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
|
McGregor R, Wu MF, Thannickal TC, Li S, Siegel JM. Opioid-induced neuroanatomical, microglial and behavioral changes are blocked by suvorexant without diminishing opioid analgesia. NATURE. MENTAL HEALTH 2024; 2:1018-1031. [PMID: 39989723 PMCID: PMC11845277 DOI: 10.1038/s44220-024-00278-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 05/31/2024] [Indexed: 02/25/2025]
Abstract
Heroin use disorder in humans and chronic opioid administration to mice result in an increase in the number and a decrease in the size of detected hypocretin (Hcrt, or orexin) neurons. Chronic morphine administration to mice increases Hcrt axonal projections to the ventral tegmental area (VTA), the level of tyrosine hydroxylase (TH) in VTA and the number of detected TH+ cells in VTA, and activates VTA and hypothalamic microglia. Co-administration of morphine with the dual Hcrt receptor antagonist suvorexant prevents morphine-induced changes in the number and size of Hcrt neurons, the increase in Hcrt projections to the VTA and microglial activation in the VTA and hypothalamus. Co-administration of suvorexant with morphine also prevents morphine anticipatory behavior and reduces opioid withdrawal symptoms. However, suvorexant does not diminish morphine analgesia. Here we show that combined administration of opioids and suvorexant may reduce the addiction potential of opioid use for pain relief in humans while maintaining the analgesic effects of opioids.
Collapse
Affiliation(s)
- Ronald McGregor
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- These authors contributed equally: Ronald McGregor, Ming-Fung Wu
| | - Ming-Fung Wu
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- These authors contributed equally: Ronald McGregor, Ming-Fung Wu
| | - Thomas C. Thannickal
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Songlin Li
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Jerome M. Siegel
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
4
|
Zhang X, Perry RJ. Metabolic underpinnings of cancer-related fatigue. Am J Physiol Endocrinol Metab 2024; 326:E290-E307. [PMID: 38294698 PMCID: PMC11901342 DOI: 10.1152/ajpendo.00378.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/01/2024]
Abstract
Cancer-related fatigue (CRF) is one of the most prevalent and detrimental complications of cancer. Emerging evidence suggests that obesity and insulin resistance are associated with CRF occurrence and severity in cancer patients and survivors. In this narrative review, we analyzed recent studies including both preclinical and clinical research on the relationship between obesity and/or insulin resistance and CRF. We also describe potential mechanisms for these relationships, though with the caveat that because the mechanisms underlying CRF are incompletely understood, the mechanisms mediating the association between obesity/insulin resistance and CRF are similarly incompletely delineated. The data suggest that, in addition to their effects to worsen CRF by directly promoting tumor growth and metastasis, obesity and insulin resistance may also contribute to CRF by inducing chronic inflammation, neuroendocrinological disturbance, and metabolic alterations. Furthermore, studies suggest that patients with obesity and insulin resistance experience more cancer-induced pain and are at more risk of emotional and behavioral disruptions correlated with CRF. However, other studies implied a potentially paradoxical impact of obesity and insulin resistance to reduce CRF symptoms. Despite the need for further investigation utilizing interventions to directly elucidate the mechanisms of cancer-related fatigue, current evidence demonstrates a correlation between obesity and/or insulin resistance and CRF, and suggests potential therapeutics for CRF by targeting obesity and/or obesity-related mediators.
Collapse
Affiliation(s)
- Xinyi Zhang
- Departments of Cellular & Molecular Physiology and Medicine (Endocrinology), Yale University School of Medicine, New Haven, Connecticut, United States
| | - Rachel J Perry
- Departments of Cellular & Molecular Physiology and Medicine (Endocrinology), Yale University School of Medicine, New Haven, Connecticut, United States
| |
Collapse
|
5
|
Valizadeh P, Momtazmanesh S, Plazzi G, Rezaei N. Connecting the dots: An updated review of the role of autoimmunity in narcolepsy and emerging immunotherapeutic approaches. Sleep Med 2024; 113:378-396. [PMID: 38128432 DOI: 10.1016/j.sleep.2023.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Narcolepsy type 1 (NT1) is a chronic disorder characterized by pathological daytime sleepiness and cataplexy due to the disappearance of orexin immunoreactive neurons in the hypothalamus. Genetic and environmental factors point towards a potential role for inflammation and autoimmunity in the pathogenesis of the disease. This study aims to comprehensively review the latest evidence on the autoinflammatory mechanisms and immunomodulatory treatments aimed at suspected autoimmune pathways in NT1. METHODS Recent relevant literature in the field of narcolepsy, its autoimmune hypothesis, and purposed immunomodulatory treatments were reviewed. RESULTS Narcolepsy is strongly linked to specific HLA alleles and T-cell receptor polymorphisms. Furthermore, animal studies and autopsies have found infiltration of T cells in the hypothalamus, supporting T cell-mediated immunity. However, the role of autoantibodies has yet to be definitively established. Increased risk of NT1 after H1N1 infection and vaccination supports the autoimmune hypothesis, and the potential role of coronavirus disease 2019 and vaccination in triggering autoimmune neurodegeneration is a recent finding. Alterations in cytokine levels, gut microbiota, and microglial activation indicate a potential role for inflammation in the disease's development. Reports of using immunotherapies in NT1 patients are limited and inconsistent. Early treatment with IVIg, corticosteroids, plasmapheresis, and monoclonal antibodies has seldomly shown some potential benefits in some studies. CONCLUSION The current body of literature supports that narcolepsy is an autoimmune disorder most likely caused by T-cell involvement. However, the potential for immunomodulatory treatments to reverse the autoinflammatory process remains understudied. Further clinical controlled trials may provide valuable insights into this area.
Collapse
Affiliation(s)
- Parya Valizadeh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sara Momtazmanesh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Giuseppe Plazzi
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy; Department of Biomedical, Metabolic, and Neural Sciences, Università Degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Rong J, Yamasaki T, Li Y, Kumata K, Zhao C, Haider A, Chen J, Xiao Z, Fujinaga M, Hu K, Mori W, Zhang Y, Xie L, Zhou X, Collier TL, Zhang MR, Liang S. Development of Novel 11C-Labeled Selective Orexin-2 Receptor Radioligands for Positron Emission Tomography Imaging. ACS Med Chem Lett 2023; 14:1419-1426. [PMID: 37849554 PMCID: PMC10577698 DOI: 10.1021/acsmedchemlett.3c00320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023] Open
Abstract
Orexin 2 receptors (OX2R) represent a vital subtype of orexin receptors intricately involved in the regulation of wakefulness, arousal, and sleep-wake cycles. Despite their importance, there are currently no positron emission tomography (PET) tracers available for imaging the OX2R in vivo. Herein, we report [11C]1 ([11C]OX2-2201) and [11C]2 ([11C]OX2-2202) as novel PET ligands. Both compounds 1 (K i = 3.6 nM) and 2 (K i = 2.2 nM) have excellent binding affinity activities toward OX2R and target selectivity (OX2/OX1 > 600 folds). In vitro autoradiography in the rat brain suggested good to excellent in vitro binding specificity for [11C]1 and [11C]2. PET imaging in rat brains indicated that the low brain uptake of [11C]2 may be due to P-glycoprotein and/or breast cancer resistance protein efflux interaction and/or low passive permeability. Continuous effort in medicinal chemistry optimization is necessary to improve the brain permeability of this scaffold.
Collapse
Affiliation(s)
- Jian Rong
- Department of Radiology and Imaging Sciences,
Emory University, Atlanta, Georgia 30322, United
States
- Division of Nuclear Medicine and Molecular Imaging,
Massachusetts General Hospital and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United
States
| | - Tomoteru Yamasaki
- Department of Advanced Nuclear Medicine Sciences,
Institute for Quantum Medical Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Yinlong Li
- Department of Radiology and Imaging Sciences,
Emory University, Atlanta, Georgia 30322, United
States
- Division of Nuclear Medicine and Molecular Imaging,
Massachusetts General Hospital and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United
States
| | - Katsushi Kumata
- Department of Advanced Nuclear Medicine Sciences,
Institute for Quantum Medical Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Chunyu Zhao
- Department of Radiology and Imaging Sciences,
Emory University, Atlanta, Georgia 30322, United
States
- Division of Nuclear Medicine and Molecular Imaging,
Massachusetts General Hospital and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United
States
| | - Achi Haider
- Department of Radiology and Imaging Sciences,
Emory University, Atlanta, Georgia 30322, United
States
- Division of Nuclear Medicine and Molecular Imaging,
Massachusetts General Hospital and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United
States
| | - Jiahui Chen
- Department of Radiology and Imaging Sciences,
Emory University, Atlanta, Georgia 30322, United
States
- Division of Nuclear Medicine and Molecular Imaging,
Massachusetts General Hospital and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United
States
| | - Zhiwei Xiao
- Department of Radiology and Imaging Sciences,
Emory University, Atlanta, Georgia 30322, United
States
- Division of Nuclear Medicine and Molecular Imaging,
Massachusetts General Hospital and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United
States
| | - Masayuki Fujinaga
- Department of Advanced Nuclear Medicine Sciences,
Institute for Quantum Medical Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Kuan Hu
- Department of Advanced Nuclear Medicine Sciences,
Institute for Quantum Medical Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Wakana Mori
- Department of Advanced Nuclear Medicine Sciences,
Institute for Quantum Medical Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences,
Institute for Quantum Medical Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences,
Institute for Quantum Medical Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Xin Zhou
- Department of Radiology and Imaging Sciences,
Emory University, Atlanta, Georgia 30322, United
States
| | - Thomas L. Collier
- Department of Radiology and Imaging Sciences,
Emory University, Atlanta, Georgia 30322, United
States
- Division of Nuclear Medicine and Molecular Imaging,
Massachusetts General Hospital and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United
States
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences,
Institute for Quantum Medical Sciences, National Institutes for Quantum
Science and Technology, Chiba 263-8555, Japan
| | - Steven Liang
- Department of Radiology and Imaging Sciences,
Emory University, Atlanta, Georgia 30322, United
States
- Division of Nuclear Medicine and Molecular Imaging,
Massachusetts General Hospital and Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United
States
| |
Collapse
|
7
|
Shan L, Linssen S, Harteman Z, den Dekker F, Shuker L, Balesar R, Breesuwsma N, Anink J, Zhou J, Lammers GJ, Swaab DF, Fronczek R. Activated Wake Systems in Narcolepsy Type 1. Ann Neurol 2023; 94:762-771. [PMID: 37395722 DOI: 10.1002/ana.26736] [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: 02/06/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
OBJECTIVE Narcolepsy type 1 (NT1) is assumed to be caused solely by a lack of hypocretin (orexin) neurotransmission. Recently, however, we found an 88% reduction in corticotropin-releasing hormone (CRH)-positive neurons in the paraventricular nucleus (PVN). We assessed the remaining CRH neurons in NT1 to determine whether they co-express vasopressin (AVP) to reflect upregulation. We also systematically assessed other wake-systems, since current NT1 treatments target histamine, dopamine, and norepinephrine pathways. METHODS In postmortem tissue of people with NT1 and matched controls, we immunohistochemically stained and quantified neuronal populations expressing: CRH and AVP in the PVN, and CRH in the Barrington nucleus; the key neuronal histamine-synthesizing enzyme, histidine decarboxylase (HDC) in the hypothalamic tuberomammillary nucleus (TMN); the rate-limited-synthesizing enzyme, tyrosine hydroxylase (TH), for dopamine in the mid-brain and for norepinephrine in the locus coeruleus (LC). RESULTS In NT1, there was: a 234% increase in the percentage of CRH cells co-expressing AVP, while there was an unchanged integrated optical density of CRH staining in the Barrington nucleus; a 36% increased number of histamine neurons expressing HDC, while the number of typical human TMN neuronal profiles was unchanged; a tendency toward an increased density of TH-positive neurons in the substantia nigra compacta; while the density of TH-positive LC neurons was unchanged. INTERPRETATION Our findings suggest an upregulation of activity by histamine neurons and remaining CRH neurons in NT1. This may explain earlier reports of normal basal plasma cortisol levels but lower levels after dexamethasone suppression. Alternatively, CRH neurons co-expressing AVP neurons are less vulnerable. ANN NEUROL 2023;94:762-771.
Collapse
Affiliation(s)
- Ling Shan
- Leiden University Medical Centre, Department of Neurology, Leiden, The Netherlands, and Sleep Wake Centre SEIN, Heemstede, The Netherlands
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Suzan Linssen
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Zoe Harteman
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Fleur den Dekker
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Lamis Shuker
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Rawien Balesar
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Nicole Breesuwsma
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Jasper Anink
- Department of (Neuro) Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Jingru Zhou
- Leiden University Medical Centre, Department of Neurology, Leiden, The Netherlands, and Sleep Wake Centre SEIN, Heemstede, The Netherlands
| | - Gert Jan Lammers
- Leiden University Medical Centre, Department of Neurology, Leiden, The Netherlands, and Sleep Wake Centre SEIN, Heemstede, The Netherlands
| | - Dick F Swaab
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Rolf Fronczek
- Leiden University Medical Centre, Department of Neurology, Leiden, The Netherlands, and Sleep Wake Centre SEIN, Heemstede, The Netherlands
| |
Collapse
|
8
|
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
|
9
|
Piilgaard L, Rose L, Justinussen JL, Hviid CG, Lemcke R, Wellendorph P, Kornum BR. Non-invasive detection of narcolepsy type I phenotypical features and disease progression by continuous home-cage monitoring of activity in two mouse models: the HCRT-KO and DTA model. Sleep 2023; 46:zsad144. [PMID: 37210587 DOI: 10.1093/sleep/zsad144] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/11/2023] [Indexed: 05/22/2023] Open
Abstract
Narcolepsy type 1 (NT1) is a neurological disorder caused by disruption of hypocretin (HCRT; or orexin) neurotransmission leading to fragmented sleep/wake states, excessive daytime sleepiness, and cataplexy (abrupt muscle atonia during wakefulness). Electroencephalography and electromyography (EEG/EMG) monitoring is the gold standard to assess NT1 phenotypical features in both humans and mice. Here, we evaluated the digital ventilated home-cage (DVC®) activity system as an alternative to detect NT1 features in two NT1 mouse models: the genetic HCRT-knockout (-KO) model, and the inducible HCRT neuron-ablation hcrt-tTA;TetO-DTA (DTA) model, including both sexes. NT1 mice exhibited an altered dark phase activity profile and increased state transitions, compared to the wild-type (WT) phenotype. An inability to sustain activity periods >40 min represented a robust activity-based NT1 biomarker. These features were observable within the first weeks of HCRT neuron degeneration in DTA mice. We also created a nest-identification algorithm to differentiate between inactivity and activity, inside and outside the nest as a sleep and wake proxy, respectively, showing significant correlations with EEG/EMG-assessed sleep/wake behavior. Lastly, we tested the sensitivity of the activity system to detect behavioral changes in response to interventions such as repeated saline injection and chocolate. Surprisingly, daily consecutive saline injections significantly reduced activity and increased nest time of HCRT-WT mice. Chocolate increased total activity in all mice, and increased the frequency of short out-of-nest inactivity episodes in HCRT-KO mice. We conclude that the DVC® system provides a useful tool for non-invasive monitoring of NT1 phenotypical features, and has the potential to monitor drug effects in NT1 mice.
Collapse
Affiliation(s)
- Louise Piilgaard
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Laura Rose
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jessica L Justinussen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Camille Gylling Hviid
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - René Lemcke
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Petrine Wellendorph
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Rahbek Kornum
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Peleg-Raibstein D, Viskaitis P, Burdakov D. Eat, seek, rest? An orexin/hypocretin perspective. J Neuroendocrinol 2023; 35:e13259. [PMID: 36994677 DOI: 10.1111/jne.13259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023]
Abstract
Seeking and ingesting nutrients is an essential cycle of life in all species. In classical neuropsychology these two behaviours are viewed as fundamentally distinct from each other, and known as appetitive and consummatory, respectively. Appetitive behaviour is highly flexible and diverse, but typically involves increased locomotion and spatial exploration. Consummatory behaviour, in contrast, typically requires reduced locomotion. Another long-standing concept is "rest and digest", a hypolocomotive response to calorie intake, thought to facilitate digestion and storage of energy after eating. Here, we note that the classical seek➔ingest➔rest behavioural sequence is not evolutionarily advantageous for all ingested nutrients. Our limited stomach capacity should be invested wisely, rather than spent on the first available nutrient. This is because nutrients are not simply calories: some nutrients are more essential for survival than others. Thus, a key choice that needs to be made soon after ingestion: to eat more and rest, or to terminate eating and search for better food. We offer a perspective on recent work suggesting how nutrient-specific neural responses shape this choice. Specifically, the hypothalamic hypocretin/orexin neurons (HONs) - cells that promote hyperlocomotive explorative behaviours - are rapidly and differentially modulated by different ingested macronutrients. Dietary non-essential (but not essential) amino acids activate HONs, while glucose depresses HONs. This nutrient-specific HON modulation engages distinct reflex arcs, seek➔ingest➔seek and seek➔ingest➔rest, respectively. We propose that these nutri-neural reflexes evolved to facilitate optimal nutrition despite the limitations of our body.
Collapse
Affiliation(s)
- Daria Peleg-Raibstein
- ETH Zürich, Department of Health Sciences and Technology, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland
| | - Paulius Viskaitis
- ETH Zürich, Department of Health Sciences and Technology, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland
| | - Denis Burdakov
- ETH Zürich, Department of Health Sciences and Technology, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland
| |
Collapse
|
11
|
Repeated measures of hypocretin-1 in Danish and Italian patients with narcolepsy and in controls. Sleep Med 2023; 101:213-220. [PMID: 36427467 DOI: 10.1016/j.sleep.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 09/15/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
STUDY OBJECTIVES The assay currently used worldwide to measure cerebrospinal fluid hypocretin-1 (CSF-hcrt-1) for diagnosing narcolepsy uses a competitive radioimmunoassay with polyclonal anti-hcrt-1 antibodies. This assay detects multiple hypocretin-1 immunoreactive species in the CSF that are all derived from full-length hcrt-1. We aimed to revalidate CSF-hcrt-1 cut-offs for narcolepsy type 1 (NT1) diagnosis and to evaluate temporal changes in CSF-hcrt-1 levels in patients suspected of having central hypersomnia. METHOD We carried out a repeat lumbar puncture with a mean follow-up of 4.0 years, to measure CSF-hcrt-1 in patients suspected of having central hypersomnia in a follow-up study. Data from CSF samples of patients with NT1 and of controls without known hypersomnia, from the Italian-Stanford and Danish populations, were examined using a receiver-operating characteristic analysis. RESULTS The optimal CSF-hcrt-1 cut-offs for identifying NT1 were 129 pg/ml and 179 pg/ml for the Italian-Stanford and Danish populations, respectively. The sensitivity was 0.93-0.99 and the specificity was 1. Follow-up lumbar puncture measurements of CSF-hcrt-1 were obtained from 73 patients. 30 of 32 patients with low CSF-hcrt-1 levels continued to be categorized as low, with an unaltered diagnosis; two patients showed a marked increase in CSF-hcrt-1, attaining normal values at follow-up. One of these patients relapsed to low CSF-hcrt-1 after follow-up. All 41 patients with normal CSF-hcrt-1 at baseline had normal CSF-hcrt-1 at follow-up. CONCLUSION CSF-hcrt-1 measurement can provide an accurate test for diagnosing NT1, although it is important to validate the CSF-hcrt-1 cut-off for specific testing locations. Stable CSF-hcrt-1 levels support the already established prognosis of narcolepsy as permanent once the disorder has fully developed.
Collapse
|
12
|
Precocious puberty in narcolepsy type 1: Orexin loss and/or neuroinflammation, which is to blame? Sleep Med Rev 2022; 65:101683. [PMID: 36096986 DOI: 10.1016/j.smrv.2022.101683] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 10/14/2022]
Abstract
Narcolepsy type 1 (NT1) is a rare neurological sleep disorder triggered by postnatal loss of the orexin/hypocretin neuropeptides. Overweight/obesity and precocious puberty are highly prevalent comorbidities of NT1, with a close temporal correlation with disease onset, suggesting a common origin. However, the underlying mechanisms remain unknown and merit further investigation. The main question we address in this review is whether the occurrence of precocious puberty in NT1 is due to the lack of orexin/hypocretin or rather to a wider hypothalamic dysfunction in the context of neuroinflammation, which is likely to accompany the disease given its autoimmune origins. Our analysis suggests that the suspected generalized neuroinflammation of the hypothalamus in NT1 would tend to delay puberty rather than hastening it. In contrast, that the brutal loss of orexin/hypocretin would favor an early reactivation of gonadotropin-releasing hormone (GnRH) secretion during the prepubertal period in vulnerable children, leading to early puberty onset. Orexin/hypocretin replacement could thus be envisaged as a potential treatment for precocious puberty in NT1. Additionally, we put forward an alternative hypothesis regarding the concomitant occurrence of sleepiness, weight gain and early puberty in NT1.
Collapse
|
13
|
Scammell TE, Saper CB. Major advances in sleep neurology: 2002–22. Lancet Neurol 2022; 21:678-680. [DOI: 10.1016/s1474-4422(22)00263-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 11/24/2022]
|
14
|
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
|
15
|
Eghtesad M, Elahdadi Salmani M, Lashkarbolouki T, Goudarzi I. Lateral Hypothalamus Corticotropin-releasing Hormone Receptor-1 Inhibition and Modulating Stress-induced Anxiety Behavior. Basic Clin Neurosci 2022; 13:373-384. [PMID: 36457881 PMCID: PMC9706292 DOI: 10.32598/bcn.2021.445.3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/15/2020] [Accepted: 10/12/2020] [Indexed: 10/28/2024] Open
Abstract
INTRODUCTION Stress is a reaction to unwanted events disturbing body homeostasis and its pathways and target areas. Stress affects the brain through the lateral hypothalamic area (LHA), the orexinergic system that mediates the effect of corticotropin-releasing hormone (CRH) through CRH Receptor Type 1 (CRHr1). Therefore, this study explores the outcome of stress exposure on anxiety development and the involvement of the LHA through LHA-CRHr1. METHODS Male Wistar rats (220-250 g) implanted with a cannula on either side of the LHA received acute or chronic stress. Subsequently, exploratory behavior was examined using the Open Field (OF), and anxiety was tested by Elevated Plus Maze (EPM). Before sacrifice, the cerebrospinal fluid (CSF) and the blood were sampled. Nissl stain was performed on fixed brain tissues. RESULTS Acute stress reduced exploration in of and increased anxiety in EPM. LHA-CRHr1 inhibition reversed the variables to increase the exploration and decrease anxiety. In contrast, chronic stress did not show any effect on anxiety-related behaviors. Chronic stress decreased the cell population in the LHA, which was prevented by the CRHr1 inhibition. However, the CRHr1 inhibition could not reverse the chronic stress-induced increase in the CSF orexin level. Furthermore, plasma corticosterone levels increased through acute or chronic stress, impeded by the inhibition of CRHr1. CONCLUSION Our results recognize LHA-CRHr1 as a capable candidate that modulates acute stress-induced anxiety development and chronic stress-induced changes in the cellular population of the region. HIGHLIGHTS Acute stress, increased immobility of the rat in open field and elevated plus maze.Chronic stress, increased orexin production while decreasing neuronal survival.The anxiety and immobility were not developed in presence of CRHr1.CRHr1 blocking reversed the chronic stress changes in corticosterone and orexin. PLAIN LANGUAGE SUMMARY Lateral Hypothalamus (LH) is a region involved in sleep and appetite regulation and recently known to play role in stress pathophysiology. The stress mediating function of the LH is performed through Corticotropin Releasing Hormone Receptor type-1 (CRHr1). This study explored the role of LH- CRHr1 in anxiety development and orexin production. Acute and chronic stress affected the behavior and molecular changes, differently. The acute stress increased the anxiety condition, while the chronic stress could only change the molecular criteria. Although we assumed that the inability of the chronic stress to develop anxiety may be attributable to habituation, the chronic stress could increase the plasma corticosterone and orexin level. All of the stress mal-changes in behavior and molecular level prevented by antagonising CRHr1 in the LH, indicating a gating function of LH-CRHr1 for stress development.
Collapse
Affiliation(s)
- Masoumeh Eghtesad
- Department of Animal Biology, School of Biology, Damghan University, Damghan, Iran
| | | | - Taghi Lashkarbolouki
- Department of Animal Biology, School of Biology, Damghan University, Damghan, Iran
| | - Iran Goudarzi
- Department of Animal Biology, School of Biology, Damghan University, Damghan, Iran
| |
Collapse
|
16
|
Gómez de San José N, Massa F, Halbgebauer S, Oeckl P, Steinacker P, Otto M. Neuronal pentraxins as biomarkers of synaptic activity: from physiological functions to pathological changes in neurodegeneration. J Neural Transm (Vienna) 2022; 129:207-230. [PMID: 34460014 PMCID: PMC8866268 DOI: 10.1007/s00702-021-02411-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 08/17/2021] [Indexed: 12/22/2022]
Abstract
The diagnosis of neurodegenerative disorders is often challenging due to the lack of diagnostic tools, comorbidities and shared pathological manifestations. Synaptic dysfunction is an early pathological event in many neurodegenerative disorders, but the underpinning mechanisms are still poorly characterised. Reliable quantification of synaptic damage is crucial to understand the pathophysiology of neurodegeneration, to track disease status and to obtain prognostic information. Neuronal pentraxins (NPTXs) are extracellular scaffolding proteins emerging as potential biomarkers of synaptic dysfunction in neurodegeneration. They are a family of proteins involved in homeostatic synaptic plasticity by recruiting post-synaptic receptors into synapses. Recent research investigates the dynamic changes of NPTXs in the cerebrospinal fluid (CSF) as an expression of synaptic damage, possibly related to cognitive impairment. In this review, we summarise the available data on NPTXs structure and expression patterns as well as on their contribution in synaptic function and plasticity and other less well-characterised roles. Moreover, we propose a mechanism for their involvement in synaptic damage and neurodegeneration and assess their potential as CSF biomarkers for neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Federico Massa
- Department of Neurology, University of Ulm, Ulm, Germany
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | | | - Patrick Oeckl
- Department of Neurology, University of Ulm, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE E.V.), Ulm, Germany
| | | | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany.
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle (Saale), Germany.
| |
Collapse
|
17
|
Cervantes González A, Belbin O. Fluid markers of synapse degeneration in synucleinopathies. J Neural Transm (Vienna) 2022; 129:187-206. [PMID: 35147800 DOI: 10.1007/s00702-022-02467-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/20/2022] [Indexed: 01/06/2023]
Abstract
The abnormal accumulation of α-synuclein in the brain is a common feature of Parkinson's disease (PD), PD dementia (PDD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA), and synucleinopathies that present with overlapping but distinct clinical symptoms that include motor and cognitive deficits. Synapse degeneration is the crucial neuropathological event in these synucleinopathies and the neuropathological correlate of connectome dysfunction. The cognitive and motor deficits resulting from the connectome dysfunction are currently measured by scalar systems that are limited in their sensitivity and largely subjective. Ideally, a marker of synapse degeneration would correlate with measures of cognitive or motor impairment, and could therefore be used as a more objective, surrogate biomarker of the core clinical features of these diseases. Furthermore, an objective surrogate biomarker that can detect and monitor the progression of synapse degeneration would improve patient management and clinical trial design, and could provide a measure of therapeutic response. Here, we review the published findings relating to candidate biomarkers of synapse degeneration in PD, PDD, DLB, and MSA patient-derived biofluids and discuss the findings in the context of the mechanisms associated with α-synuclein-mediated synapse degeneration. Understanding these mechanisms is essential not only for discovery of biomarkers, but also to improve our understanding of the earliest changes in disease pathogenesis of synucleinopathies.
Collapse
Affiliation(s)
- Alba Cervantes González
- Neurology Department, Biomedical Research Institute Sant Pau (IIB Sant Pau) and Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Olivia Belbin
- Neurology Department, Biomedical Research Institute Sant Pau (IIB Sant Pau) and Sant Pau Memory Unit, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.
| |
Collapse
|
18
|
Shan L, Balesar R, Swaab DF, Lammers GJ, Fronczek R. Reduced numbers of corticotropin‐releasing hormone neurons in narcolepsy type 1. Ann Neurol 2022; 91:282-288. [PMID: 34981555 PMCID: PMC9306683 DOI: 10.1002/ana.26300] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/31/2021] [Accepted: 12/31/2021] [Indexed: 11/14/2022]
Abstract
Narcolepsy type 1 (NT1) is a chronic sleep disorder correlated with loss of hypocretin(orexin). In NT1 post‐mortem brains, we observed 88% reduction in corticotropin‐releasing hormone (CRH)‐positive neurons in the paraventricular nucleus (PVN) and significantly less CRH‐positive fibers in the median eminence, whereas CRH‐neurons in the locus coeruleus and thalamus, and other PVN neuronal populations were spared: that is, vasopressin, oxytocin, tyrosine hydroxylase, and thyrotropin releasing hormone‐expressing neurons. Other hypothalamic cell groups, that is, the suprachiasmatic, ventrolateral preoptic, infundibular, and supraoptic nuclei and nucleus basalis of Meynert, were unaffected. The surprising selective decrease in CRH‐neurons provide novel targets for diagnostics and therapeutic interventions. ANN NEUROL 2022;91:282–288
Collapse
Affiliation(s)
- Ling Shan
- Leiden University Medical Centre, Department of Neurology Leiden The Netherlands
- Sleep Wake Centre SEIN Heemstede The Netherlands
| | - Rawien Balesar
- Department Neuropsychiatric Disorders Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences Amsterdam The Netherlands
| | - Dick F. Swaab
- Department Neuropsychiatric Disorders Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences Amsterdam The Netherlands
| | - Gert Jan Lammers
- Leiden University Medical Centre, Department of Neurology Leiden The Netherlands
- Sleep Wake Centre SEIN Heemstede The Netherlands
| | - Rolf Fronczek
- Leiden University Medical Centre, Department of Neurology Leiden The Netherlands
- Sleep Wake Centre SEIN Heemstede The Netherlands
| |
Collapse
|
19
|
Borniger JC. Cancer as a tool for preclinical psychoneuroimmunology. Brain Behav Immun Health 2021; 18:100351. [PMID: 34988496 PMCID: PMC8710415 DOI: 10.1016/j.bbih.2021.100351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/03/2021] [Accepted: 09/17/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer represents a novel homeostatic challenge to the host system. How the brain senses and responds to changes in peripheral physiology elicited by tumor growth is a largely untapped area of research. This is especially relevant given the widespread prevalence of systemic problems that people with various types of cancer experience. These include disruptions in sleep/wake cycles, cognitive function, depression, and changes in appetite/food intake, among others. Critically, many of these problems are evident prior to diagnosis, indicating that their etiology is potentially distinct from the effects of cancer treatment or the stress of a cancer diagnosis. Psychoneuroimmunology (PNI) is well equipped to tackle these types of problems, as it uses approaches from multiple disciplines to understand how specific stimuli (endogenous and environmental) are transduced into neural, endocrine, and immune signals that ultimately regulate health and behavior. In this article, I first provide a brief historical perspective of cancer and PNI, introduce the idea of cancer as a systemic homeostatic challenge, and provide examples from preclinical literature supporting this hypothesis. Given the rise of advanced tools in neuroscience (e.g., calcium imaging), we can now monitor and manipulate genetically defined neural circuits over the extended time scales necessary to disentangle distal communication between peripheral tumors and the brain.
Collapse
|
20
|
Peleg-Raibstein D, Burdakov D. Do orexin/hypocretin neurons signal stress or reward? Peptides 2021; 145:170629. [PMID: 34416308 DOI: 10.1016/j.peptides.2021.170629] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/14/2021] [Indexed: 12/23/2022]
Abstract
Hypothalamic neurons that produce the peptide transmitters orexins/hypocretins (HONs) broadcast their predominantly neuroexcitatory outputs to the entire brain via their extremely wide axonal projections. HONs were originally reported to be activated by food deprivation, and to stimulate arousal, energy expenditure, and eating. This led to extensive studies of HONs in the context of nutrient-sensing and energy balance control. While activation of HONs by body energy depletion continues to be supported by experimental evidence, it has also become clear that HONs are robustly activated not only by nutrient depletion, but also by diverse sensory stimuli (both neutral and those associated with rewarding or aversive events), seemingly unrelated to each other or to energy balance. One theory that could unify these findings is that all these stimuli signal "stress" - defined either as a potentially harmful state, or an awareness of reward deficiency. If HON activity is conceptualized as a cumulative representation of stress, then many of the reported HONs outputs - including EEG arousal, sympathetic activation, place avoidance, and exploratory behaviours - could be viewed as logical stress-counteracting responses. We discuss evidence for and against this unifying theory of HON function, including the alterations in HON activity observed in anxiety and depression disorders. We propose that, in order to orchestrate stress-countering responses, HONs need to coactivate motivation and aversion brain systems, and the impact of HON stimulation on affective states may be perceived as rewarding or aversive depending on the baseline HON activity.
Collapse
Affiliation(s)
| | - Denis Burdakov
- Department of Health Sciences and Technology, ETH Zürich, Switzerland.
| |
Collapse
|
21
|
Deurveilher S, Antonchuk M, Saumure BSC, Baldin A, Semba K. No loss of orexin/hypocretin, melanin-concentrating hormone or locus coeruleus noradrenergic neurons in a rat model of chronic sleep restriction. Eur J Neurosci 2021; 54:6027-6043. [PMID: 34355453 DOI: 10.1111/ejn.15412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022]
Abstract
Chronic sleep restriction (CSR) is common in modern society, adversely affecting cognitive performance and health. Yet how it impacts neurons regulating sleep remains unclear. Several studies using mice reported substantial losses of wake-active orexin/hypocretin and locus coeruleus (LC) noradrenergic neurons, but not rapid eye movement sleep-active melanin-concentrating hormone (MCH) neurons, following CSR. Here, we used immunohistochemistry and stereology to examine orexin, MCH and LC noradrenergic neurons in a rat model of CSR that uses programmed wheel rotation (3 h on/1 h off; '3/1' protocol). Adult male Wistar rats underwent one or four cycles of the 4-day 3/1 CSR protocol, with 2-day recovery between cycles in home cages. Time-matched control rats were housed in locked wheels/home cages. We found no significant differences in the numbers of orexin, MCH and LC noradrenergic neurons following either one- or four-cycle CSR protocol compared to respective controls. Similarly, the four-cycle CSR protocol had no effect on the densities of orexin axon terminals in the LC, noradrenergic dendrites in the LC and noradrenergic axon terminals in the frontal cortex. Body weights, however, decreased after one cycle of CSR and then increased with diminishing slope over the next three cycles. Thus, we found no evidence for loss of orexin or LC noradrenergic neurons following one and four cycles of the 4-day 3/1 CSR protocol in rats. Differences in CSR protocols and/or possible species differences in neuronal vulnerability to sleep loss may account for the discrepancy between the current results in rats and previous findings in mice.
Collapse
Affiliation(s)
- Samuel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michael Antonchuk
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Brock St C Saumure
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andrew Baldin
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kazue Semba
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
22
|
Alvente S, Berteotti C, Bastianini S, Lo Martire V, Matteoli G, Silvani A, Zoccoli G. Autonomic mechanisms of blood pressure alterations during sleep in orexin/hypocretin-deficient narcoleptic mice. Sleep 2021; 44:6124750. [PMID: 33517440 DOI: 10.1093/sleep/zsab022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/22/2020] [Indexed: 11/13/2022] Open
Abstract
STUDY OBJECTIVES Increase in arterial pressure (AP) during sleep and smaller differences in AP between sleep and wakefulness have been reported in orexin (hypocretin)-deficient mouse models of narcolepsy type 1 (NT1) and confirmed in NT1 patients. We tested whether these alterations are mediated by parasympathetic or sympathetic control of the heart and/or resistance vessels in an orexin-deficient mouse model of NT1. METHODS Thirteen orexin knock-out (ORX-KO) mice were compared with 12 congenic wild-type (WT) mice. The electroencephalogram, electromyogram, and AP of the mice were recorded in the light (rest) period during intraperitoneal infusion of atropine methyl nitrate, atenolol, or prazosin to block muscarinic cholinergic, β 1-adrenergic, or α 1-adrenergic receptors, respectively, while saline was infused as control. RESULTS AP significantly depended on a three-way interaction among the mouse group (ORX-KO vs WT), the wake-sleep state, and the drug or vehicle infused. During the control vehicle infusion, ORX-KO had significantly higher AP values during REM sleep, smaller decreases in AP from wakefulness to either non-rapid-eye-movement (non-REM) sleep or REM sleep, and greater increases in AP from non-REM sleep to REM sleep compared to WT. These differences remained significant with atropine methyl nitrate, whereas they were abolished by prazosin and, except for the smaller AP decrease from wakefulness to REM sleep in ORX-KO, also by atenolol. CONCLUSIONS Sleep-related alterations of AP due to orexin deficiency significantly depend on alterations in cardiovascular sympathetic control in a mouse model of NT1.
Collapse
Affiliation(s)
- Sara Alvente
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Chiara Berteotti
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Stefano Bastianini
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Viviana Lo Martire
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Gabriele Matteoli
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Alessandro Silvani
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Giovanna Zoccoli
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
23
|
Fronczek R, Schinkelshoek M, Shan L, Lammers GJ. The orexin/hypocretin system in neuropsychiatric disorders: Relation to signs and symptoms. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:343-358. [PMID: 34225940 DOI: 10.1016/b978-0-12-820107-7.00021-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Hypocretin-1 and 2 (or orexin A and B) are neuropeptides exclusively produced by a group of neurons in the lateral and dorsomedial hypothalamus that project throughout the brain. In accordance with this, the two different hypocretin receptors are also found throughout the brain. The hypocretin system is mainly involved in sleep-wake regulation, but also in reward mechanisms, food intake and metabolism, autonomic regulation including thermoregulation, and pain. The disorder most strongly linked to the hypocretin system is the primary sleep disorder narcolepsy type 1 caused by a lack of hypocretin signaling, which is most likely due to an autoimmune process targeting the hypocretin-producing neurons. However, the hypocretin system may also be affected, but to a lesser extent and less specifically, in various other neurological disorders. Examples are neurodegenerative diseases such as Alzheimer's, Huntington's and Parkinson's disease, immune-mediated disorders such as multiple sclerosis, neuromyelitis optica, and anti-Ma2 encephalitis, and genetic disorders such as type 1 diabetus mellitus and Prader-Willi Syndrome. A partial hypocretin deficiency may contribute to the sleep features of these disorders.
Collapse
Affiliation(s)
- Rolf Fronczek
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands; Sleep Wake Centre SEIN, Heemstede, The Netherlands.
| | - Mink Schinkelshoek
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands; Sleep Wake Centre SEIN, Heemstede, The Netherlands
| | - Ling Shan
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands; Sleep Wake Centre SEIN, Heemstede, The Netherlands; Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Gert Jan Lammers
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands; Sleep Wake Centre SEIN, Heemstede, The Netherlands
| |
Collapse
|
24
|
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
|
25
|
Arousing Effects of Electroacupuncture on the "Shuigou Point" in Rats with Disorder of Consciousness after Traumatic Brain Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6611461. [PMID: 33959185 PMCID: PMC8075666 DOI: 10.1155/2021/6611461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/31/2021] [Accepted: 04/08/2021] [Indexed: 11/22/2022]
Abstract
Orexin is an important neuropeptide that stimulates cortical activation and arousal and is involved in the regulation of wakefulness and arousal. Our previous meta-analysis showed that acupuncture fared well in the treatment of TBI-induced DOC in which “shuigou (DU 26)” was the most important and frequent point targeted. In the present study, we investigated whether electroacupuncture (EA) promotes TBI-induced unconsciousness wakefulness via orexin pathway. A TBI rat model was established using a control cortical impact (CCI) model. In the stimulated group, TBI rats received EA (15 Hz, 1.0 mA, 15 min). In the antagonist group, TBI rats were intraperitoneally injected with the orexin receptor 1 (OX1R) antagonist SB334867 and received EA. Unconsciousness time was observed in each group after TBI, and electrocorticography (ECoG) was applied to detect rats' EEG activity. Immunohistochemistry, enzyme-linked immunosorbent assay, and western blot were used to assess the levels of orexin-1(OX1) and OX1R expression in the mPFC. We show that duration of unconsciousness and the ratio of delta power in ECoG in the EA group were significantly reduced compared with those in the TBI group. EA could increase OX1 and OX1R expression in the mPFC and reduced the loss of orexin-producing neurons in LHA. However, all the efficacy of EA was blocked by the OX1R antagonist SB334867. Our findings suggest that EA promotes the recovery of consciousness of TBI-induced unconscious rats via upregulation of OX1and OX1R expression in mPFC.
Collapse
|
26
|
Adamantidis AR, Schmidt MH, Carter ME, Burdakov D, Peyron C, Scammell TE. A circuit perspective on narcolepsy. Sleep 2021; 43:5699663. [PMID: 31919524 PMCID: PMC7215265 DOI: 10.1093/sleep/zsz296] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/13/2019] [Indexed: 01/25/2023] Open
Abstract
The sleep disorder narcolepsy is associated with symptoms related to either boundary state control that include excessive daytime sleepiness and sleep fragmentation, or rapid eye movement (REM) sleep features including cataplexy, sleep paralysis, hallucinations, and sleep-onset REM sleep events (SOREMs). Although the loss of Hypocretin/Orexin (Hcrt/Ox) peptides or their receptors have been associated with the disease, here we propose a circuit perspective of the pathophysiological mechanisms of these narcolepsy symptoms that encompasses brain regions, neuronal circuits, cell types, and transmitters beyond the Hcrt/Ox system. We further discuss future experimental strategies to investigate brain-wide mechanisms of narcolepsy that will be essential for a better understanding and treatment of the disease.
Collapse
Affiliation(s)
- A R Adamantidis
- Department of Neurology, Centre for Experimental Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland.,Department of Biomedical Research, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| | - M H Schmidt
- Department of Neurology, Centre for Experimental Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland.,Ohio Sleep Medicine Institute, Dublin, OH
| | - M E Carter
- Department of Biology, Program in Neuroscience, Williams College, Williamstown, MA
| | - D Burdakov
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - C Peyron
- Center for Research in Neuroscience of Lyon, SLEEP team, CNRS UMR5292, INSERM U1028, University Lyon 1, Bron, France
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| |
Collapse
|
27
|
McGregor R, Thannickal TC, Siegel JM. Pleasure, addiction, and hypocretin (orexin). HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:359-374. [PMID: 34225941 DOI: 10.1016/b978-0-12-820107-7.00022-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The hypocretins/orexins were discovered in 1998. Within 2 years, this led to the discovery of the cause of human narcolepsy, a 90% loss of hypothalamic neurons containing these peptides. Further work demonstrated that these neurons were not simply linked to waking. Rather these neurons were active during pleasurable behaviors in waking and were silenced by aversive stimulation. This was seen in wild-type mice, rats, cats, and dogs. It was also evident in humans, with increased Hcrt release during pleasurable activities and decreased release, to the levels seen in sleep, during pain. We found that human heroin addicts have, on average, an increase of 54% in the number of detectable Hcrt neurons compared to "control" human brains and that these Hcrt neurons are substantially smaller than those in control brains. We found that in mice, chronic morphine administration induced the same changes in Hcrt neuron number and size. Our studies in the mouse allowed us to determine the specificity, dose response relations, time course of the change in the number of Hcrt neurons, and that the increased number of Hcrt neurons after opiates was not due to neurogenesis. Furthermore, we found that it took a month or longer for these anatomical changes in the mouse brain to return to baseline. Human narcoleptics, despite their prescribed use of several commonly addictive drugs, do not show significant evidence of dose escalation or substance use disorder. Similarly, mice in which the peptide has been eliminated are resistant to addiction. These findings are consistent with the concept that an increased number of Hcrt neurons may underlie and maintain opioid or cocaine use disorders.
Collapse
Affiliation(s)
- Ronald McGregor
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, CA, United States; Neurobiology Research, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Thomas C Thannickal
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, CA, United States; Neurobiology Research, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Jerome M Siegel
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, CA, United States; Neurobiology Research, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
28
|
Sagi D, de Lecea L, Appelbaum L. Heterogeneity of Hypocretin/Orexin Neurons. FRONTIERS OF NEUROLOGY AND NEUROSCIENCE 2021; 45:61-74. [PMID: 34052814 PMCID: PMC8961008 DOI: 10.1159/000514964] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 02/02/2021] [Indexed: 01/21/2023]
Abstract
The multifunctional, hypothalamic hypocretin/orexin (HCRT)-producing neurons regulate an array of physiological and behavioral states including arousal, sleep, feeding, emotions, stress, and reward. How a presumably uniform HCRT neuron population regulates such a diverse set of functions is not clear. The role of the HCRT neuropeptides may vary depending on the timing and localization of secretion and neuronal activity. Moreover, HCRT neuropeptides may not mediate all functions ascribed to HCRT neurons. Some could be orchestrated by additional neurotransmitters and neuropeptides that are expressed in HCRT neurons. We hypothesize that HCRT neurons are segregated into genetically, anatomically and functionally distinct subpopulations. We discuss accumulating data that suggest the existence of such HCRT neuron subpopulations that may effectuate the diverse functions of these neurons in mammals and fish.
Collapse
Affiliation(s)
- Dana Sagi
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Luis de Lecea
- Dept of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Lior Appelbaum
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel.,Corresponding author: Lior Appelbaum, Bar-Ilan University, Ramat-Gan 5290002, Israel. Telephone: +972-3-7384536,
| |
Collapse
|
29
|
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: 14] [Impact Index Per Article: 2.8] [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
|
30
|
Ahmadi-Soleimani SM, Mianbandi V, Azizi H, Azhdari-Zarmehri H, Ghaemi-Jandabi M, Abbasi-Mazar A, Mohajer Y, Darana SP. Coregulation of sleep-pain physiological interplay by orexin system: An unprecedented review. Behav Brain Res 2020; 391:112650. [DOI: 10.1016/j.bbr.2020.112650] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/28/2020] [Accepted: 04/08/2020] [Indexed: 12/14/2022]
|
31
|
van Steenoven I, Koel-Simmelink MJA, Vergouw LJM, Tijms BM, Piersma SR, Pham TV, Bridel C, Ferri GL, Cocco C, Noli B, Worley PF, Xiao MF, Xu D, Oeckl P, Otto M, van der Flier WM, de Jong FJ, Jimenez CR, Lemstra AW, Teunissen CE. Identification of novel cerebrospinal fluid biomarker candidates for dementia with Lewy bodies: a proteomic approach. Mol Neurodegener 2020; 15:36. [PMID: 32552841 PMCID: PMC7301448 DOI: 10.1186/s13024-020-00388-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Diagnosis of dementia with Lewy bodies (DLB) is challenging, largely due to a lack of diagnostic tools. Cerebrospinal fluid (CSF) biomarkers have been proven useful in Alzheimer's disease (AD) diagnosis. Here, we aimed to identify novel CSF biomarkers for DLB using a high-throughput proteomic approach. METHODS We applied liquid chromatography/tandem mass spectrometry with label-free quantification to identify biomarker candidates to individual CSF samples from a well-characterized cohort comprising patients with DLB (n = 20) and controls (n = 20). Validation was performed using (1) the identical proteomic workflow in an independent cohort (n = 30), (2) proteomic data from patients with related neurodegenerative diseases (n = 149) and (3) orthogonal techniques in an extended cohort consisting of DLB patients and controls (n = 76). Additionally, we utilized random forest analysis to identify the subset of candidate markers that best distinguished DLB from all other groups. RESULTS In total, we identified 1995 proteins. In the discovery cohort, 69 proteins were differentially expressed in DLB compared to controls (p < 0.05). Independent cohort replication confirmed VGF, SCG2, NPTX2, NPTXR, PDYN and PCSK1N as candidate biomarkers for DLB. The downregulation of the candidate biomarkers was somewhat more pronounced in DLB in comparison with related neurodegenerative diseases. Using random forest analysis, we identified a panel of VGF, SCG2 and PDYN to best differentiate between DLB and other clinical groups (accuracy: 0.82 (95%CI: 0.75-0.89)). Moreover, we confirmed the decrease of VGF and NPTX2 in DLB by ELISA and SRM methods. Low CSF levels of all biomarker candidates, except PCSK1N, were associated with more pronounced cognitive decline (0.37 < r < 0.56, all p < 0.01). CONCLUSION We identified and validated six novel CSF biomarkers for DLB. These biomarkers, particularly when used as a panel, show promise to improve diagnostic accuracy and strengthen the importance of synaptic dysfunction in the pathophysiology of DLB.
Collapse
Affiliation(s)
- Inger van Steenoven
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| | - Marleen J. A. Koel-Simmelink
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Leonie J. M. Vergouw
- Alzheimer Center Erasmus MC, Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Betty M. Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| | - Sander R. Piersma
- OncoProteomics Laboratory, Department of Medical Oncology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Thang V. Pham
- OncoProteomics Laboratory, Department of Medical Oncology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Claire Bridel
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Gian-Luca Ferri
- NEF-laboratory, Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Cristina Cocco
- NEF-laboratory, Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Barbara Noli
- NEF-laboratory, Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Paul F. Worley
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Mei-Fang Xiao
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Desheng Xu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Patrick Oeckl
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Markus Otto
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Wiesje M. van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Frank Jan de Jong
- Alzheimer Center Erasmus MC, Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Connie R. Jimenez
- OncoProteomics Laboratory, Department of Medical Oncology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Afina W. Lemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Mahoney CE, Mochizuki T, Scammell TE. Dual orexin receptor antagonists increase sleep and cataplexy in wild type mice. Sleep 2020; 43:zsz302. [PMID: 31830270 PMCID: PMC7294412 DOI: 10.1093/sleep/zsz302] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/26/2019] [Indexed: 11/13/2022] Open
Abstract
Orexin receptor antagonists are clinically useful for treating insomnia, but thorough blockade of orexin signaling could cause narcolepsy-like symptoms. Specifically, while sleepiness is a desirable effect, an orexin antagonist could also produce cataplexy, sudden episodes of muscle weakness often triggered by strong, positive emotions. In this study, we examined the effects of dual orexin receptor antagonists (DORAs), lemborexant (E2006) and almorexant, on sleep-wake behavior and cataplexy during the dark period in wild-type (WT) mice and prepro-orexin knockout (OXKO) mice. In WT mice, lemborexant at 10 and 30 mg/kg quickly induced NREM sleep in a dose-dependent fashion. In contrast, lemborexant did not alter sleep-wake behavior in OXKO mice. Under the baseline condition, cataplexy was rare in lemborexant-treated WT mice, but when mice were given chocolate as a rewarding stimulus, lemborexant dose-dependently increased cataplexy. Almorexant produced similar results. Collectively, these results demonstrate that DORAs potently increase NREM and REM sleep in mice via blockade of orexin signaling, and higher doses can cause cataplexy when co-administered with a likely rewarding stimulus.
Collapse
Affiliation(s)
- Carrie E Mahoney
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Takatoshi Mochizuki
- Graduate School of Science and Engineering, University of Toyama, Gofuku, Japan
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| |
Collapse
|
33
|
Scammell TE, Luo G, Borker P, Sullivan L, Biddle K, Mignot E. Treatment of narcolepsy with natalizumab. Sleep 2020; 43:5816455. [PMID: 32249922 DOI: 10.1093/sleep/zsaa050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/21/2020] [Indexed: 11/14/2022] Open
Affiliation(s)
- Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Guo Luo
- Center for Sleep Sciences and Medicine, Stanford University School of Medicine, Palo Alto, CA
| | - Priya Borker
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA.,Division of Pulmonary, Allergy and Critical Care, University of Pittsburgh, Pittsburgh, PA
| | - Lee Sullivan
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Kelsey Biddle
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Emmanuel Mignot
- Center for Sleep Sciences and Medicine, Stanford University School of Medicine, Palo Alto, CA
| |
Collapse
|
34
|
Ravel JM, Mignot EJM. [Narcolepsy: From the discovery of a wake promoting peptide to autoimmune T cell biology and molecular mimicry with flu epitopes]. Biol Aujourdhui 2019; 213:87-108. [PMID: 31829930 DOI: 10.1051/jbio/2019026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Indexed: 11/14/2022]
Abstract
Narcolepsy-cataplexy was first described in the late 19th century in Germany and France. Prevalence was established to be 0.05 % and a canine model was discovered in the 1970s. In 1983, a Japanese study found that all patients carried HLA-DR2, suggesting autoimmunity as the cause of the disease. Studies in the canine model established that dopaminergic stimulation underlies anti-narcoleptic action of psychostimulants, while antidepressants were found to suppress cataplexy through adrenergic reuptake inhibition. No HLA association was found in canines. A linkage study initiated in 1988 revealed in hypocretin (orexin) receptor two mutations as the cause of canine narcolepsy in 1999. In 1992, studies on African Americans showed that DQ0602 was a better marker than DR2 across all ethnic groups. In 2000, hypocretin-1/orexin A levels were measured in the cerebrospinal fluid (CSF) and found to be undetectable in most patients, establishing hypocretin deficiency as the cause of narcolepsy. Decreased CSF hypocretin-1 was then found to be secondary to the loss of the 70,000 neurons producing hypocretin in the hypothalamus, suggesting immune destruction of these cells as the cause of the disease. Additional genetic studies, notably genome wide associations (GWAS), found multiple genetic predisposing factors for narcolepsy. These were almost all involved in other autoimmune diseases, although a strong and unique association with T cell receptor (TCR) alpha and beta loci were observed. Nonetheless, all attempts to demonstrate presence of autoantibodies against hypocretin cells in narcolepsy failed, and the presumed autoimmune cause remained unproven. In 2009, association with strep throat infections were found, and narcolepsy onsets were found to occur more frequently in spring and summer, suggesting upper away infections as triggers. Following reports that narcolepsy cases were triggered by vaccinations and infections against influenza A 2009 pH1N1, a new pandemic strain that erupted in 2009, molecular mimicry with influenza A virus was suggested in 2010. This hypothesis was later confirmed by peptide screening showing higher activity of CD4+ T cell reactivity to a specific post-translationally amidated segment of hypocretin (HCRT-NH2) and cross-reactivity of specific TCRs with a pH1N1-specific segment of hemagglutinin that shares homology with HCRT-NH2. Strikingly, the most frequent TCR recognizing these antigens was found to carry sequences containing TRAJ24 or TRVB4-2, segments modulated by narcolepsy-associated genetic polymorphisms. Cross-reactive CD4+ T cells with these cross-reactive TCRs likely subsequently recruit CD8+ T cells that are then involved in hypocretin cell destruction. Additional flu mimics are also likely to be discovered since narcolepsy existed prior to 2009. The work that has been conducted over the years on narcolepsy offers a unique perspective on the conduct of research on the etiopathogeny of a specific disease.
Collapse
Affiliation(s)
- Jean-Marie Ravel
- Stanford Center for Sleep Sciences and Medicine, Department of Psychiatry and Behavioral Medicine, Stanford University, 3615 Porter Drive, Palo Alto, CA, USA
| | - Emmanuel J M Mignot
- Stanford Center for Sleep Sciences and Medicine, Department of Psychiatry and Behavioral Medicine, Stanford University, 3615 Porter Drive, Palo Alto, CA, USA
| |
Collapse
|
35
|
Cogswell AC, Maski K, Scammell TE, Tucker D, Orban ZS, Koralnik IJ. Children with Narcolepsy type 1 have increased T-cell responses to orexins. Ann Clin Transl Neurol 2019; 6:2566-2572. [PMID: 31730293 PMCID: PMC6917326 DOI: 10.1002/acn3.50908] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 11/25/2022] Open
Abstract
Narcolepsy type 1 (NT1) is caused by severe loss of the orexin neurons, and is highly associated with HLA DQB1*06:02. Using intracellular cytokine staining, we observed a higher frequency of IFN‐γ‐ and TNF‐α‐producing CD4+ and CD8+ T‐cells in response to orexins in 27 children with NT1 compared to 15 healthy control children. Conversely, no such difference was observed between 14 NT1 and 16 HC adults. In addition, priming with flu peptides amplified the T‐cell response to orexins in children with NT1. Our data suggests that NT1 may be caused by an autoimmune T‐cell response to orexins, possibly triggered by flu antigens.
Collapse
Affiliation(s)
- Andrew C Cogswell
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| | - Kiran Maski
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts
| | - Thomas E Scammell
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts.,Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Dominique Tucker
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| | - Zachary S Orban
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| | - Igor J Koralnik
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
36
|
Moreno-Estébanez A, Bilbao Villabeitia I, Echeverria Guibert T, Mendibe Bilbao M, Boyero Durán S, Cabral Martínez L, González-Pinto T, Agirre Beitia G, González Eizaguirre A, Rodríguez-Antigüedad A. Positive oligoclonal bands and CSF pleocytosis in narcolepsy type 1: A case report supporting the immune-mediated hypothesis. J Neuroimmunol 2019; 339:577111. [PMID: 31756639 DOI: 10.1016/j.jneuroim.2019.577111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/10/2019] [Accepted: 11/10/2019] [Indexed: 10/25/2022]
Abstract
Narcolepsy-type 1 is a neurological sleep-disorder caused by a selective loss of hypothalamic orexin/hypocretin-producing neurons whose underlying mechanism is considered to be immune-mediated. We report the case of a 16 year-old girl with excessive daytime sleepiness, hypnagogic/hypnopompic hallucinations and cataplexy, fulfilling narcolepsy-type 1 diagnostic criteria. She was HLA-DQB1*06:02/DQA1*01:02 positive. CSF analysis demonstrated positive IgG oligoclonal bands, pleocytosis and hypocretin-1 below detection limit. Other autoimmune processes were excluded, including autoimmune encephalitis. After treatment with intravenous immunoglobulins sleep-related hallucinations transiently improved for a month. This case's CSF inflammatory findings support the role of neuroinflammation in narcolepsy-type 1 development in genetically predisposed patients.
Collapse
Affiliation(s)
- Ana Moreno-Estébanez
- Neurology Department, Cruces University Hospital, Plaza Cruces S/N, 48903 Barakaldo, Basque Country, Spain.
| | - Iker Bilbao Villabeitia
- Neurology Department, Cruces University Hospital, Plaza Cruces S/N, 48903 Barakaldo, Basque Country, Spain
| | - Teresa Echeverria Guibert
- Neurophysiology Department, Cruces University Hospital, Plaza Cruces S/N, 48903 Barakaldo, Basque Country, Spain
| | - Mar Mendibe Bilbao
- Neurology Department, Cruces University Hospital, Plaza Cruces S/N, 48903 Barakaldo, Basque Country, Spain
| | - Sabas Boyero Durán
- Neurology Department, Cruces University Hospital, Plaza Cruces S/N, 48903 Barakaldo, Basque Country, Spain
| | - Laura Cabral Martínez
- Neurology Department, Cruces University Hospital, Plaza Cruces S/N, 48903 Barakaldo, Basque Country, Spain
| | - Tirso González-Pinto
- Neurology Department, Cruces University Hospital, Plaza Cruces S/N, 48903 Barakaldo, Basque Country, Spain
| | - Garazi Agirre Beitia
- Neurology Department, Cruces University Hospital, Plaza Cruces S/N, 48903 Barakaldo, Basque Country, Spain
| | - Amaia González Eizaguirre
- Neurology Department, Cruces University Hospital, Plaza Cruces S/N, 48903 Barakaldo, Basque Country, Spain
| | | |
Collapse
|
37
|
Narcolepsy — clinical spectrum, aetiopathophysiology, diagnosis and treatment. Nat Rev Neurol 2019; 15:519-539. [DOI: 10.1038/s41582-019-0226-9] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2019] [Indexed: 12/15/2022]
|
38
|
Walker WH, Borniger JC. Molecular Mechanisms of Cancer-Induced Sleep Disruption. Int J Mol Sci 2019; 20:E2780. [PMID: 31174326 PMCID: PMC6600154 DOI: 10.3390/ijms20112780] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023] Open
Abstract
Sleep is essential for health. Indeed, poor sleep is consistently linked to the development of systemic disease, including depression, metabolic syndrome, and cognitive impairments. Further evidence has accumulated suggesting the role of sleep in cancer initiation and progression (primarily breast cancer). Indeed, patients with cancer and cancer survivors frequently experience poor sleep, manifesting as insomnia, circadian misalignment, hypersomnia, somnolence syndrome, hot flushes, and nightmares. These problems are associated with a reduction in the patients' quality of life and increased mortality. Due to the heterogeneity among cancers, treatment regimens, patient populations and lifestyle factors, the etiology of cancer-induced sleep disruption is largely unknown. Here, we discuss recent advances in understanding the pathways linking cancer and the brain and how this leads to altered sleep patterns. We describe a conceptual framework where tumors disrupt normal homeostatic processes, resulting in aberrant changes in physiology and behavior that are detrimental to health. Finally, we discuss how this knowledge can be leveraged to develop novel therapeutic approaches for cancer-associated sleep disruption, with special emphasis on host-tumor interactions.
Collapse
Affiliation(s)
- William H Walker
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA.
| | - Jeremy C Borniger
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
39
|
Chowdhury S, Hung CJ, Izawa S, Inutsuka A, Kawamura M, Kawashima T, Bito H, Imayoshi I, Abe M, Sakimura K, Yamanaka A. Dissociating orexin-dependent and -independent functions of orexin neurons using novel Orexin-Flp knock-in mice. eLife 2019; 8:44927. [PMID: 31159922 PMCID: PMC6548533 DOI: 10.7554/elife.44927] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 05/09/2019] [Indexed: 12/11/2022] Open
Abstract
Uninterrupted arousal is important for survival during threatening situations. Activation of orexin/hypocretin neurons is implicated in sustained arousal. However, orexin neurons produce and release orexin as well as several co-transmitters including dynorphin and glutamate. To disambiguate orexin-dependent and -independent physiological functions of orexin neurons, we generated a novel Orexin-flippase (Flp) knock-in mouse line. Crossing with Flp-reporter or Cre-expressing mice showed gene expression exclusively in orexin neurons. Histological studies confirmed that orexin was knock-out in homozygous mice. Orexin neurons without orexin showed altered electrophysiological properties, as well as received decreased glutamatergic inputs. Selective chemogenetic activation revealed that both orexin and co-transmitters functioned to increase wakefulness, however, orexin was indispensable to promote sustained arousal. Surprisingly, such activation increased the total time spent in cataplexy. Taken together, orexin is essential to maintain basic membrane properties and input-output computation of orexin neurons, as well as to exert awake-sustaining aptitude of orexin neurons.
Collapse
Affiliation(s)
- Srikanta Chowdhury
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neural Regulation, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,CREST, JST, Honcho Kawaguchi, Saitama, Japan
| | - Chi Jung Hung
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neural Regulation, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,CREST, JST, Honcho Kawaguchi, Saitama, Japan
| | - Shuntaro Izawa
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neural Regulation, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,CREST, JST, Honcho Kawaguchi, Saitama, Japan
| | - Ayumu Inutsuka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Meiko Kawamura
- Department of Animal Model development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takashi Kawashima
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Itaru Imayoshi
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Manabu Abe
- Department of Animal Model development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kenji Sakimura
- Department of Animal Model development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neural Regulation, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,CREST, JST, Honcho Kawaguchi, Saitama, Japan
| |
Collapse
|
40
|
Abstract
Cancer is a systemic disease. In order to fully understand it, we must take a holistic view on how cancer interacts with its host. The brain monitors and responds to natural and aberrant signals arriving from the periphery, particularly those of metabolic or immune origin. As has been well described, a hallmark of cancer is marked disruption of metabolic and inflammatory processes. Depending on the salience and timing of these inputs, the brain responds via neural and humoral routes to alter whole-body physiology. These responses have consequences for tumor growth and metastasis, directly influencing patient quality of life and subsequent mortality. Additionally, environmental inputs such as light, diet, and stress, can promote inappropriate neural activity that benefits cancer. Here, I discuss evidence for brain-tumor interactions, with special emphasis on subcortical neuromodulator neural populations, and potential ways of harnessing this cross-talk as a novel approach for cancer treatment.
Collapse
Affiliation(s)
- Jeremy C Borniger
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, P154 MSLS Building, 1201 Welch Rd., Stanford, CA 94305, USA
| |
Collapse
|
41
|
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
|
42
|
Naganuma F, Bandaru SS, Absi G, Mahoney CE, Scammell TE, Vetrivelan R. Melanin-concentrating hormone neurons contribute to dysregulation of rapid eye movement sleep in narcolepsy. Neurobiol Dis 2018; 120:12-20. [PMID: 30149182 PMCID: PMC6195361 DOI: 10.1016/j.nbd.2018.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 08/02/2018] [Accepted: 08/23/2018] [Indexed: 12/22/2022] Open
Abstract
The lateral hypothalamus contains neurons producing orexins that promote wakefulness and suppress REM sleep as well as neurons producing melanin-concentrating hormone (MCH) that likely promote REM sleep. Narcolepsy with cataplexy is caused by selective loss of the orexin neurons, and the MCH neurons appear unaffected. As the orexin and MCH systems exert opposing effects on REM sleep, we hypothesized that imbalance in this REM sleep-regulating system due to activity in the MCH neurons may contribute to the striking REM sleep dysfunction characteristic of narcolepsy. To test this hypothesis, we chemogenetically activated the MCH neurons and pharmacologically blocked MCH signaling in a murine model of narcolepsy and studied the effects on sleep-wake behavior and cataplexy. To chemoactivate MCH neurons, we injected an adeno-associated viral vector containing the hM3Dq stimulatory DREADD into the lateral hypothalamus of orexin null mice that also express Cre recombinase in the MCH neurons (MCH-Cre::OX-KO mice) and into control MCH-Cre mice with normal orexin expression. In both lines of mice, activation of MCH neurons by clozapine-N-oxide (CNO) increased rapid eye movement (REM) sleep without altering other states. In mice lacking orexins, activation of the MCH neurons also increased abnormal intrusions of REM sleep manifest as cataplexy and short latency transitions into REM sleep (SLREM). Conversely, a MCH receptor 1 antagonist, SNAP 94847, almost completely eliminated SLREM and cataplexy in OX-KO mice. These findings affirm that MCH neurons promote REM sleep under normal circumstances, and their activity in mice lacking orexins likely triggers abnormal intrusions of REM sleep into non-REM sleep and wake, resulting in the SLREM and cataplexy characteristic of narcolepsy.
Collapse
Affiliation(s)
- Fumito Naganuma
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston MA-02215, USA; Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1, Fukumuro, Miyagino-ku, Sendai 983-8536, Japan
| | - Sathyajit S Bandaru
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston MA-02215, USA
| | - Gianna Absi
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston MA-02215, USA
| | - Carrie E Mahoney
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston MA-02215, USA
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston MA-02215, USA
| | - Ramalingam Vetrivelan
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston MA-02215, USA.
| |
Collapse
|
43
|
Arrigoni E, Chee MJS, Fuller PM. To eat or to sleep: That is a lateral hypothalamic question. Neuropharmacology 2018; 154:34-49. [PMID: 30503993 DOI: 10.1016/j.neuropharm.2018.11.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 12/15/2022]
Abstract
The lateral hypothalamus (LH) is a functionally and anatomically complex brain region that is involved in the regulation of many behavioral and physiological processes including feeding, arousal, energy balance, stress, reward and motivated behaviors, pain perception, body temperature regulation, digestive functions and blood pressure. Despite noteworthy experimental efforts over the past decades, the circuit, cellular and synaptic bases by which these different processes are regulated by the LH remains incompletely understood. This knowledge gap links in large part to the high cellular heterogeneity of the LH. Fortunately, the rapid evolution of newer genetic and electrophysiological tools is now permitting the selective manipulation, typically genetically-driven, of discrete LH cell populations. This, in turn, permits not only assignment of function to discrete cell groups, but also reveals that considerable synergistic and antagonistic interactions exist between key LH cell populations that regulate feeding and arousal. For example, we now know that while LH melanin-concentrating hormone (MCH) and orexin/hypocretin neurons both function as sensors of the internal metabolic environment, their roles regulating sleep and arousal are actually opposing. Additional studies have uncovered similarly important roles for subpopulations of LH GABAergic cells in the regulation of both feeding and arousal. Herein we review the role of LH MCH, orexin/hypocretin and GABAergic cell populations in the regulation of energy homeostasis (including feeding) and sleep-wake and discuss how these three cell populations, and their subpopulations, may interact to optimize and coordinate metabolism, sleep and arousal. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.
Collapse
Affiliation(s)
- Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02215, USA.
| | - Melissa J S Chee
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | - Patrick M Fuller
- Department of Neurology, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02215, USA
| |
Collapse
|
44
|
Burdakov D. Reactive and predictive homeostasis: Roles of orexin/hypocretin neurons. Neuropharmacology 2018; 154:61-67. [PMID: 30347195 DOI: 10.1016/j.neuropharm.2018.10.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/14/2018] [Accepted: 10/16/2018] [Indexed: 11/30/2022]
Abstract
Homeostasis is the maintenance of a healthy physiological equilibrium in a changing world. Reactive (feedback, counter-regulatory) and predictive (feedforward, anticipatory) homeostatic control strategies are both important for survival. For example, in energy homeostasis, the pancreas reacts to ingested glucose by releasing insulin, whereas the brain prepares the body for ingestion through anticipatory salivation based on food-associated cues. Reactive control is largely innate, whereas predictive control is often acquired or modified through associative learning, though some important predictive control strategies are innate, e.g. avoidance of fox scent in mice that never met a fox. Traditionally, the hypothalamus has been viewed as a reactive controller, sensing deviations from homeostasis to elicit counter-regulatory responses, while "higher" areas such as the cortex have been viewed as predictive controllers. However, experimental evidence argues against such neuroanatomical segregation: for example, receptors for internal homeostatic indicators are found throughout the brain, while key interoceptive hypothalamic cells also rapidly sense external cues. Here a model is proposed where the brain-wide-projecting, non-neuroendocrine, neurons of the hypothalamus, exemplified by orexin/hypocretin neurons, function as "brain government" systems that convert integrated internal and external information into reactive and predictive autonomic, cognitive, and behavioural adaptations that ensure homeostasis. Like regions of a country without a government, individual brain regions can function normally without hypothalamic guidance, but these functions are uncoordinated, producing mismatch between supply and demand of arousal, and derailing decision-making as seen in orexin-deficient narcolepsy. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.
Collapse
Affiliation(s)
- Denis Burdakov
- Swiss Federal Institute of Technology / ETH Zürich, D-HEST, Institute for Neuroscience, Schorenstrasse 16, Schwerzenbach 8603, Switzerland.
| |
Collapse
|
45
|
Burdakov D. How orexin signals bias action: Hypothalamic and accumbal circuits. Brain Res 2018; 1731:145943. [PMID: 30205111 DOI: 10.1016/j.brainres.2018.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/01/2018] [Accepted: 09/06/2018] [Indexed: 01/04/2023]
Abstract
Survival-maximizing, well-timed actions are a key responsibility of the brain. Hypothalamic neurons containing neurotransmitters orexins/hypocretins are important players in this process. Individuals without orexin neurons display inappropriately-timed transitions between arousal states, and other behavioural abnormalities including increased risk-taking. Deciphering neural circuits through which orexin neurons control brain states and behavior thus illuminates brain mechanisms of context-appropriate actions. This review outlines and puts into broader context recent examples of orexin circuit analyses in the lateral hypothalamus (LH) and the nucleus accumbens (NAc), two brain regions clasically implicated in context-appropriate actions. In the LH, orexin neurons excite GAD65-expressing neurons. The LH(GAD65) neuron excitation induces elevated locomotor activity, while inhibition of LH(GAD65) neuron natural activity depresses voluntary locomotion. The orexin → LH(GAD65) circuit may therefore assist in creating the drive to run. In the NAc shell region, orexin axons excite D2 neurons (dopamine-inhibited neurons expressing dopamine type-2 receptor). NAc(D2) cell activation increases risk-avoidance behaviors, while NAc(D2) cell inhibition reduces risk-avoidance. The excitatory orexin → NAc(D2) circuit may thus assist in reducing risk-taking, and oppose the inhibitory VTA(dopamine) → NAc(D2) circuit during computation of risk appetite. Neural computation in these local and long-range orexin circuits may thus assist in generating risk-avoiding locomotor responses to stressors known to activate orexin neurons, such as body energy depletion or potential external threats. A model is proposed where orexin-opposing, inhibitory inputs acting on the orexin target neurons may context-specifically channel orexin-induced brain excitation towards particular sets of actions.
Collapse
Affiliation(s)
- Denis Burdakov
- Swiss Federal Institute of Technology/ETH Zürich, Switzerland.
| |
Collapse
|
46
|
Bastianini S, Silvani A. Clinical implications of basic research. CLINICAL AND TRANSLATIONAL NEUROSCIENCE 2018. [DOI: 10.1177/2514183x18789327] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Stefano Bastianini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Silvani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
47
|
Thannickal TC, John J, Shan L, Swaab DF, Wu MF, Ramanathan L, McGregor R, Chew KT, Cornford M, Yamanaka A, Inutsuka A, Fronczek R, Lammers GJ, Worley PF, Siegel JM. Opiates increase the number of hypocretin-producing cells in human and mouse brain and reverse cataplexy in a mouse model of narcolepsy. Sci Transl Med 2018; 10:10/447/eaao4953. [PMID: 29950444 PMCID: PMC8235614 DOI: 10.1126/scitranslmed.aao4953] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/18/2017] [Accepted: 01/26/2018] [Indexed: 01/18/2023]
Abstract
The changes in brain function that perpetuate opiate addiction are unclear. In our studies of human narcolepsy, a disease caused by loss of immunohistochemically detected hypocretin (orexin) neurons, we encountered a control brain (from an apparently neurologically normal individual) with 50% more hypocretin neurons than other control human brains that we had studied. We discovered that this individual was a heroin addict. Studying five postmortem brains from heroin addicts, we report that the brain tissue had, on average, 54% more immunohistochemically detected neurons producing hypocretin than did control brains from neurologically normal subjects. Similar increases in hypocretin-producing cells could be induced in wild-type mice by long-term (but not short-term) administration of morphine. The increased number of detected hypocretin neurons was not due to neurogenesis and outlasted morphine administration by several weeks. The number of neurons containing melanin-concentrating hormone, which are in the same hypothalamic region as hypocretin-producing cells, did not change in response to morphine administration. Morphine administration restored the population of detected hypocretin cells to normal numbers in transgenic mice in which these neurons had been partially depleted. Morphine administration also decreased cataplexy in mice made narcoleptic by the depletion of hypocretin neurons. These findings suggest that opiate agonists may have a role in the treatment of narcolepsy, a disorder caused by hypocretin neuron loss, and that increased numbers of hypocretin-producing cells may play a role in maintaining opiate addiction.
Collapse
Affiliation(s)
- Thomas C. Thannickal
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Neurobiology Research, Veterans Administration Greater Los Angeles Healthcare System, 16111 Plummer Street, North Hills, CA 91343, USA
| | - Joshi John
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Neurobiology Research, Veterans Administration Greater Los Angeles Healthcare System, 16111 Plummer Street, North Hills, CA 91343, USA
| | - Ling Shan
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Neurobiology Research, Veterans Administration Greater Los Angeles Healthcare System, 16111 Plummer Street, North Hills, CA 91343, USA
| | - Dick F. Swaab
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Ming-Fung Wu
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Neurobiology Research, Veterans Administration Greater Los Angeles Healthcare System, 16111 Plummer Street, North Hills, CA 91343, USA
| | - Lalini Ramanathan
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Neurobiology Research, Veterans Administration Greater Los Angeles Healthcare System, 16111 Plummer Street, North Hills, CA 91343, USA
| | - Ronald McGregor
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Neurobiology Research, Veterans Administration Greater Los Angeles Healthcare System, 16111 Plummer Street, North Hills, CA 91343, USA
| | - Keng-Tee Chew
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Neurobiology Research, Veterans Administration Greater Los Angeles Healthcare System, 16111 Plummer Street, North Hills, CA 91343, USA
| | - Marcia Cornford
- Department of Pathology, Harbor University of California, Los Angeles, Medical Center, Torrance, CA 90509, USA
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Ayumu Inutsuka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Rolf Fronczek
- Leiden University Medical Centre, Department of Neurology, Leiden, Netherlands.,Sleep Wake Centre, Stichting Epilepsie Instellingen Nederland, Heemstede, Netherlands
| | - Gert Jan Lammers
- Leiden University Medical Centre, Department of Neurology, Leiden, Netherlands.,Sleep Wake Centre, Stichting Epilepsie Instellingen Nederland, Heemstede, Netherlands
| | - Paul F. Worley
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jerome M. Siegel
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Neurobiology Research, Veterans Administration Greater Los Angeles Healthcare System, 16111 Plummer Street, North Hills, CA 91343, USA.,Corresponding author.
| |
Collapse
|
48
|
Tyree SM, Borniger JC, de Lecea L. Hypocretin as a Hub for Arousal and Motivation. Front Neurol 2018; 9:413. [PMID: 29928253 PMCID: PMC5997825 DOI: 10.3389/fneur.2018.00413] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/18/2018] [Indexed: 01/01/2023] Open
Abstract
The lateral hypothalamus is comprised of a heterogeneous mix of neurons that serve to integrate and regulate sleep, feeding, stress, energy balance, reward, and motivated behavior. Within these populations, the hypocretin/orexin neurons are among the most well studied. Here, we provide an overview on how these neurons act as a central hub integrating sensory and physiological information to tune arousal and motivated behavior accordingly. We give special attention to their role in sleep-wake states and conditions of hyper-arousal, as is the case with stress-induced anxiety. We further discuss their roles in feeding, drug-seeking, and sexual behavior, which are all dependent on the motivational state of the animal. We further emphasize the application of powerful techniques, such as optogenetics, chemogenetics, and fiber photometry, to delineate the role these neurons play in lateral hypothalamic functions.
Collapse
Affiliation(s)
- Susan M Tyree
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Jeremy C Borniger
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| |
Collapse
|
49
|
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
|
50
|
Moresco M, Lecciso M, Ocadlikova D, Filardi M, Melzi S, Kornum BR, Antelmi E, Pizza F, Mignot E, Curti A, Plazzi G. Flow cytometry analysis of T-cell subsets in cerebrospinal fluid of narcolepsy type 1 patients with long-lasting disease. Sleep Med 2018. [DOI: 10.1016/j.sleep.2017.11.1150] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|