | - Juan Huang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Peng R Chen
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Yi Rao
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Yang W, Shi J, Li C, Yang J, Yu J, Huang J, Rao Y. Calcium/calmodulin-dependent protein kinase II α and β differentially regulate mammalian sleep. Commun Biol 2025; 8:11. [PMID: 39757286 DOI: 10.1038/s42003-024-07449-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/30/2024] [Indexed: 01/07/2025] Open
Abstract
While sleep is important, our understanding of its molecular mechanisms is limited. Over the last two decades, protein kinases including Ca2+/calmodulin-dependent protein kinase II (CaMKII) α and β have been implicated in sleep regulation. Of all the known mouse genetic mutants, the biggest changes in sleep is reported to be observed in adult mice with sgRNAs for Camk2b injected into their embryos: sleep is reduced by approximately 120 min (mins) over 24 h (hrs). We have reexamined the sleep phenotype in mice with either Camk2a or Camk2b gene knocked-out by conventional gene targetting. While the basal sleep is reduced in Camk2a knockout mice, it remains unaltered in Camk2b mutants. Knockout of either Camk2a or Camk2b reduces sleep rebound after deprivation, indicating their roles in sleep homeostasis. These results indicate the involvement of CaMKIIα in both basal sleep and sleep homeostasis while CaMKIIβ is mainly required physiologically for sleep homeostasis, serving as a stimulus for rigorous studies in the future.
Collapse
Affiliation(s)
- Weiwen Yang
- Chinese Institute of Brain Research, Beijing (CIBR), and Chinese Institutes for Medical Research, Beijing (CIMR), Capital Medical University, Beijing, China
- Laboratory of Neurochemical Biology, Peking-Tsinghua-NIBS (PTN) Graduate Program, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences at the Health Sciences Center, Peking University, Beijing, China
| | - Jingyi Shi
- Laboratory of Neurochemical Biology, Peking-Tsinghua-NIBS (PTN) Graduate Program, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences at the Health Sciences Center, Peking University, Beijing, China
| | - Chenggang Li
- Chinese Institute of Brain Research, Beijing (CIBR), and Chinese Institutes for Medical Research, Beijing (CIMR), Capital Medical University, Beijing, China
- Laboratory of Neurochemical Biology, Peking-Tsinghua-NIBS (PTN) Graduate Program, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences at the Health Sciences Center, Peking University, Beijing, China
| | - Jingqun Yang
- Chinese Institute of Brain Research, Beijing (CIBR), and Chinese Institutes for Medical Research, Beijing (CIMR), Capital Medical University, Beijing, China
| | - Jianjun Yu
- Chinese Institute of Brain Research, Beijing (CIBR), and Chinese Institutes for Medical Research, Beijing (CIMR), Capital Medical University, Beijing, China
- Laboratory of Neurochemical Biology, Peking-Tsinghua-NIBS (PTN) Graduate Program, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences at the Health Sciences Center, Peking University, Beijing, China
| | - Juan Huang
- Chinese Institute of Brain Research, Beijing (CIBR), and Chinese Institutes for Medical Research, Beijing (CIMR), Capital Medical University, Beijing, China
- Laboratory of Neurochemical Biology, Peking-Tsinghua-NIBS (PTN) Graduate Program, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences at the Health Sciences Center, Peking University, Beijing, China
| | - Yi Rao
- Chinese Institute of Brain Research, Beijing (CIBR), and Chinese Institutes for Medical Research, Beijing (CIMR), Capital Medical University, Beijing, China.
- Laboratory of Neurochemical Biology, Peking-Tsinghua-NIBS (PTN) Graduate Program, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences at the Health Sciences Center, Peking University, Beijing, China.
| |
Collapse
|
4
|
Lin J, Luo Z, Fan M, Liu Y, Shi X, Cai Y, Yang Z, Chen L, Pan J. Abnormal hypothalamic functional connectivity and serum arousal-promoting neurotransmitters in insomnia disorder patients: a pilot study. PeerJ 2024; 12:e18540. [PMID: 39583108 PMCID: PMC11586044 DOI: 10.7717/peerj.18540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/27/2024] [Indexed: 11/26/2024] Open
Abstract
Objective The present study aimed to investigate the functional connectivity (FC) of the anterior and posterior hypothalamus with the whole brain in insomnia disorder (ID) patients. Additionally, we explored the relationship between FC values and serum levels of arousal-promoting neurotransmitters (orexin-A and histamine) in ID patients. Methods This study enrolled 30 ID patients and 30 age- and gender-matched healthy controls. Resting-state functional magnetic resonance imaging (RS-fMRI) was employed to assess the FC of the anterior and posterior hypothalamus with the whole brain. Serum concentrations of orexin-A and histamine were measured using enzyme-linked immunosorbent assay (ELISA). Moreover, Spearman correlation analysis was conducted to investigate the relationship between FC values and serum levels of arousal-promoting neurotransmitters in ID patients. Results Our findings showed decreased FC between the posterior hypothalamus and several brain regions including the bilateral orbital superior frontal gyrus, the bilateral angular gyrus, the right anterior cingulate cortex, the left precuneus, and the right medial superior frontal gyrus in ID patients. Additionally, decreased FC was observed between the anterior hypothalamus and the right anterior cingulate cortex among ID patients. Compared to the healthy controls, ID patients showed significantly elevated serum concentrations of orexin-A and histamine. Furthermore, we identified a positive correlation between the FC of the right medial superior frontal gyrus with posterior hypothalamus and histamine levels in ID patients. Conclusion ID patients exhibited aberrant FC in brain regions related to sleep-wake regulation, particularly involving the default mode network and anterior cingulate cortex, which may correlate with the peripheral levels of histamine. These findings contribute to our understanding of the potential neuroimaging and neurohumoral mechanism underlying ID patients.
Collapse
Affiliation(s)
- Jingjing Lin
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Zhenye Luo
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Mei Fan
- Department of Psychiatry, The First Affiliated Hospital of USTC, Hefei, Anhui Province, China
| | - Yaxi Liu
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Xian Shi
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Yixian Cai
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Zhiyun Yang
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Liting Chen
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Jiyang Pan
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
5
|
Khalaf A, Lopez E, Li J, Horn A, Edlow BL, Blumenfeld H. Shared subcortical arousal systems across sensory modalities during transient modulation of attention. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613316. [PMID: 39345640 PMCID: PMC11429725 DOI: 10.1101/2024.09.16.613316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Subcortical arousal systems are known to play a key role in controlling sustained changes in attention and conscious awareness. Recent studies indicate that these systems have a major influence on short-term dynamic modulation of visual attention, but their role across sensory modalities is not fully understood. In this study, we investigated shared subcortical arousal systems across sensory modalities during transient changes in attention using block and event-related fMRI paradigms. We analyzed massive publicly available fMRI datasets collected while 1,561 participants performed visual, auditory, tactile, and taste perception tasks. Our analyses revealed a shared circuit of subcortical arousal systems exhibiting early transient increases in activity in midbrain reticular formation and central thalamus across perceptual modalities, as well as less consistent increases in pons, hypothalamus, basal forebrain, and basal ganglia. Identifying these networks is critical for understanding mechanisms of normal attention and consciousness and may help facilitate subcortical targeting for therapeutic neuromodulation.
Collapse
Affiliation(s)
- Aya Khalaf
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Erick Lopez
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Jian Li
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Andreas Horn
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Brain Circuit Therapeutics, Department of Neurology, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Movement Disorders & Neuromodulation Section, Department of Neurology, Charité – Universitätsmedizin, Berlin, Germany
| | - Brian L. Edlow
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Hal Blumenfeld
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
6
|
Bonifazi A, Del Bello F, Giorgioni G, Piergentili A, Saab E, Botticelli L, Cifani C, Micioni Di Bonaventura E, Micioni Di Bonaventura MV, Quaglia W. Targeting orexin receptors: Recent advances in the development of subtype selective or dual ligands for the treatment of neuropsychiatric disorders. Med Res Rev 2023; 43:1607-1667. [PMID: 37036052 DOI: 10.1002/med.21959] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/08/2023] [Accepted: 03/28/2023] [Indexed: 04/11/2023]
Abstract
Orexin-A and orexin-B, also named hypocretin-1 and hypocretin-2, are two hypothalamic neuropeptides highly conserved across mammalian species. Their effects are mediated by two distinct G protein-coupled receptors, namely orexin receptor type 1 (OX1-R) and type 2 (OX2-R), which share 64% amino acid identity. Given the wide expression of OX-Rs in different central nervous system and peripheral areas and the several pathophysiological functions in which they are involved, including sleep-wake cycle regulation (mainly mediated by OX2-R), emotion, panic-like behaviors, anxiety/stress, food intake, and energy homeostasis (mainly mediated by OX1-R), both subtypes represent targets of interest for many structure-activity relationship (SAR) campaigns carried out by pharmaceutical companies and academies. However, before 2017 the research was predominantly directed towards dual-orexin ligands, and limited chemotypes were investigated. Analytical characterizations, including resolved structures for both OX1-R and OX2-R in complex with agonists and antagonists, have improved the understanding of the molecular basis of receptor recognition and are assets for medicinal chemists in the design of subtype-selective ligands. This review is focused on the medicinal chemistry aspects of small molecules acting as dual or subtype selective OX1-R/OX2-R agonists and antagonists belonging to different chemotypes and developed in the last years, including radiolabeled OX-R ligands for molecular imaging. Moreover, the pharmacological effects of the most studied ligands in different neuropsychiatric diseases, such as sleep, mood, substance use, and eating disorders, as well as pain, have been discussed. Poly-pharmacology applications and multitarget ligands have also been considered.
Collapse
Affiliation(s)
- Alessandro Bonifazi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, Maryland, United States
| | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Camerino, Italy
| | - Gianfabio Giorgioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Camerino, Italy
| | | | - Elizabeth Saab
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, Maryland, United States
| | - Luca Botticelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | | | | | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Camerino, Italy
| |
Collapse
|
7
|
Zhang M, Thieux M, Arvis L, Lin JS, Guyon A, Plancoulaine S, Villanueva C, Franco P. Metabolic disturbances in children with narcolepsy: a retrospective study. Sleep 2023; 46:zsad076. [PMID: 36971181 DOI: 10.1093/sleep/zsad076] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/22/2022] [Indexed: 07/20/2023] Open
Abstract
STUDY OBJECTIVES To determine the prevalence of metabolic syndrome (MS) in children with narcolepsy and to evaluate their clinical and sleep characteristics according to the different components of MS. METHODS This retrospective study consisted of 58 de novo children with narcolepsy (median age: 12.7 years, 48.3% of boys). The recently published MS criteria in a French population of children were used. Clinical and sleep characteristics were compared between groups with different components of MS. RESULTS MS was present in 17.2% of children with narcolepsy, among whom 79.3% presented with high homeostasis model assessment for insulin resistance (HOMA-IR), 25.9% with high body mass index, 24.1% with low high-density lipoprotein cholesterol (HDL-C), and 12.1% with high triglycerides. Patients with at least two MS components had more night eating behaviors and tended to have lower percentage of slow-wave sleep and more fragmented sleep. On multiple sleep latency test, they had shorter mean sleep latencies to rapid eye movement (REM), non-REM sleep and tended to have more sleep onset REM periods (SOREMPs) than those with less than two MS components. CONCLUSIONS Insulin resistance was found to be the core metabolic disturbance in obese as well as in nonobese children with narcolepsy. Children with narcolepsy with at least two MS components presented a more severe daytime sleepiness and a higher prevalence of night-eating behaviors than those with less than two MS components. Such children might benefit from early evaluation and management in order to prevent future complications.
Collapse
Affiliation(s)
- Min Zhang
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
| | - Marine Thieux
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
- Pediatric Sleep Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon and National Reference Center for Narcolepsy, University of Lyon1, Lyon, France
| | - Laura Arvis
- Pediatric endocrinology Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Jian-Sheng Lin
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
| | - Aurore Guyon
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
- Pediatric Sleep Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon and National Reference Center for Narcolepsy, University of Lyon1, Lyon, France
| | | | - Carine Villanueva
- Pediatric endocrinology Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Patricia Franco
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
- Pediatric Sleep Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon and National Reference Center for Narcolepsy, University of Lyon1, Lyon, France
| |
Collapse
|
8
|
Seugnet L, Anaclet C, Perier M, Ghersi‐Egea J, Lin J. A marked enhancement of a BLOC-1 gene, pallidin, associated with somnolent mouse models deficient in histamine transmission. CNS Neurosci Ther 2023; 29:483-486. [PMID: 36258293 PMCID: PMC9804043 DOI: 10.1111/cns.13995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 02/06/2023] Open
Affiliation(s)
- Laurent Seugnet
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292Claude Bernard University Lyon 1BronFrance
| | - Christelle Anaclet
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292Claude Bernard University Lyon 1BronFrance
- Department of Neurological SurgeryUniversity of California, Davis School of MedicineSacramentoUSA
| | - Magali Perier
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292Claude Bernard University Lyon 1BronFrance
| | - Jean‐François Ghersi‐Egea
- Fluids and Barriers of the Central Nervous System, Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292Claude Bernard University Lyon 1BronFrance
| | - Jian‐Sheng Lin
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292Claude Bernard University Lyon 1BronFrance
| |
Collapse
|
9
|
Dhafar HO, BaHammam AS. Body Weight and Metabolic Rate Changes in Narcolepsy: Current Knowledge and Future Directions. Metabolites 2022; 12:1120. [PMID: 36422261 PMCID: PMC9693066 DOI: 10.3390/metabo12111120] [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] [Received: 10/23/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 08/26/2023] Open
Abstract
Narcolepsy is a known auto-immune disease that presents mainly in the teenage years with irresistible sleep attacks. Patients with narcolepsy, especially NT1, have been found to have a high prevalence of obesity and other metabolic derangements. This narrative review aimed to address the relationship between narcolepsy and changes in weight and metabolic rate, and discuss potential mechanisms for weight gain and metabolic changes and future research agendas on this topic. This article will provide a balanced, up-to-date critical review of the current literature, and delineate areas for future research, in order to understand the pathophysiological metabolic changes in narcolepsy. Articles using predefined keywords were searched for in PubMed and Google Scholar databases, with predefined inclusion and exclusion criteria. Compared to controls, patients with narcolepsy are more likely to be obese and have higher BMIs and waist circumferences. According to recent research, weight gain in narcolepsy patients may be higher during the disease's outset. The precise mechanisms causing this weight gain remains unknown. The available information, albeit limited, does not support differences in basal or resting metabolic rates between patients with narcolepsy and controls, other than during the time of disease onset. The evidence supporting the role of orexin in weight gain in humans with narcolepsy is still controversial, in the literature. Furthermore, the available data did not show any appreciable alterations in the levels of CSF melanin-concentrating hormone, plasma and CSF leptin, or serum growth hormone, in relation to weight gain. Other mechanisms have been proposed, including a reduction in sympathetic tone, hormonal changes, changes in eating behavior and physical activity, and genetic predisposition. The association between increased body mass index and narcolepsy is well-recognized; however, the relationship between narcolepsy and other metabolic measures, such as body fat/muscle distribution and metabolic rate independent of BMI, is not well documented, and the available evidence is inconsistent. Future longitudinal studies with larger sample sizes are needed to assess BMR in patients with narcolepsy under a standard protocol at the outset of narcolepsy, with regular follow-up.
Collapse
Affiliation(s)
- Hamza O. Dhafar
- The University Sleep Disorders Center, Department of Medicine, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
- Department of Family Medicine, Prince Mansour Military Hospital, Taif 26526, Saudi Arabia
| | - Ahmed S. BaHammam
- The University Sleep Disorders Center, Department of Medicine, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
- The Strategic Technologies Program of the National Plan for Sciences and Technology and Innovation in the Kingdom of Saudi Arabia, P.O. Box 2454, Riyadh 11324, Saudi Arabia
| |
Collapse
|
10
|
Ngo Q, Plante DT. An Update on the Misuse and Abuse Potential of Pharmacological Treatments for Central Disorders of Hypersomnolence. CURRENT SLEEP MEDICINE REPORTS 2022. [DOI: 10.1007/s40675-022-00227-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
11
|
Zhang M, Thieux M, Inocente CO, Vieux N, Arvis L, Villanueva C, Lin J, Plancoulaine S, Guyon A, Franco P. Characterization of rapid weight gain phenotype in children with narcolepsy. CNS Neurosci Ther 2022; 28:829-841. [PMID: 35212159 PMCID: PMC9062543 DOI: 10.1111/cns.13811] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVES To characterize the rapid weight gain (RWG) phenotype associated with the onset of childhood narcolepsy and to determine whether it could constitute a marker of severity of the disease. METHODS RWG was defined using the BMI z-score slope reported to one year (>0.67 SD) from symptom onset to disease diagnosis. We compared the clinical, metabolic, and sleep characteristics between patients with or without RWG at diagnosis. Pharmacological management, anthropometric, and clinical progression were also evaluated during the follow-up. RESULTS A total of 84 de novo narcoleptic pediatric patients were included; their median age at diagnosis was 12.0 years; 59.5% boys, 90.5% cataplexy, and 98.7% HLA-DQB1*06:02, 57% had RWG profile. RWG patients were younger at diagnosis than non-RWG patients, despite a shorter diagnostic delay. They had a higher BMI z-score and a higher prevalence of obesity at diagnosis, but not at symptom onset, and higher adapted Epworth Sleepiness Scale and Insomnia Severity Index scores than non-RWG patients. No differences on nocturnal polysomnography and multiple sleep latency tests were found between groups at disease diagnosis. After a median follow-up of 5 years, RWG patients still had a higher BMI z-score and a higher prevalence of obesity despite benefiting from the same therapeutic management and displaying improvement in sleepiness and school difficulties. CONCLUSIONS Narcoleptic RWG patients were younger, sleepier, and the prevalence of obesity was higher at diagnosis despite a shorter diagnostic delay than that of non-RWG patients. These patients had also a higher risk of developing a long-term obesity, despite a positive progression of their narcoleptic symptoms. RGW could then represent a maker of a more severe phenotype of childhood narcolepsy, which should inspire a prompt and more offensive management to prevent obesity and its complications.
Collapse
Affiliation(s)
- Min Zhang
- Integrative Physiology of the Brain Arousal SystemsCRNLINSERM U1028CNRS UMR5292Université Claude Bernard Lyon 1LyonFrance
| | - Marine Thieux
- Integrative Physiology of the Brain Arousal SystemsCRNLINSERM U1028CNRS UMR5292Université Claude Bernard Lyon 1LyonFrance
- Pediatric Sleep UnitHôpital Femme Mère EnfantHospices Civils de Lyon & National Reference Center for NarcolepsyUniversité Claude Bernard Lyon 1LyonFrance
| | - Clara Odilia Inocente
- Integrative Physiology of the Brain Arousal SystemsCRNLINSERM U1028CNRS UMR5292Université Claude Bernard Lyon 1LyonFrance
| | - Noemie Vieux
- Pediatric Sleep UnitHôpital Femme Mère EnfantHospices Civils de Lyon & National Reference Center for NarcolepsyUniversité Claude Bernard Lyon 1LyonFrance
| | - Laura Arvis
- Pediatric Endocrinology UnitHôpital Femme Mère EnfantHospices Civils de LyonUniversité Claude Bernard Lyon 1LyonFrance
| | - Carine Villanueva
- Pediatric Endocrinology UnitHôpital Femme Mère EnfantHospices Civils de LyonUniversité Claude Bernard Lyon 1LyonFrance
| | - Jian‐Sheng Lin
- Integrative Physiology of the Brain Arousal SystemsCRNLINSERM U1028CNRS UMR5292Université Claude Bernard Lyon 1LyonFrance
| | | | - Aurore Guyon
- Pediatric Sleep UnitHôpital Femme Mère EnfantHospices Civils de Lyon & National Reference Center for NarcolepsyUniversité Claude Bernard Lyon 1LyonFrance
| | - Patricia Franco
- Integrative Physiology of the Brain Arousal SystemsCRNLINSERM U1028CNRS UMR5292Université Claude Bernard Lyon 1LyonFrance
- Pediatric Sleep UnitHôpital Femme Mère EnfantHospices Civils de Lyon & National Reference Center for NarcolepsyUniversité Claude Bernard Lyon 1LyonFrance
| |
Collapse
|
12
|
Okechukwu CE. The neurophysiologic basis of the human sleep–wake cycle and the physiopathology of the circadian clock: a narrative review. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2022. [DOI: 10.1186/s41983-022-00468-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractThe objectives of this review were to explain the neurologic processes that control the human sleep–wake cycle as well as the pathophysiology of the human circadian clock. Non-rapid eye movement and rapid eye movement sleep are the two main phases of sleep. When triggered by circadian input from the anterior hypothalamus and sleep–wake homeostatic information from endogenous chemical signals (example, adenosine), the ventrolateral preoptic nucleus initiates the onset of sleep. Arousal in which there is a conscious monitoring of the surroundings and the ability to respond to external stimuli is known as wakefulness. It contrasts the state of sleep, in which receptivity to external stimuli is reduced. The higher the synchronous firing rates of cerebral cortex neurons, the longer the brain has been awake. Sleep–wake disturbances induced by endogenous circadian system disruptions or desynchronization between internal and external sleep–wake cycles are known as circadian rhythm sleep–wake disorder (CRSWD). Patients with CRSWD usually report chronic daytime drowsiness and/or insomnia, which interferes with their activities. CRSWD is diagnosed based on the results of some functional evaluations, which include measuring the circadian phase using core body temperature, melatonin secretion timing, sleep diaries, actigraphy, and subjective experiences (example, using the Morningness–Eveningness Questionnaire). CRSWD is classified as a dyssomnia in the second edition of the International Classification of Sleep Disorders, with six subtypes: advanced sleep phase, delayed sleep phase, irregular sleep–wake, free running, jet lag, and shift work types. CRSWD can be temporary (due to jet lag, shift work, or illness) or chronic (due to delayed sleep–wake phase disorder, advanced sleep–wake phase disorder, non-24-h sleep–wake disorder, or irregular sleep–wake rhythm disorder). The inability to fall asleep and wake up at the desired time is a common symptom of all CRSWDs.
Collapse
|
13
|
Sun Y, Tisdale R, Park S, Ma SC, Heu J, Haire M, Allocca G, Yamanaka A, Morairty SR, Kilduff TS. The development of sleep/wake disruption and cataplexy as hypocretin/orexin neurons degenerate in male vs. female Orexin/tTA; TetO-DTA Mice. Sleep 2022; 45:6532492. [PMID: 35182424 PMCID: PMC9742901 DOI: 10.1093/sleep/zsac039] [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] [Received: 10/12/2021] [Revised: 12/23/2021] [Indexed: 01/10/2023] Open
Abstract
Narcolepsy Type 1 (NT1), a sleep disorder with similar prevalence in both sexes, is thought to be due to loss of the hypocretin/orexin (Hcrt) neurons. Several transgenic strains have been created to model this disorder and are increasingly being used for preclinical drug development and basic science studies, yet most studies have solely used male mice. We compared the development of narcoleptic symptomatology in male vs. female orexin-tTA; TetO-DTA mice, a model in which Hcrt neuron degeneration can be initiated by removal of doxycycline (DOX) from the diet. EEG, EMG, subcutaneous temperature, gross motor activity, and video recordings were conducted for 24-h at baseline and 1, 2, 4, and 6 weeks after DOX removal. Female DTA mice exhibited cataplexy, the pathognomonic symptom of NT1, by Week 1 in the DOX(-) condition but cataplexy was not consistently present in males until Week 2. By Week 2, both sexes showed an impaired ability to sustain long wake bouts during the active period, the murine equivalent of excessive daytime sleepiness in NT1. Subcutaneous temperature appeared to be regulated at lower levels in both sexes as the Hcrt neurons degenerated. During degeneration, both sexes also exhibited the "Delta State", characterized by sudden cessation of activity, high delta activity in the EEG, maintenance of muscle tone and posture, and the absence of phasic EMG activity. Since the phenotypes of the two sexes were indistinguishable by Week 6, we conclude that both sexes can be safely combined in future studies to reduce cost and animal use.
Collapse
Affiliation(s)
- Yu Sun
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | | | | | - Shun-Chieh Ma
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | - Jasmine Heu
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | - Meghan Haire
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | | | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Japan,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Japan
| | | | - Thomas S Kilduff
- Corresponding author. Thomas S. Kilduff, Center for Neuroscience, Biosciences Division SRI International, 333 Ravenswood Ave Menlo Park, CA 94025 USA.
| |
Collapse
|
14
|
Plancoulaine S, Guyon A, Inocente CO, Germe P, Zhang M, Robert P, Lin JS, Franco P. Cerebrospinal Fluid Histamine Levels in Healthy Children and Potential Implication for SIDS: Observational Study in a French Tertiary Care Hospital. Front Pediatr 2022; 10:819496. [PMID: 35450108 PMCID: PMC9016218 DOI: 10.3389/fped.2022.819496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 03/09/2022] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE A defect of the waking systems could constitute a factor of vulnerability for sudden infant death syndrome (SIDS). A decrease in orexin levels, which promotes wakefulness and activates histaminergic neurons (another hypothalamic wake-promoting system) has already been demonstrated between 2 and 6 months. This work aims to study the levels of histamine (HA), tele-methylhistamine (t-MeHA), its direct metabolite, and t-MeHA/HA ratio in the cerebrospinal fluid (CSF) of healthy children, to evaluate the maturation of the histaminergic system and its possible involvement in SIDS. METHODS Seventy Eight French children between 0 and 20 years (48.7% boys) were included, all of whom had a clinical indication for lumbar puncture, but subsequently found to be normal. Measurements of HA and t-MeHA in CSF were performed by reverse phase liquid chromatography coupled to mass spectrometry detection. Statistical analyses were performed using Spearman correlations and Non-parametric pairwise ranking tests. RESULTS A negative correlation was found between age and CSF HA (r = -0.44, p < 10-4) and t-MeHA (r = -0.70, p < 10-4) levels. In pairwise comparisons, no difference in CSF HA and t-MeHA levels was observed between youngest age groups (i.e., 0-2 mo vs. 3-6 mo), but CSF HA and t-MeHA levels were significantly lower in older children (i.e., >6 mo vs. 0-6 mo). The CSF HA decrease with age was only observed in boys, who also presented global lower CSF HA levels than girls. CONCLUSION CSF HA and t-MeHA levels decrease with age in boys, and global levels are lower in boys than in girls. These results reveal changes in histaminergic transmission and metabolism during maturation. Whether lower CSF histamine values in boys compared to girls could contribute to their higher risk of SIDS warrants further research.
Collapse
Affiliation(s)
| | - Aurore Guyon
- Integrative Physiology of the Brain Arousal System, CRNL, INSERM-U1028, CNRS UMR5292, University Lyon 1, Lyon, France.,Bioprojet Biotech, Saint-Grégoire, France
| | - Clara-Odilia Inocente
- Integrative Physiology of the Brain Arousal System, CRNL, INSERM-U1028, CNRS UMR5292, University Lyon 1, Lyon, France
| | - Philippine Germe
- Integrative Physiology of the Brain Arousal System, CRNL, INSERM-U1028, CNRS UMR5292, University Lyon 1, Lyon, France
| | - Min Zhang
- Integrative Physiology of the Brain Arousal System, CRNL, INSERM-U1028, CNRS UMR5292, University Lyon 1, Lyon, France
| | | | - Jian-Sheng Lin
- Integrative Physiology of the Brain Arousal System, CRNL, INSERM-U1028, CNRS UMR5292, University Lyon 1, Lyon, France
| | - Patricia Franco
- Integrative Physiology of the Brain Arousal System, CRNL, INSERM-U1028, CNRS UMR5292, University Lyon 1, Lyon, France.,Pediatric Sleep Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, University Lyon 1, Lyon, France
| |
Collapse
|
15
|
Arrigoni E, Fuller PM. The Role of the Central Histaminergic System in Behavioral State Control. Curr Top Behav Neurosci 2022; 59:447-468. [PMID: 34595740 DOI: 10.1007/7854_2021_263] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Histamine is a small monoamine signaling molecule that plays a role in many peripheral and central physiological processes, including the regulation of wakefulness. The tuberomammillary nucleus is the sole neuronal source of histamine in the brain, and histamine neurons are thought to promote wakefulness and vigilance maintenance - under certain environmental and/or behavioral contexts - through their diffuse innervation of the cortex and other wake-promoting brain circuits. Histamine neurons also contain a number of other putative neurotransmitters, although the functional role of these co-transmitters remains incompletely understood. Within the brain histamine operates through three receptor subtypes that are located on pre- and post-synaptic membranes. Some histamine receptors exhibit constitutive activity, and hence exist in an activated state even in the absence of histamine. Newer medications used to reduce sleepiness in narcolepsy patients in fact enhance histamine signaling by blunting the constitutive activity of these histamine receptors. In this chapter, we provide an overview of the central histamine system with an emphasis on its role in behavioral state regulation and how drugs targeting histamine receptors are used clinically to treat a wide range of sleep-wake disorders.
Collapse
Affiliation(s)
- Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Patrick M Fuller
- Department of Neurological Surgery, University of California Davis School of Medicine, Davis, CA, USA
| |
Collapse
|
16
|
Shan L, Martens GJM, Swaab DF. Histamine-4 Receptor: Emerging Target for the Treatment of Neurological Diseases. Curr Top Behav Neurosci 2021; 59:131-145. [PMID: 34432256 DOI: 10.1007/7854_2021_237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
A major challenge in the field of the biogenic amine histamine is the search for new-generation histamine receptor specific drugs. Daniel Bovet and Sir James Black received their Nobel Prizes for Medicine for their work on histamine-1 receptor (H1R) and H2R antagonists to treat allergies and gastrointestinal disorders. The first H3R-targeting drug to reach the market was approved for the treatment of the neurological disorder narcolepsy in 2018. The antagonists for the most recently identified histamine receptor, H4R, are currently under clinical evaluation for their potential therapeutic effects on inflammatory diseases such as atopic dermatitis and pruritus. In this chapter, we propose that H4R antagonists are endowed with prominent anti-inflammatory and immune effects, including in the brain. To substantiate this proposition, we combine data from transcriptional analyses of postmortem human neurodegenerative disease brain samples, human genome-wide association studies (GWAS), and translational animal model studies. The results prompt us to suggest the potential involvement of the H4R in various neurodegenerative diseases and how manipulating the H4R may create new therapeutic opportunities in central nervous system diseases.
Collapse
Affiliation(s)
- Ling Shan
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands.
| | - Gerard J M Martens
- Department of Molecular Animal Physiology, Faculty of Science, Donders Institute for Brain, Cognition and Behaviour, Donders Centre for Neuroscience, Nijmegen, GA, The Netherlands
| | - Dick F Swaab
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| |
Collapse
|
17
|
A Duet Between Histamine and Oleoylethanolamide in the Control of Homeostatic and Cognitive Processes. Curr Top Behav Neurosci 2021; 59:389-410. [PMID: 34410679 DOI: 10.1007/7854_2021_236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In ballet, a pas de deux (in French it means "step of two") is a duet in which the two dancers perform ballet steps together. The suite of dances shares a common theme of partnership. How could we better describe the fine interplay between oleoylethanolamide (OEA) and histamine, two phylogenetically ancient molecules controlling metabolic, homeostatic and cognitive processes? Contrary to the pas de deux though, the two dancers presumably never embrace each other as a dancing pair but execute their "virtuoso solo" constantly exchanging interoceptive messages presumably via vagal afferents, the blood stream, the neuroenteric system. With one exception, which is in the control of liver ketogenesis, as in hepatocytes, OEA biosynthesis strictly depends on the activation of histaminergic H1 receptors. In this review, we recapitulate our main findings that evidence the interplay of histamine and OEA in the control of food consumption and eating behaviour, in the consolidation of emotional memory and mood, and finally, in the synthesis of ketone bodies. We will also summarise some of the putative underlying mechanisms for each scenario.
Collapse
|
18
|
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
|
19
|
Davis CW, Kallweit U, Schwartz JC, Krahn LE, Vaughn B, Thorpy MJ. Efficacy of pitolisant in patients with high burden of narcolepsy symptoms: pooled analysis of short-term, placebo-controlled studies. Sleep Med 2021; 81:210-217. [PMID: 33721598 DOI: 10.1016/j.sleep.2021.02.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/01/2021] [Accepted: 02/16/2021] [Indexed: 01/12/2023]
Abstract
STUDY OBJECTIVE To evaluate the efficacy of pitolisant, a histamine 3 (H3)-receptor antagonist/inverse agonist, in adult patients with high burden of narcolepsy symptoms. METHODS Data were pooled from two randomized, placebo-controlled, 7- or 8-week studies of pitolisant (titrated to a potential maximum dose of 35.6 mg/day) in adults with narcolepsy. Analyses included three independent patient subgroups: Epworth Sleepiness Scale (ESS) baseline score ≥16, Maintenance of Wakefulness Test (MWT) sleep latency ≤8 min, and ≥15 cataplexy attacks per week. RESULTS The analysis populations included 118 patients for ESS (pitolisant, n = 60; placebo, n = 58), 105 for MWT (pitolisant, n = 59; placebo, n = 46), and 31 for cataplexy (pitolisant, n = 20; placebo, n = 11). On the ESS, least-squares mean change from baseline was significantly greater for pitolisant (-6.1) compared with placebo (-2.3; P < 0.001). Significantly more pitolisant-treated patients were classified as treatment responders: ESS score reduction ≥3, 69.0% in the pitolisant group versus 35.1% in the placebo group (P = 0.001); final ESS score ≤10, 36.2% versus 10.5%, respectively (P = 0.005). On the MWT, mean sleep latency increased from 3.5 min to 10.4 min with pitolisant and from 3.4 min to 6.8 min with placebo (P = 0.017). Least-squares mean change in the weekly rate of cataplexy was significantly greater for pitolisant (-14.5; baseline, 23.9; final, 9.4) compared with placebo (-0.1; baseline, 23.1; final, 23.0; P = 0.004). Headache was the most common adverse event with pitolisant. CONCLUSIONS Pitolisant, at once-daily doses up to 35.6 mg, was efficacious for reducing excessive daytime sleepiness and cataplexy in patients with severe narcolepsy symptom burden.
Collapse
Affiliation(s)
- Craig W Davis
- Harmony Biosciences, LLC, Plymouth Meeting, PA, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Zhang M, Inocente CO, Villanueva C, Lecendreux M, Dauvilliers Y, Lin JS, Arnulf I, Gustin MP, Thieux M, Franco P. Narcolepsy with cataplexy: Does age at diagnosis change the clinical picture? CNS Neurosci Ther 2020; 26:1092-1102. [PMID: 32761857 PMCID: PMC7539846 DOI: 10.1111/cns.13438] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/06/2020] [Accepted: 06/15/2020] [Indexed: 11/29/2022] Open
Abstract
Objective To compare symptoms and sleep characteristics in patients diagnosed with narcolepsy‐cataplexy (NC) before and after the age of 18 years. Methods De novo patients with NC diagnosis completed a standardized questionnaire and interview, followed by a sleep study. The clinical and sleep measures were compared between patients diagnosed before (46 children, median age: 12 year old) and after (46 adults, median age: 28.5 year old) 18 years of age. Results The frequency of obesity (54% vs 17%), night eating (29% vs 7%), parasomnia (89% vs 43%), sleep talking (80% vs 34%), and sleep drunkenness (69% vs 24%) were higher in children than in adults, the frequency of sleep paralysis was lower (20% vs 55%) but the frequency of cataplexy and the severity of sleepiness were not different. Children scored higher than adults at the attention‐deficit/hyperactivity disorder (ADHD) scale. Depressive feelings affected not differently children (24%) and adults (32%). However, adults had lower quality of life than children. There was no difference between groups for insomnia and fatigue scores. Quality of life was essentially impacted by depressive feelings in both children and adults. Obstructive apnea‐hypopnea index (OAHI) was lower in children with higher mean and minimal oxygen saturation than in adults. No between‐group differences were found at the multiple sleep latency test. The body mass index (z‐score) was correlated with OAHI (r = .32). Conclusion At time of NC diagnosis, children have more frequent obesity, night eating, parasomnia, sleep talking, drunkenness, and ADHD symptoms than adults, even if sleepiness and cataplexy do not differ. These differences should be considered to ensure a prompt diagnosis.
Collapse
Affiliation(s)
- Min Zhang
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM-U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
| | - Clara Odilia Inocente
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM-U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
| | - Carine Villanueva
- Endocrinology Pediatric Unit, Woman Mother Child Hospital, Civil Hospices of Lyon, Lyon, France
| | - Michel Lecendreux
- Pediatric Sleep Centre, Hospital Robert-Debre, Paris, France.,National Reference Centre for Orphan Diseases, Narcolepsy, Idiopathic Hypersomnia, and Kleine-Levin Syndrome, Paris, France
| | - Yves Dauvilliers
- National Reference Network for Narcolepsy, Sleep-Wake Disorder Unit, Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier, Montpellier, France.,Inserm U1061, University of Montpellier, Neuropsychiatry: Epidemiological and Clinical Research, Montpellier, France
| | - Jian-Sheng Lin
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM-U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
| | - Isabelle Arnulf
- AP-HP, Pitié-Salpêtrière Hospital, Sleep Disorder Unit & Sorbonne University, Paris, France
| | - Marie-Paule Gustin
- Emerging Pathogens Laboratory-Fondation Mérieux, International Center for Infectiology Research (CIRI), Inserm U1111, CNRS UMR5308, ENS de Lyon, Lyon, France.,Institute of Pharmaceutic and Biological Sciences, Public Health Department, Biostatistics, University Claude Bernard Lyon 1, Villeurbanne, France
| | - Marine Thieux
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM-U1028, CNRS UMR5292, University of Lyon 1, Lyon, France.,Sleep Pediatric Unit, Woman Mother Child Hospital, Civil Hospices of Lyon, Lyon, France
| | - Patricia Franco
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM-U1028, CNRS UMR5292, University of Lyon 1, Lyon, France.,Sleep Pediatric Unit, Woman Mother Child Hospital, Civil Hospices of Lyon, Lyon, France
| |
Collapse
|
21
|
Hill E, Dale N, Wall MJ. Moderate Changes in CO 2 Modulate the Firing of Neurons in the VTA and Substantia Nigra. iScience 2020; 23:101343. [PMID: 32683315 PMCID: PMC7371905 DOI: 10.1016/j.isci.2020.101343] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/30/2020] [Accepted: 07/01/2020] [Indexed: 12/21/2022] Open
Abstract
The substantia nigra (SN) and ventral tegmental area (VTA) are vital for the control of movement, goal-directed behavior, and encoding reward. Here we show that the firing of specific neuronal subtypes in these nuclei can be modulated by physiological changes in the partial pressure of carbon dioxide (PCO2). The resting conductance of substantia nigra dopaminergic neurons in young animals (postnatal days 7-10) and GABAergic neurons in the VTA is modulated by changes in the level of CO2. We provide several lines of evidence that this CO2-sensitive conductance results from connexin 26 (Cx26) hemichannel expression. Since the levels of PCO2 in the blood will vary depending on physiological activity and pathology, this suggests that changes in PCO2 could potentially modulate motor activity, reward behavior, and wakefulness.
Collapse
Affiliation(s)
- Emily Hill
- School of Life Sciences, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK.
| | - Nicholas Dale
- School of Life Sciences, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK
| | - Mark J Wall
- School of Life Sciences, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK.
| |
Collapse
|
22
|
Exarchos I, Rogers AA, Aiani LM, Gross RE, Clifford GD, Pedersen NP, Willie JT. Supervised and unsupervised machine learning for automated scoring of sleep-wake and cataplexy in a mouse model of narcolepsy. Sleep 2020; 43:zsz272. [PMID: 31693157 PMCID: PMC7215268 DOI: 10.1093/sleep/zsz272] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/09/2019] [Indexed: 11/13/2022] Open
Abstract
Despite commercial availability of software to facilitate sleep-wake scoring of electroencephalography (EEG) and electromyography (EMG) in animals, automated scoring of rodent models of abnormal sleep, such as narcolepsy with cataplexy, has remained elusive. We optimize two machine-learning approaches, supervised and unsupervised, for automated scoring of behavioral states in orexin/ataxin-3 transgenic mice, a validated model of narcolepsy type 1, and additionally test them on wild-type mice. The supervised learning approach uses previously labeled data to facilitate training of a classifier for sleep states, whereas the unsupervised approach aims to discover latent structure and similarities in unlabeled data from which sleep stages are inferred. For the supervised approach, we employ a deep convolutional neural network architecture that is trained on expert-labeled segments of wake, non-REM sleep, and REM sleep in EEG/EMG time series data. The resulting trained classifier is then used to infer on the labels of previously unseen data. For the unsupervised approach, we leverage data dimensionality reduction and clustering techniques. Both approaches successfully score EEG/EMG data, achieving mean accuracies of 95% and 91%, respectively, in narcoleptic mice, and accuracies of 93% and 89%, respectively, in wild-type mice. Notably, the supervised approach generalized well on previously unseen data from the same animals on which it was trained but exhibited lower performance on animals not present in the training data due to inter-subject variability. Cataplexy is scored with a sensitivity of 85% and 57% using the supervised and unsupervised approaches, respectively, when compared to manual scoring, and the specificity exceeds 99% in both cases.
Collapse
Affiliation(s)
- Ioannis Exarchos
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA
| | - Anna A Rogers
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA
| | - Lauren M Aiani
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA
| | - Robert E Gross
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA
- Program in Neuroscience, Emory University, Atlanta, GA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - Gari D Clifford
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
| | - Nigel P Pedersen
- Department of Neurology, Emory University School of Medicine, Atlanta, GA
- Program in Neuroscience, Emory University, Atlanta, GA
| | - Jon T Willie
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA
- Program in Neuroscience, Emory University, Atlanta, GA
| |
Collapse
|
23
|
LC-MS/MS method for the quantification of SUVN-G3031, a novel H3 receptor inverse agonist for narcolepsy treatment. Bioanalysis 2020; 12:533-544. [PMID: 32351118 DOI: 10.4155/bio-2020-0020] [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] [Indexed: 12/12/2022] Open
Abstract
Background: A LC-MS/MS method was validated for the quantification of SUVN-G3031, a novel H3 receptor inverse agonist in clinical development for the treatment of patients with narcolepsy, with and without cataplexy. Methodology: SUVN-G3031 was extracted from plasma following acetonitrile protein precipitation, separated by Ultra HPLC and quantified using positive ESI-MS/MS. Results: The method was linear across the range of 0.1-100 ng ml-1 in plasma. Results for intra and inter-day accuracy were from 99.8 to 104% and precision (%CV) was ≤10.6%. Conclusion: The method was applied to a first-in-human study in healthy volunteers. The method is precise, accurate and highly selective for the quantification of SUVN-G3031 in human plasma.
Collapse
|
24
|
Vanda D, Zajdel P, Soural M. Imidazopyridine-based selective and multifunctional ligands of biological targets associated with psychiatric and neurodegenerative diseases. Eur J Med Chem 2019; 181:111569. [DOI: 10.1016/j.ejmech.2019.111569] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/26/2019] [Accepted: 07/28/2019] [Indexed: 12/18/2022]
|
25
|
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
|
26
|
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: 29] [Impact Index Per Article: 4.8] [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
|
27
|
Reassessing the Role of Histaminergic Tuberomammillary Neurons in Arousal Control. J Neurosci 2019; 39:8929-8939. [PMID: 31548232 DOI: 10.1523/jneurosci.1032-19.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 09/16/2019] [Accepted: 09/19/2019] [Indexed: 02/03/2023] Open
Abstract
The histaminergic neurons of the tuberomammillary nucleus (TMNHDC) of the posterior hypothalamus have long been implicated in promoting arousal. More recently, a role for GABAergic signaling by the TMNHDC neurons in arousal control has been proposed. Here, we investigated the effects of selective chronic disruption of GABA synthesis (via genetic deletion of the GABA synthesis enzyme, glutamic acid decarboxylase 67) or GABAergic transmission (via genetic deletion of the vesicular GABA transporter (VGAT)) in the TMNHDC neurons on sleep-wake in male mice. We also examined the effects of acute chemogenetic activation and optogenetic inhibition of TMNHDC neurons upon arousal in male mice. Unexpectedly, we found that neither disruption of GABA synthesis nor GABAergic transmission altered hourly sleep-wake quantities, perhaps because very few TMNHDC neurons coexpressed VGAT. Acute chemogenetic activation of TMNHDC neurons did not increase arousal levels above baseline but did enhance vigilance when the mice were exposed to a behavioral cage change challenge. Similarly, acute optogenetic inhibition had little effect upon baseline levels of arousal. In conclusion, we could not identify a role for GABA release by TMNHDC neurons in arousal control. Further, if TMNHDC neurons do release GABA, the mechanism by which they do so remains unclear. Our findings support the view that TMNHDC neurons may be important for enhancing arousal under certain conditions, such as exposure to a novel environment, but play only a minor role in behavioral and EEG arousal under baseline conditions.SIGNIFICANCE STATEMENT The histaminergic neurons of the tuberomammillary nucleus of the hypothalamus (TMNHDC) have long been thought to promote arousal. Additionally, TMNHDC neurons may counter-regulate the wake-promoting effects of histamine through co-release of the inhibitory neurotransmitter, GABA. Here, we show that impairing GABA signaling from TMNHDC neurons does not impact sleep-wake amounts and that few TMNHDC neurons contain the vesicular GABA transporter, which is presumably required to release GABA. We further show that acute activation or inhibition of TMNHDC neurons has limited effects upon baseline arousal levels and that activation enhances vigilance during a behavioral challenge. Counter to general belief, our findings support the view that TMNHDC neurons are neither necessary nor sufficient for the initiation and maintenance of arousal under baseline conditions.
Collapse
|
28
|
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
|
29
|
Thorpy MJ, Bogan RK. Update on the pharmacologic management of narcolepsy: mechanisms of action and clinical implications. Sleep Med 2019; 68:97-109. [PMID: 32032921 DOI: 10.1016/j.sleep.2019.09.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/21/2022]
Abstract
Narcolepsy is a chronic, debilitating neurological disorder of sleep-wake state instability. This instability underlies all narcolepsy symptoms, including excessive daytime sleepiness (EDS), symptoms of rapid eye movement (REM) sleep dysregulation (ie, cataplexy, hypnagogic/hypnopompic hallucinations, sleep paralysis), and disrupted nighttime sleep. Several neurotransmitter systems promote wakefulness, and various neural pathways are involved in regulating REM sleep-related muscle atonia, providing multiple targets for pharmacologic intervention to reduce EDS and cataplexy. Medications approved by the US Food and Drug Administration (FDA) for the treatment of EDS in narcolepsy include traditional stimulants (eg, amphetamines, methylphenidate), wake-promoting agents (eg, modafinil, armodafinil), and solriamfetol, which mainly act on dopaminergic and noradrenergic pathways. Sodium oxybate (thought to act via GABAB receptors) is FDA-approved for the treatment of EDS and cataplexy. Pitolisant, a histamine 3 (H3)-receptor antagonist/inverse agonist, is approved by the European Medicines Agency (EMA) for the treatment of narcolepsy with or without cataplexy in adults and by the FDA for the treatment of EDS in adults with narcolepsy. Pitolisant increases the synthesis and release of histamine in the brain and modulates the release of other neurotransmitters (eg, norepinephrine, dopamine). Antidepressants that inhibit reuptake of serotonin and/or norepinephrine are widely used off label to manage cataplexy. In many patients with narcolepsy, combination treatment with medications that act via different neural pathways is necessary for optimal symptom management. Mechanism of action, pharmacokinetics, and abuse potential are important considerations in treatment selection and subsequent medication adjustments to maximize efficacy and mitigate adverse effects in the treatment of patients with narcolepsy.
Collapse
Affiliation(s)
- Michael J Thorpy
- Sleep-Wake Disorders Center, Montefiore Medical Center, Albert Einstein College of Medicine, 3411 Wayne Ave, Bronx, NY, 10467, USA.
| | - Richard K Bogan
- SleepMed Inc., Bogan Sleep Consultants, LLC, 1333 Taylor Street, Columbia, SC, USA.
| |
Collapse
|
30
|
Huang B, Qian Z, Wang Z, Zhang J, Chen K, Xu T, Wang J, Cechetto DF, Zhao Z, Wu H. Fluctuation of primary motor cortex excitability during cataplexy in narcolepsy. Ann Clin Transl Neurol 2019; 6:210-221. [PMID: 30847354 PMCID: PMC6389735 DOI: 10.1002/acn3.670] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/17/2018] [Indexed: 12/13/2022] Open
Abstract
Objective Cataplexy is a complicated and dynamic process in narcolepsy type 1 (NT1) patients. This study aimed to clarify the distinct stages during a cataplectic attack and identify the changes of the primary motor cortex (PMC) excitability during these stages. Methods Thirty-five patients with NT1 and 29 healthy controls were recruited to this study. Cataplectic stages were distinguished from a cataplectic attack by video-polysomnogram monitoring. Transcranial magnetic stimulation motor-evoked potential (TMS-MEP) was performed to measure the excitability of PMC during quiet wakefulness, laughter without cataplexy, and each cataplectic stage. Results Based on the video and electromyogram observations, a typical cataplectic attack (CA) process is divided into four stages: triggering (CA1), resisting (CA2), atonic (CA3), and recovering stages (CA4). Compared with healthy controls, NT1 patients showed significantly decreased intracortical facilitation during quiet wakefulness. During the laughter stage, both patients and controls showed increased MEP amplitude compared with quiet wakefulness. The MEP amplitude significantly increased even higher in CA1 and 2, and then dramatically decreased in CA3 accompanied with prolonged MEP latency compared with the laughter stage and quiet wakefulness. The MEP amplitude and latency gradually recovered during CA4. Interpretation This study identifies four stages during cataplectic attack and reveals the existence of a resisting stage that might change the process of cataplexy. The fluctuation of MEP amplitude and MEP latency shows a potential participation of PMC and motor control pathway during cataplexy, and the increased MEP amplitude during CA1 and 2 strongly implies a compensatory mechanism in motor control that may resist or avoid cataplectic attack.
Collapse
Affiliation(s)
- Bei Huang
- Department of Neurology Changzheng Hospital The Second Military Medical University Shanghai China.,Department of Psychiatry Faculty of Medicine The Chinese University of Hong Kong Shatin Hong Kong
| | - Zhenying Qian
- Department of EEG Source Imaging Shanghai Mental Health Center Shanghai China
| | - Zongwen Wang
- Department of Neurology Changzheng Hospital The Second Military Medical University Shanghai China
| | - Jihui Zhang
- Department of Psychiatry Faculty of Medicine The Chinese University of Hong Kong Shatin Hong Kong
| | - Kun Chen
- Department of Neurology Changzheng Hospital The Second Military Medical University Shanghai China
| | - Tao Xu
- Department of Neurology Changzheng Hospital The Second Military Medical University Shanghai China
| | - Jijun Wang
- Department of EEG Source Imaging Shanghai Mental Health Center Shanghai China
| | - David F Cechetto
- Department of Anatomy & Cell Biology University of Western Ontario London Ontario Canada
| | - Zhongxin Zhao
- Department of Neurology Changzheng Hospital The Second Military Medical University Shanghai China
| | - Huijuan Wu
- Department of Neurology Changzheng Hospital The Second Military Medical University Shanghai China
| |
Collapse
|
31
|
Thomasy HE, Opp MR. Hypocretin Mediates Sleep and Wake Disturbances in a Mouse Model of Traumatic Brain Injury. J Neurotrauma 2019; 36:802-814. [PMID: 30136622 PMCID: PMC6387567 DOI: 10.1089/neu.2018.5810] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Traumatic brain injury (TBI) is a major cause of disability worldwide. Post-TBI sleep and wake disturbances are extremely common and difficult for patients to manage. Sleep and wake disturbances contribute to poor functional and emotional outcomes from TBI, yet effective therapies remain elusive. A more comprehensive understanding of mechanisms underlying post-TBI sleep and wake disturbance will facilitate development of effective pharmacotherapies. Previous research in human patients and animal models indicates that altered hypocretinergic function may be a major contributor to sleep-wake disturbance after TBI. In this study, we further elucidate the role of hypocretin by determining the impact of TBI on sleep-wake behavior of hypocretin knockout (HCRT KO) mice. Adult male C57BL/6J and HCRT KO mice were implanted with electroencephalography recording electrodes, and pre-injury baseline recordings were obtained. Mice were then subjected to either moderate TBI or sham surgery. Additional recordings were obtained and sleep-wake behavior determined at 3, 7, 15, and 30 days after TBI or sham procedures. At baseline, HCRT KO mice had a significantly different sleep-wake phenotype than control C57BL/6J mice. Post-TBI sleep-wake behavior was altered in a genotype-dependent manner: sleep of HCRT KO mice was not altered by TBI, whereas C57BL/6J mice had more non-rapid eye movement sleep, less wakefulness, and more short wake bouts and fewer long wake bouts. Numbers of hypocretin-positive cells were reduced in C57BL/6J mice by TBI. Collectively, these data indicate that the hypocretinergic system is involved in the alterations in sleep-wake behavior that develop after TBI in this model, and suggest potential therapeutic interventions.
Collapse
Affiliation(s)
- Hannah E. Thomasy
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | - Mark R. Opp
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, Washington
| |
Collapse
|
32
|
González B, Torres OV, Jayanthi S, Gomez N, Sosa MH, Bernardi A, Urbano FJ, García-Rill E, Cadet JL, Bisagno V. The effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors. Prog Neuropsychopharmacol Biol Psychiatry 2019; 88:222-234. [PMID: 30056065 PMCID: PMC8424782 DOI: 10.1016/j.pnpbp.2018.07.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/12/2018] [Accepted: 07/23/2018] [Indexed: 01/02/2023]
Abstract
METH use causes neuroadaptations that negatively impact the prefrontal cortex (PFC) leading to addiction and associated cognitive decline in animals and humans. In contrast, modafinil enhances cognition by increasing PFC function. Accumulated evidence indicates that psychostimulant drugs, including modafinil and METH, regulate gene expression via epigenetic modifications. In this study, we measured the effects of single-dose injections of modafinil and METH on the protein levels of acetylated histone H3 (H3ac) and H4ac, deacetylases HDAC1 and HDAC2, and of the NMDA subunit GluN1 in the medial PFC (mPFC) of mice euthanized 1 h after drug administration. To test if dopamine (DA) receptors (DRs) participate in the biochemical effects of the two drugs, we injected the D1Rs antagonist, SCH23390, or the D2Rs antagonist, raclopride, 30 min before administration of METH and modafinil. We evaluated each drug effect on glutamate synaptic transmission in D1R-expressing layer V pyramidal neurons. We also measured the enrichment of H3ac and H4ac at the promoters of several genes including DA, NE, orexin, histamine, and glutamate receptors, and their mRNA expression, since they are responsive to chronic modafinil and METH treatment. Acute modafinil and METH injections caused similar effects on total histone acetylation, increasing H3ac and decreasing H4ac, and they also increased HDAC1, HDAC2 and GluN1 protein levels in the mouse mPFC. In addition, the effects of the drugs were prevented by pre-treatment with D1Rs and D2Rs antagonists. Specifically, the changes in H4ac, HDAC2, and GluN1 were responsive to SCH23390, whereas those of H3ac and GluN1 were responsive to raclopride. Whole-cell patch clamp in transgenic BAC-Drd1a-tdTomato mice showed that METH, but not modafinil, induced paired-pulse facilitation of EPSCs, suggesting reduced presynaptic probability of glutamate release onto layer V pyramidal neurons. Analysis of histone 3/4 enrichment at specific promoters revealed: i) distinct effects of the drugs on histone 3 acetylation, with modafinil increasing H3ac at Drd1 and Adra1b promoters, but METH increasing H3ac at Adra1a; ii) distinct effects on histone 4 acetylation enrichment, with modafinil increasing H4ac at the Drd2 promoter and decreasing it at Hrh1, but METH increasing H4ac at Drd1; iii) comparable effects of both psychostimulants, increasing H3ac at Drd2, Hcrtr1, and Hrh1 promoters, decreasing H3ac at Hrh3, increasing H4ac at Hcrtr1, and decreasing H4ac at Hcrtr2, Hrh3, and Grin1 promoters. Interestingly, only METH altered mRNA levels of genes with altered histone acetylation status, inducing increased expression of Drd1a, Adra1a, Hcrtr1, and Hrh1, and decreasing Grin1. Our study suggests that although acute METH and modafinil can both increase DA neurotransmission in the mPFC, there are similar and contrasting epigenetic and transcriptional consequences that may account for their divergent clinical effects.
Collapse
Affiliation(s)
- Betina González
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Oscar V Torres
- Department of Behavioral Sciences, San Diego Mesa College, San Diego, California, United States
| | - Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, United States
| | - Natalia Gomez
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Máximo H Sosa
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Alejandra Bernardi
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Francisco J Urbano
- Laboratorio de Fisiología y Biología Molecular, Instituto de Fisiología, Biología Molecular y Neurociencias (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Edgar García-Rill
- Center for Translational Neuroscience, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jean-Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, United States
| | - Verónica Bisagno
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
33
|
Abstract
A wide variety of neuropathological abnormalities have been investigated in infants who have died of sudden infant death syndrome (SIDS). Issues which detracted from early studies included failure to use uniform definitions of SIDS and lack of appropriately matched control populations. Development of the triple risk model focused attention on the concept of an inherent susceptibility to unexpected death in certain infants, with research demonstrating a role for the neurotransmitter serotonin within the brainstem. However, it now appears that neuropathological abnormalities in SIDS infants are more complex than a simple serotonergic deficiency in certain medullary nuclei but instead could involve failure of an integrated network of neurochemical transmitters in a variety of subcortical locations. The following overview examines recent research developments looking particularly at the potential role of the peptide neurotransmitter substance P and its neurokinin-1 receptor in multiple nuclei within the brainstem, asymmetry and microdysgenesis of the hippocampus, and decreased orexin levels within dorsomedial, perifornical, and lateral levels in the hypothalamus. Whether such research will lead to identifiable biomarker for infants at risk of SIDS is yet to be established. Use of standardized and consistent methods of classifying and categorizing infant deaths will be pivotal in generating reproducible research results.
Collapse
Affiliation(s)
- Fiona M Bright
- 1 School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Robert Vink
- 2 Sansom Institute for Health Research, University of South Australia, Adelaide, South Australia, Australia
| | - Roger W Byard
- 1 School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
34
|
Provensi G, Costa A, Izquierdo I, Blandina P, Passani MB. Brain histamine modulates recognition memory: possible implications in major cognitive disorders. Br J Pharmacol 2018; 177:539-556. [PMID: 30129226 DOI: 10.1111/bph.14478] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/27/2018] [Accepted: 08/05/2018] [Indexed: 12/24/2022] Open
Abstract
Several behavioural tests have been developed to study and measure emotionally charged or emotionally neutral memories and how these may be affected by pharmacological, dietary or environmental manipulations. In this review, we describe the experimental paradigms used in preclinical studies to unravel the brain circuits involved in the recognition and memorization of environmentally salient stimuli devoid of strong emotional value. In particular, we focus on the modulatory role of the brain histaminergic system in the elaboration of recognition memory that is based on the judgement of the prior occurrence of an event, and it is believed to be a critical component of human declarative memory. The review also addresses questions that may help improve the treatment of impaired declarative memory described in several affective and neuropsychiatric disorders such as ADHD, Alzheimer's disease and major neurocognitive disorder. LINKED ARTICLES: This article is part of a themed section on New Uses for 21st Century. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.3/issuetoc.
Collapse
Affiliation(s)
- Gustavo Provensi
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Alessia Costa
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Ivan Izquierdo
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Patrizio Blandina
- Department of Neuroscience, Psychology, Drug Research and Child Health, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Maria Beatrice Passani
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
35
|
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
|
36
|
Sabetghadam A, Grabowiecka-Nowak A, Kania A, Gugula A, Blasiak E, Blasiak T, Ma S, Gundlach AL, Blasiak A. Melanin-concentrating hormone and orexin systems in rat nucleus incertus: Dual innervation, bidirectional effects on neuron activity, and differential influences on arousal and feeding. Neuropharmacology 2018; 139:238-256. [DOI: 10.1016/j.neuropharm.2018.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/20/2018] [Accepted: 07/04/2018] [Indexed: 12/24/2022]
|
37
|
Eban-Rothschild A, Appelbaum L, de Lecea L. Neuronal Mechanisms for Sleep/Wake Regulation and Modulatory Drive. Neuropsychopharmacology 2018; 43:937-952. [PMID: 29206811 PMCID: PMC5854814 DOI: 10.1038/npp.2017.294] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 11/17/2017] [Accepted: 11/24/2017] [Indexed: 12/17/2022]
Abstract
Humans have been fascinated by sleep for millennia. After almost a century of scientific interrogation, significant progress has been made in understanding the neuronal regulation and functions of sleep. The application of new methods in neuroscience that enable the analysis of genetically defined neuronal circuits with unprecedented specificity and precision has been paramount in this endeavor. In this review, we first discuss electrophysiological and behavioral features of sleep/wake states and the principal neuronal populations involved in their regulation. Next, we describe the main modulatory drives of sleep and wakefulness, including homeostatic, circadian, and motivational processes. Finally, we describe a revised integrative model for sleep/wake regulation.
Collapse
Affiliation(s)
| | - Lior Appelbaum
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| |
Collapse
|
38
|
Roman A, Meftah S, Arthaud S, Luppi PH, Peyron C. The inappropriate occurrence of rapid eye movement sleep in narcolepsy is not due to a defect in homeostatic regulation of rapid eye movement sleep. Sleep 2018. [DOI: 10.1093/sleep/zsy046] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Alexis Roman
- Center for Research in Neuroscience of Lyon (CRNL), SLEEP team, CNRS UMR5292, INSERM U1028, Université Claude Bernard-Lyon1, Lyon, France
| | - Soraya Meftah
- Center for Research in Neuroscience of Lyon (CRNL), SLEEP team, CNRS UMR5292, INSERM U1028, Université Claude Bernard-Lyon1, Lyon, France
| | - Sébastien Arthaud
- Center for Research in Neuroscience of Lyon (CRNL), SLEEP team, CNRS UMR5292, INSERM U1028, Université Claude Bernard-Lyon1, Lyon, France
| | - Pierre-Hervé Luppi
- Center for Research in Neuroscience of Lyon (CRNL), SLEEP team, CNRS UMR5292, INSERM U1028, Université Claude Bernard-Lyon1, Lyon, France
| | - Christelle Peyron
- Center for Research in Neuroscience of Lyon (CRNL), SLEEP team, CNRS UMR5292, INSERM U1028, Université Claude Bernard-Lyon1, Lyon, France
| |
Collapse
|
39
|
Holland PR. Biology of Neuropeptides: Orexinergic Involvement in Primary Headache Disorders. Headache 2018; 57 Suppl 2:76-88. [PMID: 28485849 DOI: 10.1111/head.13078] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 03/08/2017] [Indexed: 01/01/2023]
Abstract
Migraine is a very common, severe disabling condition that can last for days and strike multiple times per month. Attacks, often characterized by severe unilateral throbbing pain that is exacerbated by activity, are commonly preceded by several diverse symptoms including fatigue, irritability, and yawning. This premonitory (prodromal) phase represents the earliest identifiable feature of an attack that is a reliable predictor of ensuing headache. The diversity of these symptoms underlines the complex nature of migraine and focuses considerable attention on the hypothalamus due to its prominent role in homeostatic regulation allowing state dependent behavioral modifications. While multiple neurotransmitter and neuropeptide systems have been proposed to play a role in migraine, the current review will focus on the emerging role of the hypothalamic orexinergic system in primary headache disorders. Specifically the potential role of altered orexinergic signalling in premonitory symptomatology and the future potential of targeted orexinergic therapies that could with other approaches act during the premonitory phase to prevent the occurrence of the headache or reduce an individual's susceptibility to attacks by altering the brain's response to external and internal triggers.
Collapse
Affiliation(s)
- Philip R Holland
- Headache Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
40
|
González B, Jayanthi S, Gomez N, Torres OV, Sosa MH, Bernardi A, Urbano FJ, García-Rill E, Cadet JL, Bisagno V. Repeated methamphetamine and modafinil induce differential cognitive effects and specific histone acetylation and DNA methylation profiles in the mouse medial prefrontal cortex. Prog Neuropsychopharmacol Biol Psychiatry 2018; 82:1-11. [PMID: 29247759 PMCID: PMC6983674 DOI: 10.1016/j.pnpbp.2017.12.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/04/2017] [Accepted: 12/10/2017] [Indexed: 11/28/2022]
Abstract
Methamphetamine (METH) and modafinil are psychostimulants with different long-term cognitive profiles: METH is addictive and leads to cognitive decline, whereas modafinil has little abuse liability and is a cognitive enhancer. Increasing evidence implicates epigenetic mechanisms of gene regulation behind the lasting changes that drugs of abuse and other psychotropic compounds induce in the brain, like the control of gene expression by histones 3 and 4 tails acetylation (H3ac and H4ac) and DNA cytosine methylation (5-mC). Mice were treated with a seven-day repeated METH, modafinil or vehicle protocol and evaluated in the novel object recognition (NOR) test or sacrificed 4days after last injection for molecular assays. We evaluated total H3ac, H4ac and 5-mC levels in the medial prefrontal cortex (mPFC), H3ac and H4ac promotor enrichment (ChIP) and mRNA expression (RT-PCR) of neurotransmitter systems involved in arousal, wakefulness and cognitive control, like dopaminergic (Drd1 and Drd2), α-adrenergic (Adra1a and Adra1b), orexinergic (Hcrtr1 and Hcrtr2), histaminergic (Hrh1 and Hrh3) and glutamatergic (AMPA Gria1 and NMDA Grin1) receptors. Repeated METH and modafinil treatment elicited different cognitive outcomes in the NOR test, where modafinil-treated mice performed as controls and METH-treated mice showed impaired recognition memory. METH-treated mice also showed i) decreased levels of total H3ac and H4ac, and increased levels of 5-mC, ii) decreased H3ac enrichment at promoters of Drd2, Hcrtr1/2, Hrh1 and Grin1, and increased H4ac enrichment at Drd1, Hrh1 and Grin1, iii) increased mRNA of Drd1a, Grin1 and Gria1. Modafinil-treated mice shared none of these effects and showed increased H3ac enrichment and mRNA expression at Adra1b. Modafinil and METH showed similar effects linked to decreased H3ac in Hrh3, increased H4ac in Hcrtr1, and decreased mRNA expression of Hcrtr2. The specific METH-induced epigenetic and transcriptional changes described here may be related to the long-term cognitive decline effects of the drug and its detrimental effects on mPFC function. The lack of similar epigenetic effects of chronic modafinil administration supports this notion.
Collapse
Affiliation(s)
- Betina González
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires – Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, Maryland, United States of America
| | - Natalia Gomez
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires – Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Oscar V. Torres
- Department of Behavioral Sciences, San Diego Mesa College, San Diego, California, United States of America
| | - Máximo H. Sosa
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires – Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Alejandra Bernardi
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires – Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Francisco J. Urbano
- Laboratorio de Fisiología y Biología Molecular, Instituto de Fisiología, Biología Molecular y Neurociencias (Universidad de Buenos Aires – Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Edgar García-Rill
- Center for Translational Neuroscience, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Jean-Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, Maryland, United States of America.,Corresponding authors: Veronica Bisagno, Ph.D. Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Junín 956, piso 5, C1113-Buenos Aires, Argentina. Phone: (+54-11) 4961-6784, Fax: (+54-11) 4963-8593. Jean-Lud Cadet, MD
| | - Verónica Bisagno
- Instituto de Investigaciones Farmacológicas, Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
41
|
Yu X, Franks NP, Wisden W. Sleep and Sedative States Induced by Targeting the Histamine and Noradrenergic Systems. Front Neural Circuits 2018; 12:4. [PMID: 29434539 PMCID: PMC5790777 DOI: 10.3389/fncir.2018.00004] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/11/2018] [Indexed: 01/07/2023] Open
Abstract
Sedatives target just a handful of receptors and ion channels. But we have no satisfying explanation for how activating these receptors produces sedation. In particular, do sedatives act at restricted brain locations and circuitries or more widely? Two prominent sedative drugs in clinical use are zolpidem, a GABAA receptor positive allosteric modulator, and dexmedetomidine (DEX), a selective α2 adrenergic receptor agonist. By targeting hypothalamic neuromodulatory systems both drugs induce a sleep-like state, but in different ways: zolpidem primarily reduces the latency to NREM sleep, and is a controlled substance taken by many people to help them sleep; DEX produces prominent slow wave activity in the electroencephalogram (EEG) resembling stage 2 NREM sleep, but with complications of hypothermia and lowered blood pressure—it is used for long term sedation in hospital intensive care units—under DEX-induced sedation patients are arousable and responsive, and this drug reduces the risk of delirium. DEX, and another α2 adrenergic agonist xylazine, are also widely used in veterinary clinics to sedate animals. Here we review how these two different classes of sedatives, zolpidem and dexmedetomideine, can selectively interact with some nodal points of the circuitry that promote wakefulness allowing the transition to NREM sleep. Zolpidem enhances GABAergic transmission onto histamine neurons in the hypothalamic tuberomammillary nucleus (TMN) to hasten the transition to NREM sleep, and DEX interacts with neurons in the preoptic hypothalamic area that induce sleep and body cooling. This knowledge may aid the design of more precise acting sedatives, and at the same time, reveal more about the natural sleep-wake circuitry.
Collapse
Affiliation(s)
- Xiao Yu
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Nicholas P Franks
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Centre for Neurotechnology, Imperial College London, London, United Kingdom.,UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - William Wisden
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Centre for Neurotechnology, Imperial College London, London, United Kingdom.,UK Dementia Research Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
42
|
Hypothalamic Tuberomammillary Nucleus Neurons: Electrophysiological Diversity and Essential Role in Arousal Stability. J Neurosci 2017; 37:9574-9592. [PMID: 28874450 DOI: 10.1523/jneurosci.0580-17.2017] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 08/10/2017] [Accepted: 08/19/2017] [Indexed: 11/21/2022] Open
Abstract
Histaminergic (HA) neurons, found in the posterior hypothalamic tuberomammillary nucleus (TMN), extend fibers throughout the brain and exert modulatory influence over numerous physiological systems. Multiple lines of evidence suggest that the activity of HA neurons is important in the regulation of vigilance despite the lack of direct, causal evidence demonstrating its requirement for the maintenance of arousal during wakefulness. Given the strong correlation between HA neuron excitability and behavioral arousal, we investigated both the electrophysiological diversity of HA neurons in brain slices and the effect of their acute silencing in vivo in male mice. For this purpose, we first validated a transgenic mouse line expressing cre recombinase in histidine decarboxylase-expressing neurons (Hdc-Cre) followed by a systematic census of the membrane properties of both HA and non-HA neurons in the ventral TMN (TMNv) region. Through unsupervised hierarchical cluster analysis, we found electrophysiological diversity both between TMNv HA and non-HA neurons, and among HA neurons. To directly determine the impact of acute cessation of HA neuron activity on sleep-wake states in awake and behaving mice, we examined the effects of optogenetic silencing of TMNv HA neurons in vivo We found that acute silencing of HA neurons during wakefulness promotes slow-wave sleep, but not rapid eye movement sleep, during a period of low sleep pressure. Together, these data suggest that the tonic firing of HA neurons is necessary for the maintenance of wakefulness, and their silencing not only impairs arousal but is sufficient to rapidly and selectively induce slow-wave sleep.SIGNIFICANCE STATEMENT The function of monoaminergic systems and circuits that regulate sleep and wakefulness is often disrupted as part of the pathophysiology of many neuropsychiatric disorders. One such circuit is the posterior hypothalamic histamine (HA) system, implicated in supporting wakefulness and higher brain function, but has been difficult to selectively manipulate owing to cellular heterogeneity in this region. Here we use a transgenic mouse to interrogate both the characteristic firing properties of HA neurons and their specific role in maintaining wakefulness. Our results demonstrate that the acute, cell type-specific silencing of HA neurons during wakefulness is sufficient to not only impair arousal but to rapidly and selectively induce slow-wave sleep. This work furthers our understanding of HA-mediated mechanisms that regulate behavioral arousal.
Collapse
|
43
|
Optogenetic Investigation of Arousal Circuits. Int J Mol Sci 2017; 18:ijms18081773. [PMID: 28809797 PMCID: PMC5578162 DOI: 10.3390/ijms18081773] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/06/2017] [Accepted: 08/09/2017] [Indexed: 12/13/2022] Open
Abstract
Modulation between sleep and wake states is controlled by a number of heterogeneous neuron populations. Due to the topological proximity and genetic co-localization of the neurons underlying sleep-wake state modulation optogenetic methods offer a significant improvement in the ability to benefit from both the precision of genetic targeting and millisecond temporal control. Beginning with an overview of the neuron populations mediating arousal, this review outlines the progress that has been made in the investigation of arousal circuits since the incorporation of optogenetic techniques and the first in vivo application of optogenetic stimulation in hypocretin neurons in the lateral hypothalamus. This overview is followed by a discussion of the future progress that can be made by incorporating more recent technological developments into the research of neural circuits.
Collapse
|
44
|
Hypocretin (orexin) is critical in sustaining theta/gamma-rich waking behaviors that drive sleep need. Proc Natl Acad Sci U S A 2017. [PMID: 28630298 DOI: 10.1073/pnas.1700983114] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hcrt gene inactivation in mice leads to behavioral state instability, abnormal transitions to paradoxical sleep, and cataplexy, hallmarks of narcolepsy. Sleep homeostasis is, however, considered unimpaired in patients and narcoleptic mice. We find that whereas Hcrtko/ko mice respond to 6-h sleep deprivation (SD) with a slow-wave sleep (SWS) EEG δ (1.0 to 4.0 Hz) power rebound like WT littermates, spontaneous waking fails to induce a δ power reflecting prior waking duration. This correlates with impaired θ (6.0 to 9.5 Hz) and fast-γ (55 to 80 Hz) activity in prior waking. We algorithmically identify a theta-dominated wakefulness (TDW) substate underlying motivated behaviors and typically preceding cataplexy in Hcrtko/ko mice. Hcrtko/ko mice fully implement TDW when waking is enforced, but spontaneous TDW episode duration is greatly reduced. A reformulation of the classic sleep homeostasis model, where homeostatic pressure rises exclusively in TDW rather than all waking, predicts δ power dynamics both in Hcrtko/ko and WT mouse baseline and recovery SWS. The low homeostatic impact of Hcrtko/ko mouse spontaneous waking correlates with decreased cortical expression of neuronal activity-related genes (notably Bdnf, Egr1/Zif268, and Per2). Thus, spontaneous TDW stability relies on Hcrt to sustain θ/fast-γ network activity and associated plasticity, whereas other arousal circuits sustain TDW during SD. We propose that TDW identifies a discrete global brain activity mode that is regulated by context-dependent neuromodulators and acts as a major driver of sleep homeostasis. Hcrt loss in Hcrtko/ko mice causes impaired TDW maintenance in baseline wake and blunted δ power in SWS, reproducing, respectively, narcolepsy excessive daytime sleepiness and poor sleep quality.
Collapse
|
45
|
Low cerebrospinal fluid hypocretin levels during sudden infant death syndrome (SIDS) risk period. Sleep Med 2017; 33:57-60. [DOI: 10.1016/j.sleep.2016.12.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/15/2016] [Accepted: 12/17/2016] [Indexed: 11/20/2022]
|
46
|
Eban-Rothschild A, de Lecea L. Neuronal substrates for initiation, maintenance, and structural organization of sleep/wake states. F1000Res 2017; 6:212. [PMID: 28357049 PMCID: PMC5345773 DOI: 10.12688/f1000research.9677.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2017] [Indexed: 11/20/2022] Open
Abstract
Animals continuously alternate between sleep and wake states throughout their life. The daily organization of sleep and wakefulness is orchestrated by circadian, homeostatic, and motivational processes. Over the last decades, much progress has been made toward determining the neuronal populations involved in sleep/wake regulation. Here, we will discuss how the application of advanced
in vivo tools for cell type–specific manipulations now permits the functional interrogation of different features of sleep/wake state regulation: initiation, maintenance, and structural organization. We will specifically focus on recent studies examining the roles of wake-promoting neuronal populations.
Collapse
Affiliation(s)
- Ada Eban-Rothschild
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
47
|
Genetic Analysis of Histamine Signaling in Larval Zebrafish Sleep. eNeuro 2017; 4:eN-NWR-0286-16. [PMID: 28275716 PMCID: PMC5334454 DOI: 10.1523/eneuro.0286-16.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 02/07/2017] [Accepted: 02/09/2017] [Indexed: 01/11/2023] Open
Abstract
Pharmacological studies in mammals and zebrafish suggest that histamine plays an important role in promoting arousal. However, genetic studies using rodents with disrupted histamine synthesis or signaling have revealed only subtle or no sleep/wake phenotypes. Studies of histamine function in mammalian arousal are complicated by its production in cells of the immune system and its roles in humoral and cellular immunity, which can have profound effects on sleep/wake states. To avoid this potential confound, we used genetics to explore the role of histamine in regulating sleep in zebrafish, a diurnal vertebrate in which histamine production is restricted to neurons in the brain. Similar to rodent genetic studies, we found that zebrafish that lack histamine due to mutation of histidine decarboxylase (hdc) exhibit largely normal sleep/wake behaviors. Zebrafish containing predicted null mutations in several histamine receptors also lack robust sleep/wake phenotypes, although we are unable to verify that these mutants are completely nonfunctional. Consistent with some rodent studies, we found that arousal induced by overexpression of the neuropeptide hypocretin (Hcrt) or by stimulation of hcrt-expressing neurons is not blocked in hdc or hrh1 mutants. We also found that the number of hcrt-expressing or histaminergic neurons is unaffected in animals that lack histamine or Hcrt signaling, respectively. Thus, while acute pharmacological manipulation of histamine signaling has been shown to have profound effects on zebrafish and mammalian sleep, our results suggest that chronic loss of histamine signaling due to genetic mutations has only subtle effects on sleep in zebrafish, similar to rodents.
Collapse
|
48
|
Umehara H, Fabbri R, Provensi G, Passani MB. The hypophagic factor oleoylethanolamide differentially increases c-fos expression in appetite regulating centres in the brain of wild type and histamine deficient mice. Pharmacol Res 2016; 113:100-107. [DOI: 10.1016/j.phrs.2016.08.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/27/2016] [Accepted: 08/16/2016] [Indexed: 01/12/2023]
|
49
|
Sleep-wake patterns, non-rapid eye movement, and rapid eye movement sleep cycles in teenage narcolepsy. Sleep Med 2016; 33:47-56. [PMID: 28449905 DOI: 10.1016/j.sleep.2016.08.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 08/13/2016] [Accepted: 08/15/2016] [Indexed: 11/23/2022]
Abstract
BACKGROUND To further characterize sleep disorders associated with narcolepsy, we assessed the sleep-wake patterns, rapid eye movement (REM), and non-REM (NREM) sleep cycles in Chinese teenagers with narcolepsy. METHODS A total of 14 Chinese type 1 narcoleptic patients (13.4 ± 2.6 years of age) and 14 healthy age- and sex-matched control subjects (13.6 ± 1.8 years of age) were recruited. Ambulatory 24-h polysomnography was recorded for two days, with test subjects adapting to the instruments on day one and the study data collection performed on day two. RESULTS Compared with the controls, the narcoleptic patients showed a 1.5-fold increase in total sleep time over 24 h, characterized by enhanced slow-wave sleep and REM sleep. Frequent sleep-wake transitions were identified in nocturnal sleep with all sleep stages switching to wakefulness, with more awakenings and time spent in wakefulness after sleep onset. Despite eight cases of narcolepsy with sleep onset REM periods at night, the mean duration of NREM-REM sleep cycle episode and the ratio of REM/NREM sleep between patients and controls were not significantly different. CONCLUSION Our study identified hypersomnia in teenage narcolepsy despite excessive daytime sleepiness. Sleep fragmentation extended to all sleep stages, indicating impaired sleep-wake cycles and instability of sleep stages. The limited effects on NREM-REM sleep cycles suggest the relative conservation of ultradian regulation of sleep.
Collapse
|
50
|
Monoaminergic control of brain states and sensory processing: Existing knowledge and recent insights obtained with optogenetics. Prog Neurobiol 2016; 151:237-253. [PMID: 27634227 DOI: 10.1016/j.pneurobio.2016.09.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/18/2016] [Accepted: 09/10/2016] [Indexed: 01/18/2023]
Abstract
Monoamines are key neuromodulators involved in a variety of physiological and pathological brain functions. Classical studies using physiological and pharmacological tools have revealed several essential aspects of monoaminergic involvement in regulating the sleep-wake cycle and influencing sensory responses but many features have remained elusive due to technical limitations. The application of optogenetic tools led to the ability of monitoring and controlling neuronal populations with unprecedented temporal precision and neurochemical specificity. Here, we focus on recent advances in revealing the roles of some monoamines in brain state control and sensory information processing. We summarize the central position of monoamines in integrating sensory processing across sleep-wake states with an emphasis on research conducted using optogenetic techniques. Finally, we discuss the limitations and perspectives of new integrated experimental approaches in understanding the modulatory mechanisms of monoaminergic systems in the mammalian brain.
Collapse
|