1
|
Vringer M, Zhou J, Gool JK, Bijlenga D, Lammers GJ, Fronczek R, Schinkelshoek MS. Recent insights into the pathophysiology of narcolepsy type 1. Sleep Med Rev 2024; 78:101993. [PMID: 39241492 DOI: 10.1016/j.smrv.2024.101993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/09/2024]
Abstract
Narcolepsy type 1 (NT1) is a sleep-wake disorder in which people typically experience excessive daytime sleepiness, cataplexy and other sleep-wake disturbances impairing daily life activities. NT1 symptoms are due to hypocretin deficiency. The cause for the observed hypocretin deficiency remains unclear, even though the most likely hypothesis is that this is due to an auto-immune process. The search for autoantibodies and autoreactive T-cells has not yet produced conclusive evidence for or against the auto-immune hypothesis. Other mechanisms, such as reduced corticotrophin-releasing hormone production in the paraventricular nucleus have recently been suggested. There is no reversive treatment, and the therapeutic approach is symptomatic. Early diagnosis and appropriate NT1 treatment is essential, especially in children to prevent impaired cognitive, emotional and social development. Hypocretin receptor agonists have been designed to replace the attenuated hypocretin signalling. Pre-clinical and clinical trials have shown encouraging initial results. A better understanding of NT1 pathophysiology may contribute to faster diagnosis or treatments, which may cure or prevent it.
Collapse
Affiliation(s)
- Marieke Vringer
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jingru Zhou
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jari K Gool
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands; Department of Anatomy & Neurosciences, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Compulsivity, Impulsivity and Attention, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Denise Bijlenga
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Gert Jan Lammers
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Rolf Fronczek
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Mink S Schinkelshoek
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
2
|
Xu W, Ding W, Zhang Y, Wang S, Yan X, Xu Y, Zhi X, Liu R. The Role of T Cells in the Pathogenesis of Narcolepsy Type 1: A Narrative Review. Int J Mol Sci 2024; 25:11914. [PMID: 39595997 PMCID: PMC11593411 DOI: 10.3390/ijms252211914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Narcolepsy type 1 (NT1) is an uncommon, persistent sleep disorder distinguished by significant daytime sleepiness, episodes of cataplexy, and irregularities in rapid eye movement sleep. The etiology of NT1 is linked to the destruction of hypothalamic neurons responsible for the synthesis of the wake-promoting neuropeptide known as hypothalamic orexin. The pathophysiological mechanisms underlying NT1 remain inadequately elucidated; however, a model that incorporates the interplay of genetic predisposition, environmental influences, immune system factors, and a deficiency in hypocretin (HCRT) provides a framework for elucidating the pathogenesis of NT1. The prevalence of NT1 has been observed to rise following influenza A (H1N1) pdm09 and the administration of the Pandemrix influenza vaccine. The strong association between narcolepsy and the HLA-DQB1*06:02 allele strongly indicates an autoimmune etiology for this condition. Increasing evidence suggests that T cells play a critical role in this autoimmune-mediated HCRT neuronal loss. Studies have identified specific T cell subsets, including CD4+ and CD8+ T cells, that target HCRT neurons, contributing to their destruction. Clarifying the pathogenesis of NT1 driven by autoimmune T cells is crucial for the development of effective therapeutic interventions for this disorder. This review examines the risk factors associated with the pathogenesis of NT1, explores the role of T cells within the immune system in the progression of NT1, and evaluates immune-mediated animal models alongside prospective immunotherapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rongzeng Liu
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471003, China
| |
Collapse
|
3
|
Kukkonen JP, Jacobson LH, Hoyer D, Rinne MK, Borgland SL. International Union of Basic and Clinical Pharmacology CXIV: Orexin Receptor Function, Nomenclature and Pharmacology. Pharmacol Rev 2024; 76:625-688. [PMID: 38902035 DOI: 10.1124/pharmrev.123.000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
The orexin system consists of the peptide transmitters orexin-A and -B and the G protein-coupled orexin receptors OX1 and OX2 Orexin receptors are capable of coupling to all four families of heterotrimeric G proteins, and there are also other complex features of the orexin receptor signaling. The system was discovered 25 years ago and was immediately identified as a central regulator of sleep and wakefulness; this is exemplified by the symptomatology of the disorder narcolepsy with cataplexy, in which orexinergic neurons degenerate. Subsequent translation of these findings into drug discovery and development has resulted to date in three clinically used orexin receptor antagonists to treat insomnia. In addition to sleep and wakefulness, the orexin system appears to be a central player at least in addiction and reward, and has a role in depression, anxiety and pain gating. Additional antagonists and agonists are in development to treat, for instance, insomnia, narcolepsy with or without cataplexy and other disorders with excessive daytime sleepiness, depression with insomnia, anxiety, schizophrenia, as well as eating and substance use disorders. The orexin system has thus proved an important regulator of numerous neural functions and a valuable drug target. Orexin prepro-peptide and orexin receptors are also expressed outside the central nervous system, but their potential physiological roles there remain unknown. SIGNIFICANCE STATEMENT: The orexin system was discovered 25 years ago and immediately emerged as an essential sleep-wakefulness regulator. This discovery has tremendously increased the understanding of these processes and has thus far resulted in the market approval of three orexin receptor antagonists, which promote more physiological aspects of sleep than previous hypnotics. Further, orexin receptor agonists and antagonists with different pharmacodynamic properties are in development since research has revealed additional potential therapeutic indications. Orexin receptor signaling is complex and may represent novel features.
Collapse
Affiliation(s)
- Jyrki P Kukkonen
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Laura H Jacobson
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Daniel Hoyer
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Maiju K Rinne
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Stephanie L Borgland
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| |
Collapse
|
4
|
Chen G, Wang W, Wu H, Zhao X, Kang X, Ren J, Zhang J, Sun Y, He J, Sun S, Zhong Z, Shang D, Fan M, Cheng J, Zhang D, Su C, Lin J. Disrupted topological properties of structural brain networks present a glutamatergic neuropathophysiology in people with narcolepsy. Sleep 2024; 47:zsae002. [PMID: 38173348 DOI: 10.1093/sleep/zsae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/18/2023] [Indexed: 01/05/2024] Open
Abstract
STUDY OBJECTIVES Growing evidences have documented various abnormalities of the white matter bundles in people with narcolepsy. We sought to evaluate topological properties of brain structural networks, and their association with symptoms and neuropathophysiological features in people with narcolepsy. METHODS Diffusion tensor imaging was conducted for people with narcolepsy (n = 30) and matched healthy controls as well as symptoms assessment. Structural connectivity for each participant was generated to analyze global and regional topological properties and their correlations with narcoleptic features. Further human brain transcriptome was extracted and spatially registered for connectivity vulnerability. Genetic functional enrichment analysis was performed and further clarified using in vivo emission computed tomography data. RESULTS A wide and dramatic decrease in structural connectivities was observed in people with narcolepsy, with descending network degree and global efficiency. These metrics were not only correlated with sleep latency and awakening features, but also reflected alterations of sleep macrostructure in people with narcolepsy. Network-based statistics identified a small hyperenhanced subnetwork of cingulate gyrus that was closely related to rapid eye movement sleep behavior disorder (RBD) in narcolepsy. Further imaging genetics analysis suggested glutamatergic signatures were responsible for the preferential vulnerability of connectivity alterations in people with narcolepsy, while additional PET/SPECT data verified that structural alteration was significantly correlated with metabotropic glutamate receptor 5 (mGlutR5) and N-methyl-D-aspartate receptor (NMDA). CONCLUSIONS People with narcolepsy endured a remarkable decrease in the structural architecture, which was not only closely related to narcolepsy symptoms but also glutamatergic signatures.
Collapse
Affiliation(s)
- Guoyan Chen
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Wen Wang
- Department of Radiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Haoyang Wu
- Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Xianchao Zhao
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Xiaopeng Kang
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Jiafeng Ren
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jun Zhang
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Yingzhi Sun
- Department of Radiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jiaxiu He
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Shihui Sun
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Zhao Zhong
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Danqing Shang
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Mengmeng Fan
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jinxiang Cheng
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Dan Zhang
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Changjun Su
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jiaji Lin
- Department of Neurology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
- Department of Radiology, Chinese PLA General Hospital/Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
5
|
Roh SE, Xiao M, Delgado A, Kwak C, Savonenko A, Bakker A, Kwon HB, Worley P. Sleep and circadian rhythm disruption by NPTX2 loss of function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.559408. [PMID: 37808783 PMCID: PMC10557648 DOI: 10.1101/2023.09.26.559408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Sleep and circadian rhythm disruption (SCRD) is commonly observed in aging, especially in individuals who experience progressive cognitive decline to mild cognitive impairment (MCI) and Alzheimer's disease (AD). However, precise molecular mechanisms underlying the association between SCRD and aging are not fully understood. Orexin A is a well-characterized "sleep neuropeptide" that is expressed in hypothalamic neurons and evokes wake behavior. The importance of Orexin is exemplified in narcolepsy where it is profoundly down-regulated. Interestingly, the synaptic immediate early gene NPTX2 is co-expressed in Orexin neurons and is similarly reduced in narcolepsy. NPTX2 is also down-regulated in CSF of some cognitively normal older individuals and predicts the time of transition from normal cognition to MCI. The association between Orexin and NPTX2 is further evinced here where we observe that Orexin A and NPTX2 are highly correlated in CSF of cognitively normal aged individuals and raises the question of whether SCRD that are typically attributed to Orexin A loss of function may be modified by concomitant NPTX2 down-regulation. Is NPTX2 an effector of sleep or simply a reporter of orexin-dependent SCRD? To address this question, we examined NPTX2 KO mice and found they retain Orexin expression in the brain and so provide an opportunity to examine the specific contribution of NPTX2 to SCRD. Our results reveal that NPTX2 KO mice exhibit a disrupted circadian onset time, coupled with increased activity during the sleep phase, suggesting difficulties in maintaining states. Sleep EEG indicates distinct temporal allocation shifts across vigilance states, characterized by reduced wake and increased NREM time. Evident sleep fragmentation manifests through alterations of event occurrences during Wake and NREM, notably during light transition periods, in conjunction with an increased frequency of sleep transitions in NPTX2 KO mice, particularly between Wake and NREM. EEG spectral analysis indicated significant shifts in power across various frequency bands in the wake, NREM, and REM states, suggestive of disrupted neuronal synchronicity. An intriguing observation is the diminished occurrence of sleep spindles, one of the earliest measures of human sleep disruption, in NPTX2 KO mice. These findings highlight the effector role of NPTX2 loss of function as an instigator of SCRD and a potential mediator of sleep disruption in aging.
Collapse
Affiliation(s)
- Seung-Eon Roh
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Meifang Xiao
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ana Delgado
- Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chuljung Kwak
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alena Savonenko
- Department of Neuroanatomy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Arnold Bakker
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hyung-Bae Kwon
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Paul Worley
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
6
|
Kniazkina M, Dyachuk V. Does EGFR Signaling Mediate Orexin System Activity in Sleep Initiation? Int J Mol Sci 2023; 24:ijms24119505. [PMID: 37298454 DOI: 10.3390/ijms24119505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/21/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Sleep-wake cycle disorders are an important symptom of many neurological diseases, including Parkinson's disease, Alzheimer's disease, and multiple sclerosis. Circadian rhythms and sleep-wake cycles play a key role in maintaining the health of organisms. To date, these processes are still poorly understood and, therefore, need more detailed elucidation. The sleep process has been extensively studied in vertebrates, such as mammals and, to a lesser extent, in invertebrates. A complex, multi-step interaction of homeostatic processes and neurotransmitters provides the sleep-wake cycle. Many other regulatory molecules are also involved in the cycle regulation, but their functions remain largely unclear. One of these signaling systems is epidermal growth factor receptor (EGFR), which regulates the activity of neurons in the modulation of the sleep-wake cycle in vertebrates. We have evaluated the possible role of the EGFR signaling pathway in the molecular regulation of sleep. Understanding the molecular mechanisms that underlie sleep-wake regulation will provide critical insight into the fundamental regulatory functions of the brain. New findings of sleep-regulatory pathways may provide new drug targets and approaches for the treatment of sleep-related diseases.
Collapse
Affiliation(s)
- Marina Kniazkina
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Vyacheslav Dyachuk
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| |
Collapse
|
7
|
Pintwala SK, Fraigne JJ, Belsham DD, Peever JH. Immortal orexin cell transplants restore motor-arousal synchrony during cataplexy. Curr Biol 2023; 33:1550-1564.e5. [PMID: 37044089 DOI: 10.1016/j.cub.2023.03.077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/11/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023]
Abstract
Waking behaviors such as sitting or standing require suitable levels of muscle tone. But it is unclear how arousal and motor circuits communicate with one another so that appropriate motor tone occurs during wakefulness. Cataplexy is a peculiar condition in which muscle tone is involuntarily lost during normal periods of wakefulness. Cataplexy therefore provides a unique opportunity for identifying the signaling mechanisms that synchronize motor and arousal behaviors. Cataplexy occurs when hypothalamic orexin neurons are lost in narcolepsy; however, it is unclear if motor-arousal decoupling in cataplexy is directly or indirectly caused by orexin cell loss. Here, we used genomic, proteomic, chemogenetic, electrophysiological, and behavioral assays to determine if grafting orexin cells into the brain of cataplectic (i.e., orexin-/-) mice restores normal motor-arousal behaviors by preventing cataplexy. First, we engineered immortalized orexin cells and found that they not only produce and release orexin but also exhibit a gene profile that mimics native orexin neurons. Second, we show that engineered orexin cells thrive and integrate into host tissue when transplanted into the brain of mice. Next, we found that grafting only 200-300 orexin cells into the dorsal raphe nucleus-a region densely innervated by native orexin neurons-reduces cataplexy. Last, we show that real-time chemogenetic activation of orexin cells restores motor-arousal synchrony by preventing cataplexy. We suggest that orexin signaling is critical for arousal-motor synchrony during wakefulness and that the dorsal raphe plays a pivotal role in coupling arousal and motor behaviors.
Collapse
Affiliation(s)
- Sara K Pintwala
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jimmy J Fraigne
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Denise D Belsham
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - John H Peever
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
8
|
Yamamoto H, Nagumo Y, Ishikawa Y, Irukayama-Tomobe Y, Namekawa Y, Nemoto T, Tanaka H, Takahashi G, Tokuda A, Saitoh T, Nagase H, Funato H, Yanagisawa M. OX2R-selective orexin agonism is sufficient to ameliorate cataplexy and sleep/wake fragmentation without inducing drug-seeking behavior in mouse model of narcolepsy. PLoS One 2022; 17:e0271901. [PMID: 35867683 PMCID: PMC9307173 DOI: 10.1371/journal.pone.0271901] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/08/2022] [Indexed: 11/18/2022] Open
Abstract
Acquired loss of hypothalamic orexin (hypocretin)-producing neurons causes the chronic sleep disorder narcolepsy-cataplexy. Orexin replacement therapy using orexin receptor agonists is expected as a mechanistic treatment for narcolepsy. Orexins act on two receptor subtypes, OX1R and OX2R, the latter being more strongly implicated in sleep/wake regulation. However, it has been unclear whether the activation of only OX2R, or both OX1R and OX2R, is required to replace the endogenous orexin functions in the brain. In the present study, we examined whether the selective activation of OX2R is sufficient to rescue the phenotype of cataplexy and sleep/wake fragmentation in orexin knockout mice. Intracerebroventricular [Ala11, D-Leu15]-orexin-B, a peptidic OX2R-selective agonist, selectively activated OX2R-expressing histaminergic neurons in vivo, whereas intracerebroventricular orexin-A, an OX1R/OX2R non-selective agonist, additionally activated OX1R-positive noradrenergic neurons in vivo. Administration of [Ala11, D-Leu15]-orexin-B extended wake time, reduced state transition frequency between wake and NREM sleep, and reduced the number of cataplexy-like episodes, to the same degree as compared with orexin-A. Furthermore, intracerebroventricular orexin-A but not [Ala11, D-Leu15]-orexin-B induced drug-seeking behaviors in a dose-dependent manner in wild-type mice, suggesting that OX2R-selective agonism has a lower propensity for reinforcing/drug-seeking effects. Collectively, these findings provide a proof-of-concept for safer mechanistic treatment of narcolepsy-cataplexy through OX2R-selective agonism.
Collapse
Affiliation(s)
- Hikari Yamamoto
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yasuyuki Nagumo
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yukiko Ishikawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoko Irukayama-Tomobe
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yukiko Namekawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tsuyoshi Nemoto
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiromu Tanaka
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Genki Takahashi
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akihisa Tokuda
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tsuyoshi Saitoh
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiroshi Nagase
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiromasa Funato
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Anatomy, Faculty of Medicine, Toho University, Ota-ku, Tokyo, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas
- Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Frontiers of MIRAI in Policy and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
- * E-mail:
| |
Collapse
|
9
|
Tisdale RK, Yamanaka A, Kilduff TS. Animal models of narcolepsy and the hypocretin/orexin system: Past, present, and future. Sleep 2021; 44:6031626. [PMID: 33313880 DOI: 10.1093/sleep/zsaa278] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/04/2020] [Indexed: 11/12/2022] Open
Abstract
Animal models have advanced not only our understanding of the etiology and phenotype of the sleep disorder narcolepsy but have also informed sleep/wake regulation more generally. The identification of an inheritable narcolepsy phenotype in dogs in the 1970s allowed the establishment of a breeding colony at Stanford University, resulting in studies that provided the first insights into the genetics and neurotransmitter systems that underlie cataplexy and rapid-eye movement sleep atonia. Although the discovery of the hypocretin/orexin neuropeptides in 1998 initially seemed unrelated to sleep/wake control, the description of the phenotype of the prepro-orexin knockout (KO) mouse as strongly resembling cataplexy, the pathognomonic symptom of narcolepsy, along with identification of a mutation in hypocretin receptor-2 gene as the source of canine narcolepsy, unequivocally established the relationship between this system and narcolepsy. The subsequent discovery of hypocretin neuron degeneration in human narcolepsy demystified a disorder whose etiology had been unknown since its initial description 120 years earlier. These breakthroughs prompted the development of numerous other animal models that have allowed manipulation of the hypocretin/orexin system, thereby advancing our understanding of sleep/wake circuitry. While animal models have greatly informed understanding of this fascinating disorder and the role of the hypocretin/orexin system in sleep/wake control, the question of why these neurons degenerate in human narcolepsy is only beginning to be understood. The development of new immune-mediated narcolepsy models are likely to further inform the etiology of this sleep disorder and animal models will undoubtedly play a critical role in the development of novel narcolepsy therapeutics.
Collapse
Affiliation(s)
- Ryan K Tisdale
- Center for Neuroscience, Biosciences Division, SRI International
| | - 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
- Center for Neuroscience, Biosciences Division, SRI International
| |
Collapse
|
10
|
Berteotti C, Liguori C, Pace M. Dysregulation of the orexin/hypocretin system is not limited to narcolepsy but has far-reaching implications for neurological disorders. Eur J Neurosci 2020; 53:1136-1154. [PMID: 33290595 DOI: 10.1111/ejn.15077] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 12/23/2022]
Abstract
Neuropeptides orexin A and B (OX-A/B, also called hypocretin 1 and 2) are released selectively by a population of neurons which projects widely into the entire central nervous system but is localized in a restricted area of the tuberal region of the hypothalamus, caudal to the paraventricular nucleus. The OX system prominently targets brain structures involved in the regulation of wake-sleep state switching, and also orchestrates multiple physiological functions. The degeneration and dysregulation of the OX system promotes narcoleptic phenotypes both in humans and animals. Hence, this review begins with the already proven involvement of OX in narcolepsy, but it mainly discusses the new pre-clinical and clinical insights of the role of OX in three major neurological disorders characterized by sleep impairment which have been recently associated with OX dysfunction, such as Alzheimer's disease, stroke and Prader Willi syndrome, and have been emerged over the past 10 years to be strongly associated with the OX dysfunction and should be more considered in the future. In the light of the impairment of the OX system in these neurological disorders, it is conceivable to speculate that the integrity of the OX system is necessary for a healthy functioning body.
Collapse
Affiliation(s)
- Chiara Berteotti
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Claudio Liguori
- Sleep Medicine Centre, Neurology Unit, University Hospital Tor Vergata, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Marta Pace
- Genetics and Epigenetics of Behaviour Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
11
|
Harris T, Bugescu R, Kelly J, Makela A, Sotzen M, Sisk C, Atkin G, Pratt R, Crockett E, Leinninger G. DLK1 Expressed in Mouse Orexin Neurons Modulates Anxio-Depressive Behavior but Not Energy Balance. Brain Sci 2020; 10:brainsci10120975. [PMID: 33322758 PMCID: PMC7764426 DOI: 10.3390/brainsci10120975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 11/16/2022] Open
Abstract
Lateral hypothalamic area (LHA) neurons expressing the neuropeptide orexin (OX) are implicated in obesity and anxio-depression. However, these neurons release OX as well as a host of other proteins that might contribute to normal physiology and disease states. We hypothesized that delta-like homolog 1 (DLK1), a protein reported to be co-expressed by all OX neurons, contributes to the regulation of energy balance and/or anxio-depression. Consistent with previous reports, we found that all rat OX neurons co-express DLK1. Yet, in mice and humans only a subset of OX neurons co-expressed DLK1. Since human OX-DLK1 distribution is more similar to mice than rats, mice are a comparable model to assess the human physiologic role of DLK1. We therefore used a viral lesion strategy to selectively delete DLK1 within the LHA of adult mice (DLK1Null) to reveal its role in body weight and behavior. Adult-onset DLK1 deletion had no impact on body weight or ingestive behavior. However, DLK1Null mice engaged in more locomotor activity than control mice and had decreased anxiety and depression measured via the elevated plus maze and forced swim tests. These data suggest that DLK1 expression via DLK1-expressing OX neurons primarily contributes to anxio-depression behaviors without impacting body weight.
Collapse
Affiliation(s)
- Tatiyana Harris
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (T.H.); (R.B.); (J.K.); (A.M.); (M.S.)
| | - Raluca Bugescu
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (T.H.); (R.B.); (J.K.); (A.M.); (M.S.)
| | - Jaylyn Kelly
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (T.H.); (R.B.); (J.K.); (A.M.); (M.S.)
| | - Anna Makela
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (T.H.); (R.B.); (J.K.); (A.M.); (M.S.)
| | - Morgan Sotzen
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (T.H.); (R.B.); (J.K.); (A.M.); (M.S.)
| | - Cheryl Sisk
- Neuroscience Program, Department of Psychology, Michigan State University, East Lansing, MI 48824, USA;
| | - Graham Atkin
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA;
| | - Rebecca Pratt
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA;
| | - Elahé Crockett
- Department of Medicine, Michigan State University, East Lansing, MI 48824, USA;
| | - Gina Leinninger
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA; (T.H.); (R.B.); (J.K.); (A.M.); (M.S.)
- Correspondence:
| |
Collapse
|
12
|
Yaeger JD, Krupp KT, Gale JJ, Summers CH. Counterbalanced microcircuits for Orx1 and Orx2 regulation of stress reactivity. MEDICINE IN DRUG DISCOVERY 2020. [DOI: 10.1016/j.medidd.2020.100059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
13
|
Wang D, Zhang J, Bai Y, Zheng X, Alizamini MM, Shang W, Yang Q, Li M, Li Y, Sui N. Melanin-concentrating hormone in rat nucleus accumbens or lateral hypothalamus differentially impacts morphine and food seeking behaviors. J Psychopharmacol 2020; 34:478-489. [PMID: 31909693 DOI: 10.1177/0269881119895521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Identifying neural substrates that are differentially affected by drugs of abuse and natural rewards is key to finding a target for an efficacious treatment for substance abuse. Melanin-concentrating hormone is a polypeptide with an inhibitory effect on the mesolimbic dopamine system. Here we test the hypothesis that melanin-concentrating hormone in the lateral hypothalamus and nucleus accumbens shell is differentially involved in the regulation of morphine and food-rewarded behaviors. METHODS Male Sprague-Dawley rats were trained with morphine (5.0 mg/kg, subcutaneously) or food pellets (standard chow, 10-14 g) to induce a conditioned place preference, immediately followed by extinction training. Melanin-concentrating hormone (1.0 µg/side) or saline was infused into the nucleus accumbens shell or lateral hypothalamus before the reinstatement primed by morphine or food, and locomotor activity was simultaneously monitored. As the comparison, melanin-concentrating hormone was also microinjected into the nucleus accumbens shell or lateral hypothalamus before the expression of food or morphine-induced conditioned place preference. RESULTS Microinfusion of melanin-concentrating hormone into the nucleus accumbens shell (but not into the lateral hypothalamus) prevented the reinstatement of morphine conditioned place preference but had no effect on the reinstatement of food conditioned place preference. In contrast, microinfusion of melanin-concentrating hormone into the lateral hypothalamus (but not in the nucleus accumbens shell) inhibited the reinstatement of food conditioned place preference but had no effect on the reinstatement of morphine conditioned place preference. CONCLUSIONS These results suggest a clear double dissociation of melanin-concentrating hormone in morphine/food rewarding behaviors and melanin-concentrating hormone in the nucleus accumbens shell. Melanin-concentrating hormone could be a potential target for therapeutic intervention for morphine abuse without affecting natural rewards.
Collapse
Affiliation(s)
- Dongmei Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Jianjun Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Yunjing Bai
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Xigeng Zheng
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Mirmohammadali M Alizamini
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Wen Shang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Qingxiong Yang
- School of Karst Science, Guizhou Normal University/State Engineering Technology Institute for Karst Desertification Control, Guiyang, China
| | - Ming Li
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Yonghui Li
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Nan Sui
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
14
|
Update on narcolepsy. J Neurol 2019; 266:1809-1815. [DOI: 10.1007/s00415-019-09310-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/01/2019] [Accepted: 04/02/2019] [Indexed: 12/20/2022]
|
15
|
Tanaka S, Honda Y, Takaku S, Koike T, Oe S, Hirahara Y, Yoshida T, Takizawa N, Takamori Y, Kurokawa K, Kodama T, Yamada H. Involvement of PLAGL1/ZAC1 in hypocretin/orexin transcription. Int J Mol Med 2019; 43:2164-2176. [PMID: 30896835 PMCID: PMC6445593 DOI: 10.3892/ijmm.2019.4143] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 03/19/2019] [Indexed: 12/16/2022] Open
Abstract
The hypocretin/orexin neuropeptide system coordinates the regulation of various physiological processes. Our previous study reported that a reduction in the expression of pleomorphic adenoma gene-like 1 (Plagl1), which encodes a C2H2 zinc-finger transcription factor, occurs in hypocretin neuron-ablated transgenic mice, suggesting that PLAGL1 is co-expressed in hypocretin neurons and regulates hypocretin transcription. The present study examined whether canonical prepro-hypocretin transcription is functionally modulated by PLAGL1. Double immunostaining indicated that the majority of hypocretin neurons were positive for PLAGL1 immunore-activity in the nucleus. Notably, PLAGL1 immunoreactivity in hypocretin neurons was altered in response to several conditions affecting hypocretin function. An uneven localization of PLAGL1 was detected in the nuclei of hypocretin neurons following sleep deprivation. Chromatin immunoprecipitation revealed that endogenous PLAGL1 may bind to a putative PLAGL1-binding site in the proximal region of the hypocretin gene, in the murine hypothalamus. In addition, electroporation of the PLAGL1 expression vector into the fetal hypothalamus promoted hypothalamic hypocretin transcription. These results suggested that PLAGL1 may regulate hypothalamic hypocretin transcription.
Collapse
Affiliation(s)
- Susumu Tanaka
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka 573‑1010, Japan
| | - Yoshiko Honda
- SLEEP Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156‑8506, Japan
| | - Shizuka Takaku
- SLEEP Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156‑8506, Japan
| | - Taro Koike
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka 573‑1010, Japan
| | - Souichi Oe
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka 573‑1010, Japan
| | - Yukie Hirahara
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka 573‑1010, Japan
| | - Takashi Yoshida
- Department of Urology and Andrology, Kansai Medical University, Hirakata, Osaka 573‑1191, Japan
| | - Nae Takizawa
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka 573‑1010, Japan
| | - Yasuharu Takamori
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka 573‑1010, Japan
| | - Kiyoshi Kurokawa
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka 573‑1010, Japan
| | - Tohru Kodama
- SLEEP Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156‑8506, Japan
| | - Hisao Yamada
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Osaka 573‑1010, Japan
| |
Collapse
|
16
|
Abstract
Narcolepsy is the most common neurological cause of chronic sleepiness. The discovery about 20 years ago that narcolepsy is caused by selective loss of the neurons producing orexins (also known as hypocretins) sparked great advances in the field. Here, we review the current understanding of how orexin neurons regulate sleep-wake behaviour and the consequences of the loss of orexin neurons. We also summarize the developing evidence that narcolepsy is an autoimmune disorder that may be caused by a T cell-mediated attack on the orexin neurons and explain how these new perspectives can inform better therapeutic approaches.
Collapse
Affiliation(s)
- Carrie E Mahoney
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Andrew Cogswell
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Igor J Koralnik
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Abstract
Purpose of Review The aim of this review was to summarize collected data on the role of orexin and orexin neurons in the control of sleep and blood pressure. Recent Findings Although orexins (hypocretins) have been known for only 20 years, an impressive amount of data is now available regarding their physiological role. Hypothalamic orexin neurons are responsible for the control of food intake and energy expenditure, motivation, circadian rhythm of sleep and wake, memory, cognitive functions, and the cardiovascular system. Multiple studies show that orexinergic stimulation results in increased blood pressure and heart rate and that this effect may be efficiently attenuated by orexinergic antagonism. Increased activity of orexinergic neurons is also observed in animal models of hypertension. Summary Pharmacological intervention in the orexinergic system is now one of the therapeutic possibilities in insomnia. Although the role of orexin in the control of blood pressure is well described, we are still lacking clinical evidence that this is a possibility for a new approach in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Mariusz Sieminski
- Department of Emergency Medicine, Medical University of Gdansk, Smoluchowskiego 17, 80-235, Gdansk, Poland.
| | - Jacek Szypenbejl
- Department of Emergency Medicine, Medical University of Gdansk, Smoluchowskiego 17, 80-235, Gdansk, Poland
| | - Eemil Partinen
- Department of Neurology, University of Helsinki, Helsinki, Finland
- Vitalmed Helsinki Sleep Clinic, Helsinki, Finland
| |
Collapse
|
18
|
Moorman DE. The hypocretin/orexin system as a target for excessive motivation in alcohol use disorders. Psychopharmacology (Berl) 2018; 235:1663-1680. [PMID: 29508004 PMCID: PMC5949267 DOI: 10.1007/s00213-018-4871-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/20/2018] [Indexed: 12/17/2022]
Abstract
The hypocretin/orexin (ORX) system has been repeatedly demonstrated to regulate motivation for drugs of abuse, including alcohol. In particular, ORX seems to be critically involved in highly motivated behaviors, as is observed in high-seeking individuals in a population, in the seeking of highly palatable substances, and in models of dependence. It seems logical that this system could be considered as a potential target for treatment for addiction, particularly alcohol addiction, as ORX pharmacological manipulations significantly reduce drinking. However, the ORX system also plays a role in a wide range of other behaviors, emotions, and physiological functions and is disrupted in a number of non-dependence-associated disorders. It is therefore important to consider how the ORX system might be optimally targeted for potential treatment for alcohol use disorders either in combination with or separate from its role in other functions or diseases. This review will focus on the role of ORX in alcohol-associated behaviors and whether and how this system could be targeted to treat alcohol use disorders while avoiding impacts on other ORX-relevant functions. A brief overview of the ORX system will be followed by a discussion of some of the factors that makes it particularly intriguing as a target for alcohol addiction treatment, a consideration of some potential challenges associated with targeting this system and, finally, some future directions to optimize new treatments.
Collapse
Affiliation(s)
- David E Moorman
- Department of Psychological and Brain Sciences, Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, 528 Tobin Hall, 135 Hicks Way, Amherst, MA, 01003, USA.
| |
Collapse
|
19
|
Enevoldsen LH, Tindborg M, Hovmand NL, Christoffersen C, Ellingsgaard H, Suetta C, Stallknecht BM, Jennum PJ, Kjær A, Gammeltoft S. Functional brown adipose tissue and sympathetic activity after cold exposure in humans with type 1 narcolepsy. Sleep 2018; 41:4996398. [DOI: 10.1093/sleep/zsy092] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Indexed: 11/14/2022] Open
Affiliation(s)
- Lotte Hahn Enevoldsen
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Denmark
| | - Marie Tindborg
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | | | - Christina Christoffersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, Denmark
| | | | - Charlotte Suetta
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Denmark
| | - Bente Merete Stallknecht
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | | | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Denmark
| | | |
Collapse
|
20
|
Narp Mediates Antidepressant-Like Effects of Electroconvulsive Seizures. Neuropsychopharmacology 2018; 43:1088-1098. [PMID: 29052614 PMCID: PMC5854807 DOI: 10.1038/npp.2017.252] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 10/11/2017] [Accepted: 10/15/2017] [Indexed: 12/28/2022]
Abstract
Growing recognition of persistent cognitive defects associated with electroconvulsive therapy (ECT), a highly effective and commonly used antidepressant treatment, has spurred interest in identifying its mechanism of action to guide development of safer treatment options. However, as repeated seizure activity elicits a bewildering array of electrophysiological and biochemical effects, this goal has remained elusive. We have examined whether deletion of Narp, an immediate early gene induced by electroconvulsive seizures (ECS), blocks its antidepressant efficacy. Based on multiple measures, we infer that Narp knockout mice undergo normal seizure activity in this paradigm, yet fail to display antidepressant-like behavioral effects of ECS. Although Narp deletion does not suppress ECS-induced proliferation in the dentate gyrus, it blocks dendritic outgrowth of immature granule cell neurons in the dentate molecular layer induced by ECS. Taken together, these findings indicate that Narp contributes to the antidepressant action of ECT and implicate the ability of ECS to induce dendritic arborization of differentiating granule cells as a relevant step in eliciting this response.
Collapse
|
21
|
Ma S, Hangya B, Leonard CS, Wisden W, Gundlach AL. Dual-transmitter systems regulating arousal, attention, learning and memory. Neurosci Biobehav Rev 2018; 85:21-33. [PMID: 28757457 PMCID: PMC5747977 DOI: 10.1016/j.neubiorev.2017.07.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 07/16/2017] [Indexed: 01/12/2023]
Abstract
An array of neuromodulators, including monoamines and neuropeptides, regulate most behavioural and physiological traits. In the past decade, dramatic progress has been made in mapping neuromodulatory circuits, in analysing circuit dynamics, and interrogating circuit function using pharmacogenetic, optogenetic and imaging methods This review will focus on several distinct neural networks (acetylcholine/GABA/glutamate; histamine/GABA; orexin/glutamate; and relaxin-3/GABA) that originate from neural hubs that regulate wakefulness and related attentional and cognitive processes, and highlight approaches that have identified dual transmitter roles in these behavioural functions. Modulation of these different neural networks might be effective treatments of diseases related to arousal/sleep dysfunction and of cognitive dysfunction in psychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Sherie Ma
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia.
| | - Balázs Hangya
- 'Lendület' Laboratory of Systems Neuroscience, Department of Cellular and Network Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | | | - William Wisden
- Department of Life Sciences, Imperial College London, London, UK
| | - Andrew L Gundlach
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia; Department of Anatomy and Neuroscience, The University of Melbourne, Victoria, Australia.
| |
Collapse
|
22
|
Mavanji V, Butterick TA, Duffy CM, Nixon JP, Billington CJ, Kotz CM. Orexin/hypocretin treatment restores hippocampal-dependent memory in orexin-deficient mice. Neurobiol Learn Mem 2017; 146:21-30. [PMID: 29107703 DOI: 10.1016/j.nlm.2017.10.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/02/2017] [Accepted: 10/27/2017] [Indexed: 12/31/2022]
Abstract
Orexin A is produced in neurons of the lateral, perifornical and dorsomedial regions of the lateral hypothalamic area, which then project widely throughout the central nervous system to regulate arousal state, sleep-wake architecture, energy homeostasis and cognitive processes. Disruption of orexin signaling leads to sleep disturbances and increased body mass index, but recent studies also indicate that orexin neuron activation improves learning and memory. We hypothesized that hippocampal orexin receptor activation improves memory. To test this idea, we obtained orexin/ataxin-3 (O/A3) mice, which become deficient in orexin neurons by about 12 weeks of age. We first measured hippocampal orexin receptor 1 (OX1R) gene expression and protein levels, then tested acquisition and consolidation of two-way active avoidance (TWAA) memory, a hippocampal-dependent learning and memory task. Finally, we determined if exogenous intra-hippocampal OXA treatment could reverse cognitive impairment (as determined by TWAA) in OA/3 mice. We showed that OX1R mRNA expression and protein levels were significantly elevated in O/A3 mice, indicating the potential for preserved orexin responsiveness. The O/A3 mice were significantly impaired in TWAA memory vs. control mice, but OXA treatment (both acute and chronic) reversed these memory deficits. These results demonstrate that orexin plays an important role in hippocampal-dependent consolidation of two-way active avoidance memory, and orexin replacement can rescue the cognitive impairment.
Collapse
Affiliation(s)
- Vijayakumar Mavanji
- Research Service, Veterans Affairs Health Care System, Minneapolis, MN 55417 USA
| | - Tammy A Butterick
- Research Service, Veterans Affairs Health Care System, Minneapolis, MN 55417 USA; Department of Food Science and Nutrition, University of Minnesota, St Paul, MN 55108 USA; Minnesota Obesity Center, St Paul, MN 55108 USA
| | - Cayla M Duffy
- Research Service, Veterans Affairs Health Care System, Minneapolis, MN 55417 USA; Department of Food Science and Nutrition, University of Minnesota, St Paul, MN 55108 USA; Minnesota's Discovery, Research and Innovation Economy, Brain Conditions, University of Minnesota, Minneapolis, MN 55455
| | - Joshua P Nixon
- Research Service, Veterans Affairs Health Care System, Minneapolis, MN 55417 USA; Department of Food Science and Nutrition, University of Minnesota, St Paul, MN 55108 USA
| | - Charles J Billington
- Research Service, Veterans Affairs Health Care System, Minneapolis, MN 55417 USA; Minnesota Obesity Center, St Paul, MN 55108 USA; Department of Medicine, University of Minnesota, Minneapolis, MN 55455 USA
| | - Catherine M Kotz
- Research Service, Veterans Affairs Health Care System, Minneapolis, MN 55417 USA; Minnesota Obesity Center, St Paul, MN 55108 USA; Geriatric Research Education Clinical Center, Veterans Affairs Health Care System, Minneapolis, MN 55417 USA; Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN 55455 USA.
| |
Collapse
|
23
|
Neurochemical Heterogeneity Among Lateral Hypothalamic Hypocretin/Orexin and Melanin-Concentrating Hormone Neurons Identified Through Single-Cell Gene Expression Analysis. eNeuro 2017; 4:eN-NWR-0013-17. [PMID: 28966976 PMCID: PMC5617207 DOI: 10.1523/eneuro.0013-17.2017] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 08/14/2017] [Accepted: 08/25/2017] [Indexed: 02/06/2023] Open
Abstract
The lateral hypothalamic area (LHA) lies at the intersection of multiple neural and humoral systems and orchestrates fundamental aspects of behavior. Two neuronal cell types found in the LHA are defined by their expression of hypocretin/orexin (Hcrt/Ox) and melanin-concentrating hormone (MCH) and are both important regulators of arousal, feeding, and metabolism. Conflicting evidence suggests that these cell populations have a more complex signaling repertoire than previously appreciated, particularly in regard to their coexpression of other neuropeptides and the machinery for the synthesis and release of GABA and glutamate. Here, we undertook a single-cell expression profiling approach to decipher the neurochemical phenotype, and heterogeneity therein, of Hcrt/Ox and MCH neurons. In transgenic mouse lines, we used single-cell quantitative polymerase chain reaction (qPCR) to quantify the expression of 48 key genes, which include neuropeptides, fast neurotransmitter components, and other key markers, which revealed unexpected neurochemical diversity. We found that single MCH and Hcrt/Ox neurons express transcripts for multiple neuropeptides and markers of both excitatory and inhibitory fast neurotransmission. Virtually all MCH and approximately half of the Hcrt/Ox neurons sampled express both the machinery for glutamate release and GABA synthesis in the absence of a vesicular GABA release pathway. Furthermore, we found that this profile is characteristic of a subpopulation of LHA glutamatergic neurons but contrasts with a broad population of LHA GABAergic neurons. Identifying the neurochemical diversity of Hcrt/Ox and MCH neurons will further our understanding of how these populations modulate postsynaptic excitability through multiple signaling mechanisms and coordinate diverse behavioral outputs.
Collapse
|
24
|
Liu H, Kong X, Chen F. Mkrn3 functions as a novel ubiquitin E3 ligase to inhibit Nptx1 during puberty initiation. Oncotarget 2017; 8:85102-85109. [PMID: 29156706 PMCID: PMC5689596 DOI: 10.18632/oncotarget.19347] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 06/26/2017] [Indexed: 12/12/2022] Open
Abstract
Central precocious puberty (CPP) is attributed to the disorder of some trigger factors those can activate the hypothalamic-pituitary-gonadal axis controlled by GnRH neurons. Many recent studies reveal one of those trigger factors, Makorin ring finger protein 3 (Mkrn3), whose loss-of-function mutations are implicated in CPP. Although Mkrn3 contained zinc Ring finger domain is considered as a putative E3 ubiquitin ligase, its actual function is never reported. Here, our results demonstrated that in mice hypothalamus before and when puberty initiated, Mkrn3 expressed the reversed tendency with Nptx1, which is an important secreted protein for neuron development. Furthermore, our data manifested that Mkrn3 interacted and suppressed Nptx1 activity. And the Ring finger domain of Mkrn3 contained was determined to be essential for binding with Nptx1 for its polyubiquitination during the puberty initiation. Our study shed light on the molecular insights into the function of Mkrn3 in the events of puberty initiation.
Collapse
Affiliation(s)
- Huifang Liu
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangxin Kong
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengling Chen
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Singh P, Thakur MK. Histone Deacetylase 2 Inhibition Attenuates Downregulation of Hippocampal Plasticity Gene Expression during Aging. Mol Neurobiol 2017; 55:2432-2442. [PMID: 28364391 DOI: 10.1007/s12035-017-0490-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/14/2017] [Indexed: 12/31/2022]
Abstract
The brain undergoes several anatomical, biochemical, and molecular changes during aging, which subsequently result in downregulation of synaptic plasticity genes and decline of memory. However, the regulation of these genes during aging is not clearly understood. Previously, we reported that the expression of histone deacetylase (HDAC)2 was upregulated in the hippocampus of old mice and negatively correlated with the decline in recognition memory. As HDAC2 regulates key synaptic plasticity neuronal immediate early genes (IEGs), we have examined their expression and epigenetic regulation. We noted that the expression of neuronal IEGs decreased both at mRNA and protein level in the hippocampus of old mice. To explore the underlying regulation, we analyzed the binding of HDAC2 and level of histone acetylation at the promoter of neuronal IEGs. While the binding of HDAC2 was higher, H3K9 and H3K14 acetylation level was lower at the promoter of these genes in old as compared to young and adult mice. Further, we inhibited HDAC2 non-specifically by sodium butyrate and specifically by antisense oligonucleotide to recover epigenetic modification, expression of neuronal IEGs, and memory in old mice. Inhibition of HDAC2 increased histone H3K9 and H3K14 acetylation level at the promoter of neuronal IEGs, their expression, and recognition memory in old mice as compared to control. Thus, inhibition of HDAC2 can be used as a therapeutic target to recover decline in memory due to aging and associated neurological disorders.
Collapse
Affiliation(s)
- Padmanabh Singh
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India
| | - M K Thakur
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India.
| |
Collapse
|
26
|
Villano I, Messina A, Valenzano A, Moscatelli F, Esposito T, Monda V, Esposito M, Precenzano F, Carotenuto M, Viggiano A, Chieffi S, Cibelli G, Monda M, Messina G. Basal Forebrain Cholinergic System and Orexin Neurons: Effects on Attention. Front Behav Neurosci 2017; 11:10. [PMID: 28197081 PMCID: PMC5281635 DOI: 10.3389/fnbeh.2017.00010] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 01/12/2017] [Indexed: 12/27/2022] Open
Abstract
The basal forebrain (BF) cholinergic system has an important role in attentive functions. The cholinergic system can be activated by different inputs, and in particular, by orexin neurons, whose cell bodies are located within the postero-lateral hypothalamus. Recently the orexin-producing neurons have been proved to promote arousal and attention through their projections to the BF. The aim of this review article is to summarize the evidence showing that the orexin system contributes to attentional processing by an increase in cortical acetylcholine release and in cortical neurons activity.
Collapse
Affiliation(s)
- Ines Villano
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Antonietta Messina
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Fiorenzo Moscatelli
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy; Department of Motor, Human and Health Science, University of Rome, "Foro Italico"Rome, Italy
| | - Teresa Esposito
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Vincenzo Monda
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Maria Esposito
- Department of Mental Health, Physical and Preventive Medicine, Second University of Naples Naples, Italy
| | - Francesco Precenzano
- Department of Mental Health, Physical and Preventive Medicine, Second University of Naples Naples, Italy
| | - Marco Carotenuto
- Department of Mental Health, Physical and Preventive Medicine, Second University of NaplesNaples, Italy; Neapolitan Brain Group (NBG), Clinic of Child and Adolescent Neuropsychiatry, Department of Mental, Physical Health and Preventive Medicine, Second University of NaplesNaples, Italy
| | - Andrea Viggiano
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno Salerno, Italy
| | - Sergio Chieffi
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Giuseppe Cibelli
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Giovanni Messina
- Department of Experimental Medicine, Second University of NaplesNaples, Italy; Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| |
Collapse
|
27
|
Cristino L, Imperatore R, Palomba L, Di Marzo V. The Endocannabinoid System in Leptin-Driven Changes of Orexinergic Signaling Under Physiological and Pathological Conditions. ENDOCANNABINOIDS AND LIPID MEDIATORS IN BRAIN FUNCTIONS 2017:1-26. [DOI: 10.1007/978-3-319-57371-7_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
28
|
Tanaka S, Takizawa N, Honda Y, Koike T, Oe S, Toyoda H, Kodama T, Yamada H. Hypocretin/orexin loss changes the hypothalamic immune response. Brain Behav Immun 2016; 57:58-67. [PMID: 27318095 DOI: 10.1016/j.bbi.2016.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 12/26/2022] Open
Abstract
Hypocretin, also known as orexin, maintains the vigilance state and regulates various physiological processes, such as arousal, sleep, food intake, energy expenditure, and reward. Previously, we found that when wild-type mice and hypocretin/ataxin-3 littermates (which are depleted of hypothalamic hypocretin-expressing neurons postnatally) were administered lipopolysaccharide (LPS), the two genotypes exhibited significant differences in their sleep/wake cycle, including differences in the degree of increase in sleep periods and in recovery from sickness behaviour. In the present study, we examined changes in the hypothalamic vigilance system and in the hypothalamic expression of inflammatory factors in response to LPS in hypocretin/ataxin-3 mice. Peripheral immune challenge with LPS affected the hypothalamic immune response and vigilance states. This response was altered by the loss of hypocretin. Hypocretin expression was inhibited after LPS injection in both hypocretin/ataxin-3 mice and their wild-type littermates, but expression was completely abolished only in hypocretin/ataxin-3 mice. Increases in the number of histidine decarboxylase (HDC)-positive cells and in Hdc mRNA expression were found in hypocretin/ataxin-3 mice, and this increase was suppressed by LPS. Hypocretin loss did not impact the change in expression of hypothalamic inflammatory factors in response to LPS, except for interferon gamma and colony stimulating factor 3. The number of c-Fos-positive/HDC-positive cells in hypocretin/ataxin-3 mice administered LPS injections was elevated, even during the rest period, in all areas, suggesting that there is an increase in the activity of histaminergic neurons in hypocretin/ataxin-3 mice following LPS injection. Taken together, our results suggest a novel role for hypocretin in the hypothalamic response to peripheral immune challenge. Our findings contribute to the understanding of the pathophysiology of narcolepsy.
Collapse
Affiliation(s)
- Susumu Tanaka
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Japan; SLEEP Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - Nae Takizawa
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Japan
| | - Yoshiko Honda
- SLEEP Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Taro Koike
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Japan
| | - Souichi Oe
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Japan
| | - Hiromi Toyoda
- SLEEP Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tohru Kodama
- SLEEP Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hisao Yamada
- Department of Anatomy and Cell Science, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
29
|
Srivas S, Thakur MK. Epigenetic regulation of neuronal immediate early genes is associated with decline in their expression and memory consolidation in scopolamine-induced amnesic mice. Mol Neurobiol 2016; 54:5107-5119. [PMID: 27553230 DOI: 10.1007/s12035-016-0047-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/08/2016] [Indexed: 01/08/2023]
Abstract
Recently, we reported a correlation of scopolamine mediated decline in memory consolidation with increase in the expression of DNA methyltransferase 1 (DNMT1) and histone deacetylase 2 (HDAC2) in the mouse hippocampus. Memory consolidation is a protein synthesis-dependent process which involves the expression of synaptic plasticity genes, particularly neuronal immediate early genes (IEGs). However, the mechanism of regulation of these genes during decline in memory is poorly understood. Therefore, we have studied the epigenetic regulation of expression of neuronal IEGs in scopolamine-induced amnesic mice. Scopolamine significantly impaired memory consolidation as tested by radial arm maze, and the expression of neuronal IEGs was downregulated in the hippocampus as revealed by qRT-PCR and Western blotting. Further, methylated DNA immunoprecipitation (MeDIP) analysis showed increase in DNA methylation, while chromatin immunoprecipitation (ChIP) revealed decrease in H3K9/14 acetylation at the promoter of neuronal IEGs. Taken together, the present study shows that increased DNA methylation and decreased histone acetylation at the promoter of neuronal IEGs are associated with decline in their expression and memory consolidation during scopolamine-induced amnesia. These findings suggest that the epigenetic regulation through altered DNA methylation and histone acetylation might be explored further to develop potential therapeutic interventions for amnesia.
Collapse
Affiliation(s)
- Sweta Srivas
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Institute of Sciences, Banaras Hindu University, Varanasi, 221 005, India
| | - Mahendra K Thakur
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Institute of Sciences, Banaras Hindu University, Varanasi, 221 005, India.
| |
Collapse
|
30
|
Guo SJ, Cui Y, Huang ZZ, Liu H, Zhang XQ, Jiang JX, Xin WJ. Orexin A-mediated AKT signaling in the dentate gyrus contributes to the acquisition, expression and reinstatement of morphine-induced conditioned place preference. Addict Biol 2016; 21:547-59. [PMID: 25757577 DOI: 10.1111/adb.12236] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Accumulating evidence indicates that the hippocampal dentate gyrus (DG), a critical brain region contributing to learning and memory, is involved in the addiction and relapse to abused drugs. Emerging studies also suggest the role of orexin signaling in the rewarding behavior induced by repeated exposure to opiates. In the present study, we investigated the dynamic adaptation of orexin signaling in the DG and its functional significance in the acquisition, expression, maintenance of and relapse to rewarding behavior induced by morphine. Repeated place conditioning with morphine significantly increased the orexin A content released from the lateral hypothalamic area projecting neurons into the DG. Local infusions of orexin A into the DG sensitized the acquisition of and relapse to the conditioned place preference induced by morphine. The application of the orexin receptor type 1 (OXR1) antagonist SB334867 significantly abolished the acquisition, expression and maintenance of the conditioned place preference induced by repeated exposure to morphine. Furthermore, the significant increase of the phosphorylation of AKT in the DG was associated with preference for the morphine-paired chamber in rats, which was reversed by the local administration of an OXR1 antagonist. Thus, these findings suggested that the dynamic upregulation of orexin A signaling, via the AKT pathway in the DG, may promote the acquisition and maintenance of opioid-induced craving behaviors and may increase sensitivity to the rewarding effect of subsequent opioids.
Collapse
Affiliation(s)
- Sui-Jun Guo
- Department of Psychology; Guangzhou Medical University; China
| | - Yu Cui
- Department of Physiology and Pain Research Center; Zhongshan Medical School; Sun Yat-Sen University; China
| | - Zhen-Zhen Huang
- Department of Physiology and Pain Research Center; Zhongshan Medical School; Sun Yat-Sen University; China
| | - Huan Liu
- Department of Physiology and Pain Research Center; Zhongshan Medical School; Sun Yat-Sen University; China
| | - Xue-Qin Zhang
- Department of Psychology; Guangzhou Medical University; China
| | - Jin-Xiang Jiang
- Department of Psychology; Guangzhou Medical University; China
| | - Wen-Jun Xin
- Department of Physiology and Pain Research Center; Zhongshan Medical School; Sun Yat-Sen University; China
| |
Collapse
|
31
|
Ferrari LL, Agostinelli LJ, Krashes MJ, Lowell BB, Scammell TE, Arrigoni E. Dynorphin inhibits basal forebrain cholinergic neurons by pre- and postsynaptic mechanisms. J Physiol 2016; 594:1069-85. [PMID: 26613645 DOI: 10.1113/jp271657] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/24/2015] [Indexed: 12/29/2022] Open
Abstract
KEY POINTS The basal forebrain is an important component of the ascending arousal system and may be a key site through which the orexin neurons promote arousal. It has long been known that orexin-A and -B excite basal forebrain cholinergic neurons, but orexin-producing neurons also make the inhibitory peptide dynorphin. Using whole-cell recordings in brain slices, we found that dynorphin-A directly inhibits basal forebrain cholinergic neurons via κ-opioid receptors, and decreases afferent excitatory synaptic input to these neurons. While the effects of dynorphin-A and orexin-A desensitize over multiple applications, co-application of dynorphin-A and orexin-A produces a sustained response that reverses depending on the membrane potential of basal forebrain cholinergic neurons. At -40 mV the net effect of the co-application is inhibition by dynorphin-A, whereas at -70 mV the excitatory response to orexin-A prevails. ABSTRACT The basal forebrain (BF) is an essential component of the ascending arousal systems and may be a key site through which the orexin (also known as hypocretin) neurons drive arousal and promote the maintenance of normal wakefulness. All orexin neurons also make dynorphin, and nearly all brain regions innervated by the orexin neurons express kappa opiate receptors, the main receptor for dynorphin. This is remarkable because orexin excites target neurons including BF neurons, but dynorphin has inhibitory effects. We identified the sources of dynorphin input to the magnocellular preoptic nucleus and substantia innominata (MCPO/SI) in mice and determined the effects of dynorphin-A on MCPO/SI cholinergic neurons using patch-clamp recordings in brain slices. We found that the orexin neurons are the main source of dynorphin input to the MCPO/SI region, and dynorphin-A inhibits MCPO/SI cholinergic neurons through κ-opioid receptors by (1) activation of a G protein-coupled inwardly rectifying potassium current, (2) inhibition of a voltage-gated Ca(2+) current and (3) presynaptic depression of the glutamatergic input to these neurons. The responses both to dynorphin-A and to orexin-A desensitize, but co-application of dynorphin-A and orexin-A produces a sustained response. In addition, the polarity of the response to the co-application depends on the membrane potential of BF neurons; at -40 mV the net effect of the co-application is inhibition by dynorphin-A, whereas at -70 mV the excitatory response to orexin-A prevails. This suggests that depending on their state of activation, BF cholinergic neurons can be excited or inhibited by signals from the orexin neurons.
Collapse
Affiliation(s)
- L L Ferrari
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - L J Agostinelli
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - M J Krashes
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, 20892-1453, USA
| | - B B Lowell
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - T E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - E Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| |
Collapse
|
32
|
Abstract
Initially implicated in the regulation of feeding, orexins/hypocretins are now acknowledged to play a major role in the control of a wide variety of biological processes, such as sleep, energy expenditure, pain, cardiovascular function and neuroendocrine regulation, a feature that makes them one of the most pleiotropic families of hypothalamic neuropeptides. While the orexigenic effect of orexins is well described, their central effects on energy expenditure and particularly on brown adipose tissue (BAT) thermogenesis are not totally unraveled. Better understanding of these actions and their possible interrelationship with other hypothalamic systems controlling thermogenesis, such as AMP-activated protein kinase (AMPK) and endoplasmic reticulum (ER) stress, will help to clarify the exact role and pathophysiological relevance of these neuropeptides have on energy balance.
Collapse
Affiliation(s)
- Johan Fernø
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; Department of Clinical Science, K. G. Jebsen Center for Diabetes Research, University of Bergen, N-5021 Bergen, Norway.
| | - Rosa Señarís
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) 15706, Spain
| | - Manuel Tena-Sempere
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) 15706, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica (IMIBIC)/Hospital Reina Sofía, 14004 Córdoba, Spain; FiDiPro Program, Department of Physiology, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) 15706, Spain.
| |
Collapse
|
33
|
Yelin-Bekerman L, Elbaz I, Diber A, Dahary D, Gibbs-Bar L, Alon S, Lerer-Goldshtein T, Appelbaum L. Hypocretin neuron-specific transcriptome profiling identifies the sleep modulator Kcnh4a. eLife 2015; 4:e08638. [PMID: 26426478 PMCID: PMC4718730 DOI: 10.7554/elife.08638] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 09/30/2015] [Indexed: 12/31/2022] Open
Abstract
Sleep has been conserved throughout evolution; however, the molecular and neuronal mechanisms of sleep are largely unknown. The hypothalamic hypocretin/orexin (Hcrt) neurons regulate sleep\wake states, feeding, stress, and reward. To elucidate the mechanism that enables these various functions and to identify sleep regulators, we combined fluorescence cell sorting and RNA-seq in hcrt:EGFP zebrafish. Dozens of Hcrt-neuron–specific transcripts were identified and comprehensive high-resolution imaging revealed gene-specific localization in all or subsets of Hcrt neurons. Clusters of Hcrt-neuron–specific genes are predicted to be regulated by shared transcription factors. These findings show that Hcrt neurons are heterogeneous and that integrative molecular mechanisms orchestrate their diverse functions. The voltage-gated potassium channel Kcnh4a, which is expressed in all Hcrt neurons, was silenced by the CRISPR-mediated gene inactivation system. The mutant kcnh4a (kcnh4a-/-) larvae showed reduced sleep time and consolidation, specifically during the night, suggesting that Kcnh4a regulates sleep. DOI:http://dx.doi.org/10.7554/eLife.08638.001 Sleep appears to be essential for all animals. The loss of a type of brain cell called the Hypocretin/Orexin (Hcrt) neurons causes the sleep disorder narcolepsy, which disturbs sleep patterns. These neurons also control several other fundamental behaviors and activities, including eating and processing rewards, but it is not clear how Hcrt neurons are able to influence multiple behaviors. The development and activity of a cell depends to a large extent on the genes it expresses. Yelin-Bekerman et al. have now used genetic techniques to identify a set of genes that are specifically expressed in the Hcrt neurons of zebrafish. Some of these genes are expressed in all of the Hcrt neurons, and some are only expressed in certain subsets of them. Computational methods also revealed a set of “transcription factor” proteins that regulate the expression of clusters of these genes. Yelin-Bekerman et al. focused on a gene called kcnh4a, and found that this encodes an ion channel protein that allows potassium ions to exit the neurons and stop neuronal activity (this activity is also known as an “action potential”). This gene is expressed in all Hcrt neurons. Further experiments showed that zebrafish that lack the potassium channel sleep less during the night. This therefore suggests that the potassium channel is important for regulating sleep. Future studies of the genes that are enriched in Hcrt neurons could uncover the mechanisms that enable the neurons to play a role in such a diverse range of processes, including feeding and sleep-wake cycles. These studies should enhance our understanding of the role of sleep and may help to develop treatments for metabolic and sleep disorders. DOI:http://dx.doi.org/10.7554/eLife.08638.002
Collapse
Affiliation(s)
- Laura Yelin-Bekerman
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.,The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Idan Elbaz
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.,The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Alex Diber
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.,The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | | | | | - Shahar Alon
- Media Lab, Massachusetts Institute of Technology, Cambridge, United states
| | - Tali Lerer-Goldshtein
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.,The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Lior Appelbaum
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.,The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| |
Collapse
|
34
|
Shan L, Dauvilliers Y, Siegel JM. Interactions of the histamine and hypocretin systems in CNS disorders. Nat Rev Neurol 2015; 11:401-13. [PMID: 26100750 DOI: 10.1038/nrneurol.2015.99] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Histamine and hypocretin neurons are localized to the hypothalamus, a brain area critical to autonomic function and sleep. Narcolepsy type 1, also known as narcolepsy with cataplexy, is a neurological disorder characterized by excessive daytime sleepiness, impaired night-time sleep, cataplexy, sleep paralysis and short latency to rapid eye movement (REM) sleep after sleep onset. In narcolepsy, 90% of hypocretin neurons are lost; in addition, two groups reported in 2014 that the number of histamine neurons is increased by 64% or more in human patients with narcolepsy, suggesting involvement of histamine in the aetiology of this disorder. Here, we review the role of the histamine and hypocretin systems in sleep-wake modulation. Furthermore, we summarize the neuropathological changes to these two systems in narcolepsy and discuss the possibility that narcolepsy-associated histamine abnormalities could mediate or result from the same processes that cause the hypocretin cell loss. We also review the changes in the hypocretin and histamine systems, and the associated sleep disruptions, in Parkinson disease, Alzheimer disease, Huntington disease and Tourette syndrome. Finally, we discuss novel therapeutic approaches for manipulation of the histamine system.
Collapse
Affiliation(s)
- Ling Shan
- Department of Psychiatry and Brain Research Institute, UCLA School of Medicine, Veterans' Affairs Greater Los Angeles Healthcare System (VA GLAHS), 16111 Plummer Street North Hills, 151A3, CA 91343, USA
| | - Yves Dauvilliers
- Centre de Référence Nationale Maladies Rares, Narcolepsie et Hypersomnie Idiopathique, Département de Neurologie, Hôpital Gui-de-Chauliac, INSERM U1061, 80 avenue Augustin Fliche, Montpellier 34295, France
| | - Jerome M Siegel
- Department of Psychiatry and Brain Research Institute, UCLA School of Medicine, Veterans' Affairs Greater Los Angeles Healthcare System (VA GLAHS), 16111 Plummer Street North Hills, 151A3, CA 91343, USA
| |
Collapse
|
35
|
|
36
|
Brown JA, Woodworth HL, Leinninger GM. To ingest or rest? Specialized roles of lateral hypothalamic area neurons in coordinating energy balance. Front Syst Neurosci 2015; 9:9. [PMID: 25741247 PMCID: PMC4332303 DOI: 10.3389/fnsys.2015.00009] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 01/15/2015] [Indexed: 12/26/2022] Open
Abstract
Survival depends on an organism’s ability to sense nutrient status and accordingly regulate intake and energy expenditure behaviors. Uncoupling of energy sensing and behavior, however, underlies energy balance disorders such as anorexia or obesity. The hypothalamus regulates energy balance, and in particular the lateral hypothalamic area (LHA) is poised to coordinate peripheral cues of energy status and behaviors that impact weight, such as drinking, locomotor behavior, arousal/sleep and autonomic output. There are several populations of LHA neurons that are defined by their neuropeptide content and contribute to energy balance. LHA neurons that express the neuropeptides melanin-concentrating hormone (MCH) or orexins/hypocretins (OX) are best characterized and these neurons play important roles in regulating ingestion, arousal, locomotor behavior and autonomic function via distinct neuronal circuits. Recently, another population of LHA neurons containing the neuropeptide Neurotensin (Nts) has been implicated in coordinating anorectic stimuli and behavior to regulate hydration and energy balance. Understanding the specific roles of MCH, OX and Nts neurons in harmonizing energy sensing and behavior thus has the potential to inform pharmacological strategies to modify behaviors and treat energy balance disorders.
Collapse
Affiliation(s)
- Juliette A Brown
- Department of Pharmacology and Toxicology, Michigan State University East Lansing, MI, USA ; Center for Integrative Toxicology East Lansing, MI, USA
| | | | - Gina M Leinninger
- Center for Integrative Toxicology East Lansing, MI, USA ; Department of Physiology, Michigan State University East Lansing, MI, USA
| |
Collapse
|
37
|
Elbaz I, Lerer-Goldshtein T, Okamoto H, Appelbaum L. Reduced synaptic density and deficient locomotor response in neuronal activity-regulated pentraxin 2a mutant zebrafish. FASEB J 2014; 29:1220-34. [PMID: 25466900 DOI: 10.1096/fj.14-258350] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 11/12/2014] [Indexed: 11/11/2022]
Abstract
Neuronal-activity-regulated pentraxin (NARP/NPTX2/NP2) is a secreted synaptic protein that regulates the trafficking of glutamate receptors and mediates learning, memory, and drug addiction. The role of NPTX2 in regulating structural synaptic plasticity and behavior in a developing vertebrate is indefinite. We characterized the expression of nptx2a in larvae and adult zebrafish and established a transcription activator-like effector nuclease (TALEN)-mediated nptx2a mutant (nptx2a(-/-)) to study the role of Nptx2a in regulating structural synaptic plasticity and behavior. Similar to mammals, the zebrafish nptx2a was expressed in excitatory neurons in the brain and spinal cord. Its expression was induced in response to a mechanosensory stimulus but did not change during day and night. Behavioral assays showed that loss of Nptx2a results in reduced locomotor response to light-to-dark transition states and to a sound stimulus. Live imaging of synapses using the transgenic nptx2a:GAL4VP16 zebrafish and a fluorescent presynaptic synaptophysin (SYP) marker revealed reduced synaptic density in the axons of the spinal motor neurons and the anterodorsal lateral-line ganglion (gAD), which regulate locomotor activity and locomotor response to mechanosensory stimuli, respectively. These results suggest that Nptx2a affects locomotor response to external stimuli by mediating structural synaptic plasticity in excitatory neuronal circuits.
Collapse
Affiliation(s)
- Idan Elbaz
- *The Mina & Everard Goodman Faculty of Life Sciences and The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel; and Laboratory for Developmental Gene Regulation, RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - Tali Lerer-Goldshtein
- *The Mina & Everard Goodman Faculty of Life Sciences and The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel; and Laboratory for Developmental Gene Regulation, RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - Hitoshi Okamoto
- *The Mina & Everard Goodman Faculty of Life Sciences and The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel; and Laboratory for Developmental Gene Regulation, RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - Lior Appelbaum
- *The Mina & Everard Goodman Faculty of Life Sciences and The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel; and Laboratory for Developmental Gene Regulation, RIKEN Brain Science Institute, Wako, Saitama, Japan
| |
Collapse
|
38
|
Li J, Hu Z, de Lecea L. The hypocretins/orexins: integrators of multiple physiological functions. Br J Pharmacol 2014; 171:332-50. [PMID: 24102345 DOI: 10.1111/bph.12415] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 07/16/2013] [Accepted: 08/02/2013] [Indexed: 12/28/2022] Open
Abstract
The hypocretins (Hcrts), also known as orexins, are two peptides derived from a single precursor produced in the posterior lateral hypothalamus. Over the past decade, the orexin system has been associated with numerous physiological functions, including sleep/arousal, energy homeostasis, endocrine, visceral functions and pathological states, such as narcolepsy and drug abuse. Here, we review the discovery of Hcrt/orexins and their receptors and propose a hypothesis as to how the orexin system orchestrates these multifaceted physiological functions.
Collapse
Affiliation(s)
- Jingcheng Li
- Department of Physiology, Third Military Medical University, Chongqing, China
| | | | | |
Collapse
|
39
|
Miskimon M, Han S, Lee JJ, Ringkamp M, Wilson MA, Petralia RS, Dong X, Worley PF, Baraban JM, Reti IM. Selective expression of Narp in primary nociceptive neurons: role in microglia/macrophage activation following nerve injury. J Neuroimmunol 2014; 274:86-95. [PMID: 25005116 DOI: 10.1016/j.jneuroim.2014.06.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 05/13/2014] [Accepted: 06/19/2014] [Indexed: 11/19/2022]
Abstract
Neuronal activity regulated pentraxin (Narp) is a secreted protein implicated in regulating synaptic plasticity via its association with the extracellular surface of AMPA receptors. We found robust Narp immunostaining in dorsal root ganglia (DRG) that is largely restricted to small diameter neurons, and in the superficial layers of the dorsal horn of the spinal cord. In double staining studies of DRG, we found that Narp is expressed in both IB4- and CGRP-positive neurons, markers of distinct populations of nociceptive neurons. Although a panel of standard pain behavioral assays were unaffected by Narp deletion, we found that Narp knockout mice displayed an exaggerated microglia/macrophage response in the dorsal horn of the spinal cord to sciatic nerve transection 3days after surgery compared with wild type mice. As other members of the pentraxin family have been implicated in regulating innate immunity, these findings suggest that Narp, and perhaps other neuronal pentraxins, also regulate inflammation in the nervous system.
Collapse
Affiliation(s)
- M Miskimon
- Department of Neuroscience, Johns Hopkins University, United States
| | - S Han
- Department of Psychiatry, Johns Hopkins University, United States
| | - J J Lee
- Department of Psychiatry, Johns Hopkins University, United States
| | - M Ringkamp
- Department of Neurosurgery, Johns Hopkins University, United States
| | - M A Wilson
- Department of Neuroscience, Johns Hopkins University, United States
| | - R S Petralia
- NIDCD, NIH, Johns Hopkins University, United States
| | - X Dong
- Department of Neuroscience, Johns Hopkins University, United States
| | - P F Worley
- Department of Neuroscience, Johns Hopkins University, United States
| | - J M Baraban
- Department of Neuroscience, Johns Hopkins University, United States; Department of Psychiatry, Johns Hopkins University, United States
| | - I M Reti
- Department of Neuroscience, Johns Hopkins University, United States; Department of Psychiatry, Johns Hopkins University, United States; Laboratory of Origin, United States.
| |
Collapse
|
40
|
Hoyer D, Jacobson LH. Orexin in sleep, addiction and more: is the perfect insomnia drug at hand? Neuropeptides 2013; 47:477-88. [PMID: 24215799 DOI: 10.1016/j.npep.2013.10.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 10/13/2013] [Accepted: 10/14/2013] [Indexed: 12/29/2022]
Abstract
Orexins A and B (hypocretins 1 and 2) and their two receptors (OX1R and OX2R) were discovered in 1998 by two different groups. Orexin A and B are derived from the differential processing of a common precursor, the prepro-orexin peptide. The neuropeptides are expressed in a few thousand cells located in the lateral hypothalamus (LH), but their projections and receptor distribution are widespread throughout the brain. Remarkably, prepro peptide and double (OX1R/OX2R) receptor knock out (KO) mice reproduce a sleep phenotype known in humans and dogs as narcolepsy/cataplexy. In humans, this disease is characterized by the absence of orexin producing cells in the LH, and severely depleted levels of orexin the cerebrospinal fluid. Null mutation of the individual OX1R or OX2R in mice substantially ameliorates the narcolepsy/cataplexy phenotype compared to the OX1R/OX2R KO, and highlights specific roles of the individual receptors in sleep architecture, the OX1R KO demonstrating an a attenuated sleep phenotype relative to the OX2R KO. It has therefore been suggested that orexin is a master regulator of the sleep-wake cycle, with high activity of the LH orexin cells during wake and almost none during sleep. Less than 10years later, the first orexin antagonist, almorexant, a dual orexin receptor antagonist (DORA), was reported to be effective in inducing sleep in volunteers and insomnia patients. Although development was stopped for almorexant and for Glaxo's DORA SB-649868, no less than 4 orexin receptor antagonists have reached phase II for insomnia, including Filorexant (MK-6096) and Suvorexant (MK-4305) from Merck. Suvorexant has since progressed to Phase III and dossier submission to the FDA. These four compounds are reported as DORAs, however, they equilibrate very slowly at one and/or the other orexin receptor, and thus at equilibrium may show more or less selectivity for OX1R or OX2R. The appropriate balance of antagonism of the two receptors for sleep is a point of debate, although in rodent models OX2R antagonism alone appears sufficient to induce sleep, whereas OX1R antagonism is largely devoid of this effect. Orexin is involved in a number of other functions including reward and feeding, where OX1R (possibly OX2R) antagonists display anti-addictive properties in rodent models of alcohol, smoking, and drug self-administration. However, despite early findings in feeding and appetite control, orexin receptor antagonists have not produced the anticipated effects in models of increased food intake or obesity in rodents, nor have they shown marked effects on weight in the existing clinical trials. The role of orexin in a number of other domains such as pain, mood, anxiety, migraine and neurodegenerative diseases is an active area of research. The progress of the orexin field is thus extraordinary, and the community awaits the clinical testing of more receptor selective antagonists in sleep and other disorders, as well as that of orexin agonists, with the latter expected to produce positive outcomes in narcolepsy/cataplexy and other conditions.
Collapse
Affiliation(s)
- Daniel Hoyer
- Department of Pharmacology & Therapeutics, School of Medicine, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville Campus, Kenneth Myer Building, at Genetics Lane, on Royal Parade, University of Melbourne, Parkville 3010, Australia.
| | | |
Collapse
|
41
|
Blouin AM, Lee JJ, Tao B, Smith DR, Johnson AW, Baraban JM, Reti IM. Narp knockout mice show normal reactivity to novelty but attenuated recovery from neophobia. Behav Brain Res 2013; 257:178-81. [PMID: 24120400 DOI: 10.1016/j.bbr.2013.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 10/02/2013] [Indexed: 10/26/2022]
Abstract
Narp knockout (KO) mice demonstrate cognitive inflexibility and addictive behavior, which are associated with abnormal reactivity to a novel stimulus. To assess reactivity to novelty, we tested Narp KO and wild-type (WT) mice on a neophobia procedure. Both Narp KO and WT mice showed a similar decrease in consumption upon initial exposure to a novel flavor, but Narp KO mice did not increase consumption with subsequent exposures to the novel flavor like the WT mice. Therefore, Narp KO mice do not have abnormal reactivity to novelty but show deficits in adapting behavior to reflect the updated value of a stimulus.
Collapse
Affiliation(s)
- Ashley M Blouin
- Department of Psychiatry and Behavioral Sciences Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Hypocretin/orexin neurons contribute to hippocampus-dependent social memory and synaptic plasticity in mice. J Neurosci 2013; 33:5275-84. [PMID: 23516292 DOI: 10.1523/jneurosci.3200-12.2013] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Hypocretin/orexin (Hcrt)-producing neurons in the lateral hypothalamus project throughout the brain, including to the hippocampus, where Hcrt receptors are widely expressed. Hcrt neurons activate these targets to orchestrate global arousal state, wake-sleep architecture, energy homeostasis, stress adaptation, and reward behaviors. Recently, Hcrt has been implicated in cognitive functions and social interaction. In the present study, we tested the hypothesis that Hcrt neurons are critical to social interaction, particularly social memory, using neurobehavioral assessment and electrophysiological approaches. The validated "two-enclosure homecage test" devices and procedure were used to test sociability, preference for social novelty (social novelty), and recognition memory. A conventional direct contact social test was conducted to corroborate the findings. We found that adult orexin/ataxin-3-transgenic (AT) mice, in which Hcrt neurons degenerate by 3 months of age, displayed normal sociability and social novelty with respect to their wild-type littermates. However, AT mice displayed deficits in long-term social memory. Nasal administration of exogenous Hcrt-1 restored social memory to an extent in AT mice. Hippocampal slices taken from AT mice exhibited decreases in degree of paired-pulse facilitation and magnitude of long-term potentiation, despite displaying normal basal synaptic neurotransmission in the CA1 area compared to wild-type hippocampal slices. AT hippocampi had lower levels of phosphorylated cAMP response element-binding protein (pCREB), an activity-dependent transcription factor important for synaptic plasticity and long-term memory storage. Our studies demonstrate that Hcrt neurons play an important role in the consolidation of social recognition memory, at least in part through enhancements of hippocampal synaptic plasticity and cAMP response element-binding protein phosphorylation.
Collapse
|
43
|
Furutani N, Hondo M, Kageyama H, Tsujino N, Mieda M, Yanagisawa M, Shioda S, Sakurai T. Neurotensin co-expressed in orexin-producing neurons in the lateral hypothalamus plays an important role in regulation of sleep/wakefulness states. PLoS One 2013; 8:e62391. [PMID: 23620827 PMCID: PMC3631195 DOI: 10.1371/journal.pone.0062391] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 03/24/2013] [Indexed: 02/03/2023] Open
Abstract
Both orexin and neurotensin are expressed in the lateral hypothalamic area (LHA) and have been implicated in the regulation of feeding, motor activity and the reward system. A double label immunofluorescence and in situ hybridization studies showed that neurotensin colocalizes with orexin in neurons of the LHA. Pharmacological studies suggested that neurotensin excites orexin-producing neurons (orexin neurons) through activation of neurotensin receptor-2 (NTSR-2) and non-selective cation channels. In situ hybridization study showed that most orexin neurons express neurotensin receptor-2 mRNA but not neurotensin receptor-1 (Ntsr-1) mRNA. Immunohistochemical studies showed that neurotensin-immunoreactive fibers make appositions to orexin neurons. A neurotensin receptor antagonist decreased Fos expression in orexin neurons and wakefulness time in wild type mice when administered intraperitoneally. However, the antagonist did not evoke any effect on these parameters in orexin neuron-ablated mice. These observations suggest the importance of neurotensin in maintaining activity of orexin neurons. The evidence presented here expands our understanding of the regulatory mechanism of orexin neurons.
Collapse
Affiliation(s)
- Naoki Furutani
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Mari Hondo
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa, Japan
- Center for Behavioral Molecular Genetics, University of Tsukuba, Tsukuba, Japan
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - Haruaki Kageyama
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Natsuko Tsujino
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Michihiro Mieda
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Masashi Yanagisawa
- Center for Behavioral Molecular Genetics, University of Tsukuba, Tsukuba, Japan
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Seiji Shioda
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Takeshi Sakurai
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa, Japan
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
- * E-mail:
| |
Collapse
|
44
|
Tsujino N, Sakurai T. Role of orexin in modulating arousal, feeding, and motivation. Front Behav Neurosci 2013; 7:28. [PMID: 23616752 PMCID: PMC3629303 DOI: 10.3389/fnbeh.2013.00028] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 03/25/2013] [Indexed: 01/15/2023] Open
Abstract
Orexin deficiency results in narcolepsy in humans, dogs, and rodents, suggesting that the orexin system is particularly important for maintenance of wakefulness. However, orexin neurons are “multi-tasking” neurons that regulate sleep/wake states as well as feeding behavior, emotion, and reward processes. Orexin deficiency causes abnormalities in energy homeostasis, stress-related behavior, and reward systems. Orexin excites waking-active monoaminergic and cholinergic neurons in the hypothalamus and brain stem regions to maintain a long, consolidated waking period. Orexin neurons also have reciprocal links with the hypothalamic nuclei, which regulates feeding. Moreover, the responsiveness of orexin neurons to peripheral metabolic cues suggests that these neurons have an important role as a link between energy homeostasis and vigilance states. The link between orexin and the ventral tegmental nucleus serves to motivate an animal to engage in goal-directed behavior. This review focuses on the interaction of orexin neurons with emotion, reward, and energy homeostasis systems. These connectivities are likely to be highly important to maintain proper vigilance states.
Collapse
Affiliation(s)
- Natsuko Tsujino
- Department of Molecular Neuroscience and Integrative Physiology, Graduate School of Medical Science, Kanazawa University Kanazawa, Japan
| | | |
Collapse
|
45
|
Translational profiling of hypocretin neurons identifies candidate molecules for sleep regulation. Genes Dev 2013; 27:565-78. [PMID: 23431030 DOI: 10.1101/gad.207654.112] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hypocretin (orexin; Hcrt)-containing neurons of the hypothalamus are essential for the normal regulation of sleep and wake behaviors and have been implicated in feeding, anxiety, depression, and reward. The absence of these neurons causes narcolepsy in humans and model organisms. However, little is known about the molecular phenotype of these cells; previous attempts at comprehensive profiling had only limited sensitivity or were inaccurate. We generated a Hcrt translating ribosome affinity purification (bacTRAP) line for comprehensive translational profiling of all ribosome-bound transcripts in these neurons in vivo. From this profile, we identified >6000 transcripts detectably expressed above background and 188 transcripts that are highly enriched in these neurons, including all known markers of the cells. Blinded analysis of in situ hybridization databases suggests that ~60% of these are expressed in a Hcrt marker-like pattern. Fifteen of these were confirmed with double labeling and microscopy, including the transcription factor Lhx9. Ablation of this gene results in a >30% loss specifically of Hcrt neurons, without a general disruption of hypothalamic development. Polysomnography and activity monitoring revealed a profound hypersomnolence in these mice. These data provide an in-depth and accurate profile of Hcrt neuron gene expression and suggest that Lhx9 may be important for specification or survival of a subset of these cells.
Collapse
|
46
|
Blouin AM, Han S, Pearce AM, Cheng K, Lee JJ, Johnson AW, Wang C, During MJ, Holland PC, Shaham Y, Baraban JM, Reti IM. Role of medial prefrontal cortex Narp in the extinction of morphine conditioned place preference. Learn Mem 2013; 20:75-9. [PMID: 23322555 DOI: 10.1101/lm.028621.112] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Narp knockout (KO) mice demonstrate an impaired extinction of morphine conditioned place preference (CPP). Because the medial prefrontal cortex (mPFC) has been implicated in extinction learning, we tested whether Narp cells in this region play a role in the extinction of morphine CPP. We found that intracranial injections of adenoassociated virus (AAV) expressing wild-type (WT) Narp into the mPFC of Narp KO mice rescued the extinction and the injection of AAV expressing a dominant negative form of Narp (NarpN) into the mPFC of WT mice impaired the extinction of morphine CPP. These findings suggest that Narp in the mPFC mediates the extinction of morphine CPP.
Collapse
Affiliation(s)
- Ashley M Blouin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Vital hypothalamic neurons regulating hunger, wakefulness, reward-seeking, and body weight are often defined by unique expression of hypothalamus-specific neuropeptides. Gene-ablation studies show that some of these peptides, notably orexin/hypocretin (hcrt/orx), are themselves critical for stable states of consciousness and metabolic health. However, neuron-ablation studies often reveal more severe phenotypes, suggesting key roles for co-expressed transmitters. Indeed, most hypothalamic neurons, including hcrt/orx cells, contain fast transmitters glutamate and GABA, as well as several neuropeptides. What are the roles and relations between different transmitters expressed by the same neuron? Here, we consider signaling codes for releasing different transmitters in relation to transmitter and receptor diversity in behaviorally defined, widely projecting “peptidergic” neurons, such as hcrt/orx cells. We then discuss latest optogenetic studies of endogenous transmitter release from defined sets of axons in situ, which suggest that recently characterized vital peptidergic neurons [e.g., hcrt/orx, proopiomelanocortin (POMC), and agouti-related peptide (AgRP) cells], as well as classical modulatory neurons (e.g., dopamine and acetylcholine cells), all use fast transmitters to control their postsynaptic targets. These optogenetic insights are complemented by recent observations of behavioral deficiencies caused by genetic ablation of fast transmission from specific neuropeptidergic and aminergic neurons. Powerful and fast (millisecond-scale) GABAergic and glutamatergic signaling from neurons previously considered to be primarily “modulatory” raises new questions about the roles of slower co-transmitters they co-express.
Collapse
|
48
|
Tsunematsu T, Yamanaka A. The Role of Orexin/Hypocretin in the Central Nervous System and Peripheral Tissues. SLEEP HORMONES 2012; 89:19-33. [DOI: 10.1016/b978-0-12-394623-2.00002-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
49
|
Leinninger GM. Lateral thinking about leptin: a review of leptin action via the lateral hypothalamus. Physiol Behav 2011; 104:572-81. [PMID: 21550356 DOI: 10.1016/j.physbeh.2011.04.060] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Revised: 04/19/2011] [Accepted: 04/28/2011] [Indexed: 12/30/2022]
Abstract
The lateral hypothalamic area (LHA) was initially described as a "feeding center" but we are now beginning to understand that the LHA contributes to other aspects of physiology as well. Indeed, the best-characterized neuronal populations of the LHA (which contain melanin-concentrating hormone (MCH) or the hypocretins/orexins (OX)) are not strictly orexigenic, but also have roles in regulation of the autonomic and sympathetic nervous systems as well as in modulating motivated behavior. Leptin is an anorectic hormone that regulates energy homeostasis and the mesolimbic DA system (which transduces the wanting of food, drugs of abuse, and sex) in part, via actions at the LHA. At least three populations of LHA neurons are regulated by leptin: those containing MCH, OX or the long form of the leptin receptor, LepRb. The emerging picture of leptin interaction with these LHA populations suggests that the LHA is not merely regulating feeding, but is a crucial integrator of energy balance and motivated behavior.
Collapse
Affiliation(s)
- Gina M Leinninger
- Division of Metabolism, Endocrinology and Diabetes, Department of Medicine, University of Michigan Medical School, 100 Wall Street, Ann Arbor, MI 48105, USA
| |
Collapse
|
50
|
Tanaka S, Kodama T, Nonaka T, Toyoda H, Arai M, Fukazawa M, Honda Y, Honda M, Mignot E. Transcriptional regulation of the hypocretin/orexin gene by NR6A1. Biochem Biophys Res Commun 2010; 403:178-83. [PMID: 21056546 PMCID: PMC3012206 DOI: 10.1016/j.bbrc.2010.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 11/01/2010] [Indexed: 12/30/2022]
Abstract
The hypocretin (also known as orexin) neuropeptide system coordinates the regulation of various physiological processes. A reduction in Nr6a1 expression was observed in hypocretin neuron-ablated transgenic mice. To show that prepro-hypocretin transcription is functionally modulated by NR6A1, we performed chromatin immunoprecipitation (ChIP) analysis, double-immunostaining, a luciferase reporter assay, and an in utero electroporation study. ChIP analysis showed that endogenous NR6A1 binds to a putative NR6A1-binding site. Double-immunostaining indicated almost all hypocretin neurons were positive for NR6A1 immunoreactivity. NR6A1 overexpression in SH-SY5Y cells modulated hypocretin promoter activity, an effect that was countered by lacking a putative NR6A1-binding site. Electroporation with Nr6a1 in the foetal hypothalamus promoted hypocretin transcription as compared to GFP-electroporation. These experiments confirmed that NR6A1 works as a regulator for hypocretin transcription.
Collapse
Affiliation(s)
- Susumu Tanaka
- Department of Sleep Disorders Research, Tokyo Institute of Psychiatry, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|