201
|
Bonifazi A, Del Bello F, Giorgioni G, Piergentili A, Saab E, Botticelli L, Cifani C, Micioni Di Bonaventura E, Micioni Di Bonaventura MV, Quaglia W. Targeting orexin receptors: Recent advances in the development of subtype selective or dual ligands for the treatment of neuropsychiatric disorders. Med Res Rev 2023; 43:1607-1667. [PMID: 37036052 DOI: 10.1002/med.21959] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/08/2023] [Accepted: 03/28/2023] [Indexed: 04/11/2023]
Abstract
Orexin-A and orexin-B, also named hypocretin-1 and hypocretin-2, are two hypothalamic neuropeptides highly conserved across mammalian species. Their effects are mediated by two distinct G protein-coupled receptors, namely orexin receptor type 1 (OX1-R) and type 2 (OX2-R), which share 64% amino acid identity. Given the wide expression of OX-Rs in different central nervous system and peripheral areas and the several pathophysiological functions in which they are involved, including sleep-wake cycle regulation (mainly mediated by OX2-R), emotion, panic-like behaviors, anxiety/stress, food intake, and energy homeostasis (mainly mediated by OX1-R), both subtypes represent targets of interest for many structure-activity relationship (SAR) campaigns carried out by pharmaceutical companies and academies. However, before 2017 the research was predominantly directed towards dual-orexin ligands, and limited chemotypes were investigated. Analytical characterizations, including resolved structures for both OX1-R and OX2-R in complex with agonists and antagonists, have improved the understanding of the molecular basis of receptor recognition and are assets for medicinal chemists in the design of subtype-selective ligands. This review is focused on the medicinal chemistry aspects of small molecules acting as dual or subtype selective OX1-R/OX2-R agonists and antagonists belonging to different chemotypes and developed in the last years, including radiolabeled OX-R ligands for molecular imaging. Moreover, the pharmacological effects of the most studied ligands in different neuropsychiatric diseases, such as sleep, mood, substance use, and eating disorders, as well as pain, have been discussed. Poly-pharmacology applications and multitarget ligands have also been considered.
Collapse
Affiliation(s)
- Alessandro Bonifazi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, Maryland, United States
| | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Camerino, Italy
| | - Gianfabio Giorgioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Camerino, Italy
| | | | - Elizabeth Saab
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, Maryland, United States
| | - Luca Botticelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | | | | | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Camerino, Italy
| |
Collapse
|
202
|
Abstract
Diseases associated with nicotine dependence in the form of habitual tobacco use are a major cause of premature death in the United States. The majority of tobacco smokers will relapse within the first month of attempted abstinence. Smoking cessation agents increase the likelihood that smokers can achieve long-term abstinence. Nevertheless, currently available smoking cessation agents have limited utility and fail to prevent relapse in the majority of smokers. Pharmacotherapy is therefore an effective strategy to aid smoking cessation efforts but considerable risk of relapse persists even when the most efficacious medications currently available are used. The past decade has seen major breakthroughs in our understanding of the molecular, cellular, and systems-level actions of nicotine in the brain that contribute to the development and maintenance of habitual tobacco use. In parallel, large-scale human genetics studies have revealed allelic variants that influence vulnerability to tobacco use disorder. These advances have revealed targets for the development of novel smoking cessation agents. Here, we summarize current efforts to develop smoking cessation therapeutics and highlight opportunities for future efforts.
Collapse
Affiliation(s)
- Dana Lengel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paul J. Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Drug Discovery Institute (DDI), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
203
|
Zhao J, Yan S, Li M, Sun L, Dong M, Yin M, Shen J, Zhao Z. NPFR regulates the synthesis and metabolism of lipids and glycogen via AMPK: Novel targets for efficient corn borer management. Int J Biol Macromol 2023; 247:125816. [PMID: 37451386 DOI: 10.1016/j.ijbiomac.2023.125816] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
RNA biopesticides are regarded as "the third revolution in the history of pesticides" due to their extensive advantages such as precision, high efficiency, green, pollution-free, etc. In the current study, two target genes encoding neuropeptide F receptor (NPFR) and AMP-activated protein kinase (AMPK), which are essential for insect feeding, cellular energy homeostasis and nutrient availability, were selected to design RNA pesticides. We achieved high RNA interference (RNAi) efficiency of npfr via a star polycation nanocarrier-based double-stranded RNA (dsRNA) delivery system. The food consumption of Ostrinia furnacalis is largely suppressed, which leads to a good protective effect on corn leaves. We determined the mechanism of the above genes. NPFR binds to the Gα protein and activates the intracellular second messengers cAMP and Ca2+, which in turn phosphorylate AMPK to regulate the synthesis and metabolism of lipids and glycogen. We then adopted a highly efficient bacteria-based expression system for the production of large amounts of dsRNA segments targeting npfr and ampk simultaneously and subsequently complexed them with nanocarriers to develop a novel dual-target RNA pesticide. Our RNA nanopesticide dramatically inhibits larval feeding, growth and development, and its controlling effect is even better than that of the widely used anti-feedant azadirachtin.
Collapse
Affiliation(s)
- Jiajia Zhao
- Department of Entomology and MARA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, PR China
| | - Shuo Yan
- Department of Entomology and MARA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, PR China
| | - Mingshan Li
- Department of Entomology and MARA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, PR China
| | - Lina Sun
- Department of Entomology and MARA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, PR China
| | - Min Dong
- Department of Entomology and MARA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, PR China
| | - Meizhen Yin
- State Key Lab of Chemical Resource Engineering, Beijing Lab of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Jie Shen
- Department of Entomology and MARA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, PR China.
| | - Zhangwu Zhao
- Department of Entomology and MARA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, PR China.
| |
Collapse
|
204
|
Li S, Liu M, Cao S, Liu B, Li D, Wang Z, Sun H, Cui Y, Shi Y. The Mechanism of the Gut-Brain Axis in Regulating Food Intake. Nutrients 2023; 15:3728. [PMID: 37686760 PMCID: PMC10490484 DOI: 10.3390/nu15173728] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/16/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
With the increasing prevalence of energy metabolism disorders such as diabetes, cardiovascular disease, obesity, and anorexia, the regulation of feeding has become the focus of global attention. The gastrointestinal tract is not only the site of food digestion and absorption but also contains a variety of appetite-regulating signals such as gut-brain peptides, short-chain fatty acids (SCFAs), bile acids (BAs), bacterial proteins, and cellular components produced by gut microbes. While the central nervous system (CNS), as the core of appetite regulation, can receive and integrate these appetite signals and send instructions to downstream effector organs to promote or inhibit the body's feeding behaviour. This review will focus on the gut-brain axis mechanism of feeding behaviour, discussing how the peripheral appetite signal is sensed by the CNS via the gut-brain axis and the role of the central "first order neural nuclei" in the process of appetite regulation. Here, elucidation of the gut-brain axis mechanism of feeding regulation may provide new strategies for future production practises and the treatment of diseases such as anorexia and obesity.
Collapse
Affiliation(s)
- Shouren Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.L.); (M.L.)
| | - Mengqi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.L.); (M.L.)
| | - Shixi Cao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.L.); (M.L.)
| | - Boshuai Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.L.); (M.L.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Defeng Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.L.); (M.L.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Zhichang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.L.); (M.L.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Hao Sun
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.L.); (M.L.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Yalei Cui
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.L.); (M.L.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Yinghua Shi
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.L.); (M.L.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| |
Collapse
|
205
|
Bouâouda H, Jha PK. Orexin and MCH neurons: regulators of sleep and metabolism. Front Neurosci 2023; 17:1230428. [PMID: 37674517 PMCID: PMC10478345 DOI: 10.3389/fnins.2023.1230428] [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: 05/28/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023] Open
Abstract
Sleep-wake and fasting-feeding are tightly coupled behavioral states that require coordination between several brain regions. The mammalian lateral hypothalamus (LH) is a functionally and anatomically complex brain region harboring heterogeneous cell populations that regulate sleep, feeding, and energy metabolism. Significant attempts were made to understand the cellular and circuit bases of LH actions. Rapid advancements in genetic and electrophysiological manipulation help to understand the role of discrete LH cell populations. The opposing action of LH orexin/hypocretin and melanin-concentrating hormone (MCH) neurons on metabolic sensing and sleep-wake regulation make them the candidate to explore in detail. This review surveys the molecular, genetic, and neuronal components of orexin and MCH signaling in the regulation of sleep and metabolism.
Collapse
Affiliation(s)
- Hanan Bouâouda
- Pharmacology Institute, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Pawan Kumar Jha
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
206
|
Mogavero MP, Godos J, Grosso G, Caraci F, Ferri R. Rethinking the Role of Orexin in the Regulation of REM Sleep and Appetite. Nutrients 2023; 15:3679. [PMID: 37686711 PMCID: PMC10489991 DOI: 10.3390/nu15173679] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/12/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Orexin plays a significant role in the modulation of REM sleep, as well as in the regulation of appetite and feeding. This review explores, first, the current evidence on the role of orexin in the modulation of sleep and wakefulness and highlights that orexin should be considered essentially as a neurotransmitter inhibiting REM sleep and, to a much lesser extent, a wake promoting agent. Subsequently, the relationship between orexin, REM sleep, and appetite regulation is examined in detail, shedding light on their interconnected nature in both physiological conditions and diseases (such as narcolepsy, sleep-related eating disorder, idiopathic hypersomnia, and night eating syndrome). Understanding the intricate relationship between orexin, REM sleep, and appetite regulation is vital for unraveling the complex mechanisms underlying sleep-wake patterns and metabolic control. Further research in this field is encouraged in order to pave the way for novel therapeutic approaches to sleep disorders and metabolic conditions associated with orexin dysregulation.
Collapse
Affiliation(s)
- Maria P. Mogavero
- Department of Psychology, Vita-Salute San Raffaele University, 20132 Milan, Italy;
- San Raffaele Scientific Institute, Division of Neuroscience, Sleep Disorders Center, 20127 Milan, Italy
| | - Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.); (G.G.)
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (J.G.); (G.G.)
| | - Filippo Caraci
- Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute—IRCCS, 94018 Troina, Italy;
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| | - Raffaele Ferri
- Sleep Research Centre, Oasi Research Institute—IRCCS, 94018 Troina, Italy
| |
Collapse
|
207
|
Wang S, Zhao Y, Hu X. Exploring the mechanism of Suanzaoren decoction in treatment of insomnia based on network pharmacology and molecular docking. Front Pharmacol 2023; 14:1145532. [PMID: 37670944 PMCID: PMC10475534 DOI: 10.3389/fphar.2023.1145532] [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: 01/16/2023] [Accepted: 08/08/2023] [Indexed: 09/07/2023] Open
Abstract
Objective: To explore the functional mechanisms of Suanzaoren decoction (SZRD) for treating insomnia using network pharmacology and molecular docking. Methods: The active ingredients and corresponding targets of SZRD were obtained from the Traditional Chinese Medicine Systems Pharmacology database, and then, the names of the target proteins were standardized using the UniProt database. The insomnia-related targets were obtained from the GeneCards, DisGeNET, and DrugBank databases. Next, a Venn diagram comprising the drug and disease targets was created, and the intersecting targets were used to draw the active ingredient-target network diagram using Cytoscape software. Next, the STRING database was used to build a protein-protein interaction network, followed by cluster analysis using the MCODE plug-in. The Database for Annotation, Visualization, Integrated Discovery (i.e., DAVID), and the Metascape database were used for Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. AutoDock Vina and Pymol software were used for molecular docking. Results: SZRD contained 138 active ingredients, corresponding to 239 targets. We also identified 2,062 insomnia-related targets, among which, 95 drug and disease targets intersected. The GO analysis identified 490, 62, and 114 genes related to biological processes, cellular components, and molecular functions, respectively. Lipid and atherosclerosis, chemical carcinogen-receptor activation, and neuroactive ligand-receptor interaction were the most common pathways in the KEGG analysis. Molecular docking demonstrated that the primary active components of SZRD for insomnia had good binding capabilities with the core proteins in PPI network. Conclusion: Insomnia treatment with SZRD involves multiple targets and signaling pathways, which may improve insomnia by reducing inflammation, regulating neurotransmitters.
Collapse
Affiliation(s)
- Shuxiao Wang
- Internal Encephalopathy of Traditonal Chinese Medicine, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Yan Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xingang Hu
- Internal Encephalopathy of Traditonal Chinese Medicine, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
208
|
Strickland JC, Hatton KW, Hays LR, Rayapati AO, Lile JA, Rush CR, Stoops WW. Use of drug purchase tasks in medications development research: orexin system regulation of cocaine and drug demand. Behav Pharmacol 2023; 34:275-286. [PMID: 37403694 PMCID: PMC10328554 DOI: 10.1097/fbp.0000000000000731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
Commodity purchase tasks provide a useful method for evaluating behavioral economic demand in the human laboratory. Recent research has shown how responding to purchase tasks for blinded drug administration can be used to study abuse liability. This analysis uses data from a human laboratory study to highlight how similar procedures may be particularly useful for understanding momentary changes in drug valuation when screening novel interventions. Eight nontreatment-seeking participants with cocaine use disorder (one with partial data) were enrolled in a cross-over, double-blind, randomized inpatient study. Participants were maintained on the Food and Drug Administration-approved insomnia medication suvorexant (oral; 0, 5, 10, 20 mg/day) in randomized order with experimental sessions completed after at least 3 days of maintenance on each suvorexant dose. Experimental sessions included administration of a sample dose of 0, 10 and 30 mg/70 kg intravenous cocaine. Analyses focused on purchase tasks for the blinded sample dose as well as alcohol, cigarettes and chocolate completed 15 min after the sample dose. As expected based on abuse liability, near zero demand was observed for placebo with dose-related increases in cocaine demand. Suvorexant maintenance increased cocaine demand in a dose-related manner with the greatest increase observed for the 10 mg/kg cocaine dose. Increased demand under suvorexant maintenance was also observed for alcohol. No effect of cocaine administration was observed for alcohol, cigarette, or chocolate demand. These data support the validity of demand procedures for measuring blinded drug demand. Findings also parallel self-administration data from this study by showing increases in cocaine use motivation under suvorexant maintenance.
Collapse
Affiliation(s)
- Justin C Strickland
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kevin W Hatton
- Department of Anesthesiology, University of Kentucky College of Medicine
| | - Lon R Hays
- Department of Psychiatry, University of Kentucky College of Medicine
| | - Abner O Rayapati
- Department of Psychiatry, University of Kentucky College of Medicine
| | - Joshua A Lile
- Department of Psychiatry, University of Kentucky College of Medicine
- Department of Behavioral Science, University of Kentucky College of Medicine
- Department of Psychology, University of Kentucky College of Arts and Sciences
| | - Craig R Rush
- Department of Psychiatry, University of Kentucky College of Medicine
- Department of Behavioral Science, University of Kentucky College of Medicine
- Department of Psychology, University of Kentucky College of Arts and Sciences
| | - William W Stoops
- Department of Psychiatry, University of Kentucky College of Medicine
- Department of Behavioral Science, University of Kentucky College of Medicine
- Department of Psychology, University of Kentucky College of Arts and Sciences
- Center on Drug and Alcohol Research, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
209
|
Mazur F, Całka J. Hypothalamic orexins as possible therapeutic agents in threat and spatial memory disorders. Front Behav Neurosci 2023; 17:1228056. [PMID: 37576933 PMCID: PMC10412936 DOI: 10.3389/fnbeh.2023.1228056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Orexin-A and orexin-B, neuropeptides produced exclusively in the lateral hypothalamus, have been implicated in various functions, including memory. Their levels are elevated in certain pathological states, such as PTSD, and lowered in other states, e.g., memory deficits. Recent developments have shown the possibilities of using orexins to modulate memory. Their administration can improve the results of test animals in paradigms such as passive avoidance (PA), cued fear conditioning (CFC), and the Morris water maze (MWM), with differences between the orexin used and the route of drug administration. Blocking orexin receptors in different brain structures produces opposing effects of memory impairments in given paradigms. Therefore, influencing the orexinergic balance of the brain becomes a viable way to ameliorate memory deficits, shift PTSD-induced recall of stressful memories to an extinction path, or regulate other memory processes.
Collapse
Affiliation(s)
- Filip Mazur
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Masuria in Olsztyn, Olsztyn, Poland
| | | |
Collapse
|
210
|
Dauvilliers Y, Mignot E, Del Río Villegas R, Du Y, Hanson E, Inoue Y, Kadali H, Koundourakis E, Meyer S, Rogers R, Scammell TE, Sheikh SI, Swick T, Szakács Z, von Rosenstiel P, Wu J, Zeitz H, Murthy NV, Plazzi G, von Hehn C. Oral Orexin Receptor 2 Agonist in Narcolepsy Type 1. N Engl J Med 2023; 389:309-321. [PMID: 37494485 DOI: 10.1056/nejmoa2301940] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
BACKGROUND Narcolepsy type 1 is caused by severe loss or lack of brain orexin neuropeptides. METHODS We conducted a phase 2, randomized, placebo-controlled trial of TAK-994, an oral orexin receptor 2-selective agonist, in patients with narcolepsy type 1. Patients with confirmed narcolepsy type 1 according to clinical criteria were randomly assigned to receive twice-daily oral TAK-994 (30 mg, 90 mg, or 180 mg) or placebo. The primary end point was the mean change from baseline to week 8 in average sleep latency (the time it takes to fall asleep) on the Maintenance of Wakefulness Test (range, 0 to 40 minutes; normal ability to stay awake, ≥20 minutes). Secondary end points included the change in the Epworth Sleepiness Scale (ESS) score (range, 0 to 24, with higher scores indicating greater daytime sleepiness; normal, <10) and the weekly cataplexy rate. RESULTS Of the 73 patients, 17 received TAK-994 at a dose of 30 mg twice daily, 20 received 90 mg twice daily, 19 received 180 mg twice daily, and 17 received placebo. The phase 2 trial and an extension trial were terminated early owing to hepatic adverse events. Primary end-point data were available for 41 patients (56%); the main reason for missing data was early trial termination. Least-squares mean changes to week 8 in average sleep latency on the MWT were 23.9 minutes in the 30-mg group, 27.4 minutes in the 90-mg group, 32.6 minutes in the 180-mg group, and -2.5 minutes in the placebo group (difference vs. placebo, 26.4 minutes in the 30-mg group, 29.9 minutes in the 90-mg group, and 35.0 minutes the 180-mg group; P<0.001 for all comparisons). Least-squares mean changes to week 8 in the ESS score were -12.2 in the 30-mg group, -13.5 in the 90-mg group, -15.1 in the 180-mg group, and -2.1 in the placebo group (difference vs. placebo, -10.1 in the 30-mg group, -11.4 in the 90-mg group, and -13.0 in the 180-mg group). Weekly incidences of cataplexy at week 8 were 0.27 in the 30-mg group, 1.14 in the 90-mg group, 0.88 in the 180-mg group, and 5.83 in the placebo group (rate ratio vs. placebo, 0.05 in the 30-mg group, 0.20 in the 90-mg group, and 0.15 in the 180-mg group). A total of 44 of 56 patients (79%) receiving TAK-994 had adverse events, most commonly urinary urgency or frequency. Clinically important elevations in liver-enzyme levels occurred in 5 patients, and drug-induced liver injury meeting Hy's law criteria occurred in 3 patients. CONCLUSIONS In a phase 2 trial involving patients with narcolepsy type 1, an orexin receptor 2 agonist resulted in greater improvements on measures of sleepiness and cataplexy than placebo over a period of 8 weeks but was associated with hepatotoxic effects. (Funded by Takeda Development Center Americas; TAK-994-1501 and TAK-994-1504 ClinicalTrials.gov numbers, NCT04096560 and NCT04820842.).
Collapse
Affiliation(s)
- Yves Dauvilliers
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Emmanuel Mignot
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Rafael Del Río Villegas
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Yeting Du
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Elizabeth Hanson
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Yuichi Inoue
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Harisha Kadali
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Elena Koundourakis
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Seetha Meyer
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Raquel Rogers
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Thomas E Scammell
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Sarah I Sheikh
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Todd Swick
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Zoltan Szakács
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Philipp von Rosenstiel
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Jingtao Wu
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Heidi Zeitz
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - N Venkatesha Murthy
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Giuseppe Plazzi
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| | - Christian von Hehn
- From the Sleep and Wake Disorders Center, Department of Neurology, Gui de Chauliac Hospital, and the University of Montpellier, INSERM Institute for Neurosciences of Montpellier - both in Montpellier, France (Y. Dauvilliers); the Stanford Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University Medical School, Palo Alto, CA (E.M.); the Neurophysiology and Sleep Disorders Unit, Vithas Hospitals, and Universidad CEU San Pablo, CEU Universities - both in Madrid (R.R.V.); Takeda Development Center Americas, Lexington (Y. Du, E.H., H.K., E.K., S.M., R.R., S.I.S., T.S., P.R., J.W., H.Z., N.V.M., C.H.), and the Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston (T.E.S.) - both in Massachusetts; Japan Somnology Center, Institute of Neuropsychiatry, and the Department of Somnology, Tokyo Medical University - both in Tokyo (Y.I.); the State Health Center, Budapest, Hungary (Z.S.); and IRCCS, Istituto delle Scienze Neurologiche di Bologna, Bologna, and the Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena - both in Italy (G.P.)
| |
Collapse
|
211
|
Maness EB, Blumenthal SA, Burk JA. Dual orexin/hypocretin receptor antagonism attenuates NMDA receptor hypofunction-induced attentional impairments in a rat model of schizophrenia. Behav Brain Res 2023; 450:114497. [PMID: 37196827 PMCID: PMC10330488 DOI: 10.1016/j.bbr.2023.114497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
Schizophrenia is a neuropsychiatric condition that is associated with impaired attentional processing and performance. Failure to support increasing attentional load may result, in part, from inhibitory failure in attention-relevant cortical regions, and available antipsychotics often fail to address this issue. Orexin/hypocretin receptors are found throughout the brain and are expressed on neurons relevant to both attention and schizophrenia, highlighting them as a potential target to treat schizophrenia-associated attentional dysfunction. In the present experiment, rats (N = 14) trained in a visual sustained attention task that required discrimination of trials which presented a visual signal from trials during which no signal was presented. Once trained, rats were then co-administered the psychotomimetic N-methyl-D-aspartate (NMDA) receptor antagonist dizocilpine (MK-801: 0 or 0.1 mg/kg, intraperitoneal injections) and the dual orexin receptor antagonist filorexant (MK-6096: 0, 0.1, or 1 mM, intracerebroventricular infusions) prior to task performance across six sessions. Dizocilpine impaired overall accuracy during signal trials, slowed reaction times for correctly-responded trials, and increased the number of omitted trials throughout the task. Dizocilpine-induced increases in signal trial deficits, correct response latencies, and errors of omission were reduced following infusions of the 0.1 mM, but not 1 mM, dose of filorexant. As such, orexin receptor blockade may improve attentional deficits in a state of NMDA receptor hypofunction.
Collapse
Affiliation(s)
- Eden B Maness
- Department of Psychological Sciences, College of William and Mary, Williamsburg, VA 23187, USA; VA Boston Healthcare System and Department of Psychiatry, Harvard Medical School, West Roxbury, MA 02132, USA.
| | - Sarah A Blumenthal
- Center for Translational Social Neuroscience, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Joshua A Burk
- Department of Psychological Sciences, College of William and Mary, Williamsburg, VA 23187, USA
| |
Collapse
|
212
|
Dawson M, Terstege DJ, Jamani N, Tsutsui M, Pavlov D, Bugescu R, Epp JR, Leinninger GM, Sargin D. Hypocretin/orexin neurons encode social discrimination and exhibit a sex-dependent necessity for social interaction. Cell Rep 2023; 42:112815. [PMID: 37459234 DOI: 10.1016/j.celrep.2023.112815] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 05/20/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023] Open
Abstract
The hypothalamus plays a crucial role in the modulation of social behavior by encoding internal states. The hypothalamic hypocretin/orexin neurons, initially identified as regulators of sleep and appetite, are important for emotional and motivated behaviors. However, their role in social behavior remains unclear. Using fiber photometry and behavioral analysis, we show here that hypocretin neurons differentially encode social discrimination based on the nature of social encounters. The optogenetic inhibition of hypocretin neuron activity or blocking of hcrt-1 receptors reduces the amount of time mice are engaged in social interaction in males but not in females. Reduced hcrt-1 receptor signaling during social interaction is associated with altered activity in the insular cortex and ventral tegmental area in males. Our data implicating hypocretin neurons as sexually dimorphic regulators within social networks have significant implications for the treatment of neuropsychiatric diseases with social dysfunction, particularly considering varying prevalence among sexes.
Collapse
Affiliation(s)
- Matthew Dawson
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Dylan J Terstege
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Naila Jamani
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Mio Tsutsui
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Dmitrii Pavlov
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Raluca Bugescu
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Jonathan R Epp
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Gina M Leinninger
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Derya Sargin
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
213
|
Jang HB, Ahn D, Kim HK, Guan X, Fan Y, Lee BH, Kim HY. Mediation of lateral hypothalamus orexin input to lateral habenula in the inhibitory effects of mechanical stimulation on psychomotor responses induced by cocaine. Front Mol Neurosci 2023; 16:1195939. [PMID: 37501724 PMCID: PMC10369078 DOI: 10.3389/fnmol.2023.1195939] [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: 03/29/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
Introduction The lateral hypothalamus (LH) plays an important physiological role in brain function and also plays an important role in substance abuse. The neuropeptides called orexin (or hypocretins) have been identified as being located exclusively in the cell bodies of the LH. Our previous studies have demonstrated that mechanical stimulation (MS) of the ulnar nerve produces strong inhibitory effects on cocaine addiction-like behaviors through activation of LH projection to the lateral habenula (LHb). Methods Therefore, the present study hypothesized that ulnar MS would suppress the psychomotor responses induced by cocaine through the orexinergic LH-to-LHb pathway. Results Ulnar MS attenuated cocaine enhancement of locomotor activity and 50-kHz ultrasonic vocalizations, which was prevented by antagonism of orexin-receptor type 2 (OX2R) in the LHb. Injection of orexin-A into the LHb reduced the cocaine-induced psychomotor responses. MS of the ulnar nerve excited LH orexinergic neurons. In addition, the excitation of LHb neurons by MS was blocked by the systemic administration of an OX2R antagonist. Discussion These findings suggest that MS applied to the ulnar nerve recruits an orexinergic LH-to-LHb pathway to suppress the psychomotor responses induced by cocaine.
Collapse
Affiliation(s)
- Han Byeol Jang
- Department of Physiology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea
| | - DanBi Ahn
- Department of Physiology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea
| | - Hyung Kyu Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Xiaowei Guan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Fan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bae Hwan Lee
- Department of Physiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hee Young Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
214
|
Koob GF, Vendruscolo L. Theoretical Frameworks and Mechanistic Aspects of Alcohol Addiction: Alcohol Addiction as a Reward Deficit/Stress Surfeit Disorder. Curr Top Behav Neurosci 2023. [PMID: 37421551 DOI: 10.1007/7854_2023_424] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2023]
Abstract
Alcohol use disorder (AUD) can be defined by a compulsion to seek and take alcohol, the loss of control in limiting intake, and the emergence of a negative emotional state when access to alcohol is prevented. Alcohol use disorder impacts multiple motivational mechanisms and can be conceptualized as a disorder that includes a progression from impulsivity (positive reinforcement) to compulsivity (negative reinforcement). Compulsive drug seeking that is associated with AUD can be derived from multiple neuroadaptations, but the thesis argued herein is that a key component involves the construct of negative reinforcement. Negative reinforcement is defined as drug taking that alleviates a negative emotional state. The negative emotional state that drives such negative reinforcement is hypothesized to derive from the dysregulation of specific neurochemical elements that are involved in reward and stress within basal forebrain structures that involve the ventral striatum and extended amygdala, respectively. Specific neurochemical elements in these structures include decreases in reward neurotransmission (e.g., decreases in dopamine and opioid peptide function in the ventral striatum) and the recruitment of brain stress systems (e.g., corticotropin-releasing factor [CRF]) in the extended amygdala, which contributes to hyperkatifeia and greater alcohol intake that is associated with dependence. Glucocorticoids and mineralocorticoids may play a role in sensitizing the extended amygdala CRF system. Other components of brain stress systems in the extended amygdala that may contribute to the negative motivational state of withdrawal include norepinephrine in the bed nucleus of the stria terminalis, dynorphin in the nucleus accumbens, hypocretin and vasopressin in the central nucleus of the amygdala, and neuroimmune modulation. Decreases in the activity of neuropeptide Y, nociception, endocannabinoids, and oxytocin in the extended amygdala may also contribute to hyperkatifeia that is associated with alcohol withdrawal. Such dysregulation of emotional processing may also significantly contribute to pain that is associated with alcohol withdrawal and negative urgency (i.e., impulsivity that is associated with hyperkatifeia during hyperkatifeia). Thus, an overactive brain stress response system is hypothesized to be activated by acute excessive drug intake, to be sensitized during repeated withdrawal, to persist into protracted abstinence, and to contribute to the compulsivity of AUD. The combination of the loss of reward function and recruitment of brain stress systems provides a powerful neurochemical basis for a negative emotional state that is responsible for the negative reinforcement that at least partially drives the compulsivity of AUD.
Collapse
Affiliation(s)
- George F Koob
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
| | - Leandro Vendruscolo
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
215
|
Saitoh T, Sakurai T. The Present and Future of Synthetic Orexin Receptor Agonists. Peptides 2023:171051. [PMID: 37422012 DOI: 10.1016/j.peptides.2023.171051] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/16/2023] [Accepted: 07/02/2023] [Indexed: 07/10/2023]
Abstract
The neuropeptide orexin/hypocretin plays a crucial role in various physiological processes, including the regulation of sleep/wakefulness, appetite, emotion and the reward system. Dysregulation of orexin signaling has been implicated in hypersomnia, especially in narcolepsy, which is a chronic neurological disorder characterized by excessive daytime sleepiness (EDS), sudden loss of muscle tone while awake (cataplexy), sleep paralysis, and hallucinations. Small-molecule orexin receptor agonists have emerged as promising therapeutics for these disorders, and significant progress has been made in this field in the past decade. This review summarizes recent advances in the design and synthesis of orexin receptor agonists, with a focus on peptidic and small-molecule OX2R-selective, dual, and OX1R-selective agonists. The review discusses the key structural features and pharmacological properties of these agonists, as well as their potential therapeutic applications. DATA AVAILABILITY: No data was used for the research described in the article.
Collapse
Affiliation(s)
- Tsuyoshi Saitoh
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Takeshi Sakurai
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
216
|
Kostansek JA, Latona GJ, Heruye SH, Matthews S, Bockman CS, Simeone KA, Simeone TA. Orexin receptors regulate hippocampal sharp wave-ripple complexes in ex vivo slices. Eur J Pharmacol 2023; 950:175763. [PMID: 37146705 PMCID: PMC10311575 DOI: 10.1016/j.ejphar.2023.175763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/13/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Orexin is a neuromodulatory peptide produced by lateral hypothalamic orexin neurons and binds to G-protein-coupled orexin-1 receptor and orexin-2 receptors. Whether orexin modulates learning and memory is not fully understood. Orexin has biphasic effects on learning and memory: promoting learning and memory at homeostatic levels and inhibiting at supra- and sub-homeostatic levels. Hippocampal sharp wave-ripples encode memory information and are essential for memory consolidation and retrieval. The role of orexin on sharp wave-ripples in hippocampal CA1 remains unknown. Here, we used multi-electrode array recordings in acute ex vivo hippocampal slices to determine the effects of orexin receptor antagonists on sharp wave-ripples. Bath-application of either the orexin-1 receptor antagonist N-(2-Methyl-6-benzoxazolyl)-N'-1,5-naphthyridin-4-yl urea (SB-334867) or the orexin-2 receptor antagonist N-Ethyl-2-[(6-methoxy-3-pyridinyl)[(2-methylphenyl)sulfonyl]amino]-N-(3-pyridinylmethyl)-acetamide (EMPA) reduced sharp wave and ripple incidence, sharp wave amplitude, and sharp wave duration. SB-334867 and EMPA effects on sharp wave amplitude and duration were equivalent, whereas EMPA exhibited a greater reduction of sharp wave and ripple incidence. EMPA also increased ripple duration, whereas SB-334867 had no effect. Inhibition of both orexin receptors with a dual orexin receptor antagonist N-[1,1'-Biphenyl]-2-yl-1-[2-[(1-methyl-1H-benzimidazol-2-yl)thio]acetyl-2-pyrrolidinedicarboxamide (TCS-1102) had effects similar to EMPA, however, sharp wave amplitude and duration were unaffected. Region-specific expression of orexin receptors suggests orexin may regulate sharp wave generation in CA3, dentate gyrus-mediated sharp wave modification, sharp wave propagation to CA1, and local ripple emergence in CA1. Our study indicates an orexin contribution to hippocampal sharp wave-ripple complexes and suggests a mechanism by which sub-homeostatic concentrations of orexin may inhibit learning and memory function.
Collapse
Affiliation(s)
- Joseph A Kostansek
- Creighton University, School of Medicine, Department of Pharmacology & Neuroscience, Omaha, NE, 68174, USA.
| | - Gavin J Latona
- Creighton University, School of Medicine, Department of Pharmacology & Neuroscience, Omaha, NE, 68174, USA
| | - Segewkal H Heruye
- Creighton University, School of Medicine, Department of Pharmacology & Neuroscience, Omaha, NE, 68174, USA
| | - Stephanie Matthews
- Creighton University, School of Medicine, Department of Pharmacology & Neuroscience, Omaha, NE, 68174, USA
| | - Charles S Bockman
- Creighton University, School of Medicine, Department of Pharmacology & Neuroscience, Omaha, NE, 68174, USA
| | - Kristina A Simeone
- Creighton University, School of Medicine, Department of Pharmacology & Neuroscience, Omaha, NE, 68174, USA
| | - Timothy A Simeone
- Creighton University, School of Medicine, Department of Pharmacology & Neuroscience, Omaha, NE, 68174, USA.
| |
Collapse
|
217
|
Costello A, Linning-Duffy K, Vandenbrook C, Donohue K, O'Hara BF, Kim A, Lonstein JS, Yan L. Effects of light therapy on sleep/wakefulness, daily rhythms, and the central orexin system in a diurnal rodent model of seasonal affective disorder. J Affect Disord 2023; 332:299-308. [PMID: 37060954 PMCID: PMC10161688 DOI: 10.1016/j.jad.2023.04.012] [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: 08/25/2022] [Revised: 03/31/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
BACKGROUND Bright light therapy (BLT) is the first-line treatment for seasonal affective disorder. However, the neural mechanisms underlying BLT are unclear. To begin filling this gap, the present study examined the impact of BLT on sleep/wakefulness, daily rhythms, and the wakefulness-promoting orexin/hypocretin system in a diurnal rodent, Nile grass rats (Arvicanthis niloticus). METHODS Male and female grass rats were housed under a 12:12 h light/dark cycle with dim light (50 lx) during the day. The experimental group received daily 1-h early morning BLT (full-spectrum white light, 10,000 lx), while the control group received narrowband red light for 4 weeks. Sleep/wakefulness and in-cage locomotor activity were monitored, followed by examination of hypothalamic prepro-orexin and orexin receptors OX1R and OX2R expression in corticolimbic brain regions. RESULTS The BLT group had higher wakefulness during light treatment, better nighttime sleep quality, and improved daily rhythm entrainment compared to controls. The impact of BLT on the orexin system was sex- and brain region-specific, with males showing higher OX1R and OX2R in the CA1, while females showed higher prepro-orexin but lower OX1R and OX2R in the BLA, compared to same-sex controls. LIMITATIONS The present study focused on the orexin system in a limited number of brain regions at a single time point. Sex wasn't a statistical factor, as male and female cohorts were run independently. CONCLUSIONS The diurnal grass rats show similar behavioral responses to BLT as humans, thus could be a good model for further elucidating the neural mechanisms underlying the therapeutic effects of BLT.
Collapse
Affiliation(s)
- Allison Costello
- Department of Psychology, Michigan State University, United States of America.
| | | | | | - Kevin Donohue
- Department of Electrical and Computer Engineering, Michigan State University, United States of America
| | - Bruce F O'Hara
- Department of Biology, University of Kentucky, United States of America
| | - Antony Kim
- Department of Architecture, UC Berkeley, United States of America
| | - Joseph S Lonstein
- Department of Psychology, Michigan State University, United States of America; Neuroscience Program, Michigan State University, United States of America
| | - Lily Yan
- Department of Psychology, Michigan State University, United States of America; Neuroscience Program, Michigan State University, United States of America
| |
Collapse
|
218
|
Zupančič M, Tretiakov E, Máté Z, Erdélyi F, Szabó G, Clotman F, Hökfelt T, Harkany T, Keimpema E. Brain-wide mapping of efferent projections of glutamatergic (Onecut3 + ) neurons in the lateral mouse hypothalamus. Acta Physiol (Oxf) 2023; 238:e13973. [PMID: 37029761 PMCID: PMC10909463 DOI: 10.1111/apha.13973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/09/2023]
Abstract
AIM This study mapped the spatiotemporal positions and connectivity of Onecut3+ neuronal populations in the developing and adult mouse brain. METHODS We generated fluorescent reporter mice to chart Onecut3+ neurons for brain-wide analysis. Moreover, we crossed Onecut3-iCre and Mapt-mGFP (Tau-mGFP) mice to visualize axonal projections. A dual Cre/Flp-dependent AAV construct in Onecut3-iCre cross-bred with Slc17a6-FLPo mice was used in an intersectional strategy to map the connectivity of glutamatergic lateral hypothalamic neurons in the adult mouse. RESULTS We first found that Onecut3 marks a hitherto undescribed Slc17a6+ /Vglut2+ neuronal cohort in the lateral hypothalamus, with the majority expressing thyrotropin-releasing hormone. In the adult, Onecut3+ /Vglut2+ neurons of the lateral hypothalamus had both intra- and extrahypothalamic efferents, particularly to the septal complex and habenula, where they targeted other cohorts of Onecut3+ neurons and additionally to the neocortex and hippocampus. This arrangement suggests that intrinsic reinforcement loops could exist for Onecut3+ neurons to coordinate their activity along the brain's midline axis. CONCLUSION We present both a toolbox to manipulate novel subtypes of hypothalamic neurons and an anatomical arrangement by which extrahypothalamic targets can be simultaneously entrained.
Collapse
Affiliation(s)
- Maja Zupančič
- Department of Molecular Neurosciences, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Evgenii Tretiakov
- Department of Molecular Neurosciences, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Zoltán Máté
- Institute of Experimental Medicine, Hungarian Academy of SciencesBudapestHungary
| | - Ferenc Erdélyi
- Institute of Experimental Medicine, Hungarian Academy of SciencesBudapestHungary
| | - Gábor Szabó
- Institute of Experimental Medicine, Hungarian Academy of SciencesBudapestHungary
| | - Frédéric Clotman
- Animal Molecular and Cellular Biology Group, Louvain Institute of Biomolecular Science and TechnologyUniversité Catholique de LouvainLouvain‐la‐NeuveBelgium
| | - Tomas Hökfelt
- Department of Neuroscience, Biomedicum 7DKarolinska InstitutetSolnaSweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain ResearchMedical University of ViennaViennaAustria
- Department of Neuroscience, Biomedicum 7DKarolinska InstitutetSolnaSweden
| | - Erik Keimpema
- Department of Molecular Neurosciences, Center for Brain ResearchMedical University of ViennaViennaAustria
| |
Collapse
|
219
|
van Lemmen M, van der Schrier R, Dahan A, van Velzen M, Sarton E, Niesters M. Pharmacology of viable mechanism agnostic respiratory stimulants for the reversal of drug-induced respiratory depression in humans. Expert Opin Drug Metab Toxicol 2023; 19:671-679. [PMID: 37795596 DOI: 10.1080/17425255.2023.2262386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/20/2023] [Indexed: 10/06/2023]
Abstract
INTRODUCTION Drug-induced respiratory depression is potentially fatal and can be caused by various drugs such as synthetic opioids and tranquilizers. The only class of respiratory depressants that has a specific reversal agent are opioids, such as naloxone. These reversal agents have limited utility in situations of polysubstance ingestion with agents from multiple respiratory depressant classes. Hence, there is an unmet need for drugs that stimulate breathing irrespective of the underlying cause of respiratory depression, i.e. mechanism agnostic respiratory stimulants. AREAS COVERED In this review, we discuss agnostic respiratory stimulants, tested in humans with promising results, i.e. ampakines, drugs that act at the carotid bodies, N-methyl-D-aspartate receptor antagonist ketamine, and orexin receptor-2-agonist danavorexton, and others that demonstrated positive effects in animals but not yet in humans. EXPERT OPINION Rapid, effective rescuing of individuals who overdosed on respiratory depressants saves lives. While naloxone is the preferred drug for reversing opioid-induced respiratory depression, its effectiveness is limited in cases involving non-opioids. While several agnostic respiratory stimulants showed promise in humans, further research is needed to optimize dosing, evaluate safety and efficacy in deeper respiratory depression (apnea). Additionally, future studies should combine agnostic stimulants with naloxone, to improve rapid, effective rescue from drug overdoses.
Collapse
Affiliation(s)
- Maarten van Lemmen
- Department of Anesthesiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Albert Dahan
- Department of Anesthesiology, Leiden University Medical Center, Leiden, the Netherlands
- PainLess Foundation, Leiden, the Netherlands
| | - Monique van Velzen
- Department of Anesthesiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Elise Sarton
- Department of Anesthesiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marieke Niesters
- Department of Anesthesiology, Leiden University Medical Center, Leiden, the Netherlands
- PainLess Foundation, Leiden, the Netherlands
| |
Collapse
|
220
|
Raskin M, Malone C, Hilz EN, Smits JAJ, Telch MJ, Otto MW, Shumake J, Lee HJ, Monfils MH. CO 2 reactivity is associated with individual differences in appetitive extinction memory. Physiol Behav 2023; 266:114183. [PMID: 37031791 PMCID: PMC10840099 DOI: 10.1016/j.physbeh.2023.114183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/17/2023] [Accepted: 04/04/2023] [Indexed: 04/11/2023]
Abstract
Pavlovian conditioning can underly the maladaptive behaviors seen in psychiatric disorders such as anxiety and addiction. In both the lab and the clinic, these responses can be attenuated through extinction learning, but often return with the passage of time, stress, or a change in context. Extinction to fear and reward cues are both subject to these return of behavior phenomena and have overlap in neurocircuitry, yet it is unknown whether they share any common predictors. The orexin system has been implicated in both fear and appetitive extinction and can be activated through a CO2 challenge. We previously found that behavioral CO2 reactivity predicts fear extinction and orexin activation. Here, we sought to extend our previous findings to determine whether CO2 reactivity might also predict extinction memory for appetitive light-food conditioning. We find that the same subcomponent of behavioral CO2 reactivity that predicted fear extinction also predicts appetitive extinction, but in the opposite direction. We show evidence that this subcomponent remains stable across two CO2 challenges, suggesting it may be a stable trait of both behavioral CO2 reactivity and appetitive extinction phenotype. Our findings further the possibility for CO2 reactivity to be used as a transdiagnostic screening tool to determine whether an individual would be a good candidate for exposure therapy.
Collapse
Affiliation(s)
- Marissa Raskin
- The University of Texas at Austin, Institute for Neuroscience, United States
| | - Cassidy Malone
- The University of Texas at Austin, Department of Psychology, United States
| | - Emily N Hilz
- The University of Texas at Austin, Department of Psychology, United States
| | - Jasper A J Smits
- The University of Texas at Austin, Department of Psychology, United States; The University of Texas at Austin, Institute for Mental Health Research, United States
| | - Michael J Telch
- The University of Texas at Austin, Department of Psychology, United States; The University of Texas at Austin, Institute for Mental Health Research, United States
| | | | - Jason Shumake
- The University of Texas at Austin, Department of Psychology, United States; The University of Texas at Austin, Institute for Mental Health Research, United States
| | - Hongjoo J Lee
- The University of Texas at Austin, Institute for Neuroscience, United States; The University of Texas at Austin, Department of Psychology, United States
| | - Marie-H Monfils
- The University of Texas at Austin, Institute for Neuroscience, United States; The University of Texas at Austin, Department of Psychology, United States; The University of Texas at Austin, Institute for Mental Health Research, United States.
| |
Collapse
|
221
|
Rivas M, Ferreira A, Torterolo P, Benedetto L. Hypocretins, sleep, and maternal behavior. Front Behav Neurosci 2023; 17:1184885. [PMID: 37456808 PMCID: PMC10347526 DOI: 10.3389/fnbeh.2023.1184885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
The postpartum period is a demanding time during which mothers experience numerous physiological adaptations that enable them to care for their offspring while maintaining their wellbeing. Hypocretins, also known as orexins, are neuropeptides synthesized by hypothalamic neurons that play a fundamental role in several functions, including the promotion of wakefulness and motivated behaviors, such as maternal care. In this regard, several findings suggest that the activity of the hypocretinergic system increases in the early postpartum period and begins to decline as weaning approaches. In particular, hypocretins within the medial preoptic area, a crucial region during this period, modulate both maternal behavior and sleep. Although further studies are necessary to obtain a comprehensive understanding of the role of hypocretins in lactating females, current research suggests that this system participates in promoting active components of maternal behavior and regulating wakefulness and sleep adjustments during the postpartum period, potentially leading to increased wakefulness during this stage. These adaptive adjustments enable the mother to cope with the continuously changing demands of the pups.
Collapse
Affiliation(s)
- Mayda Rivas
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Annabel Ferreira
- Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Pablo Torterolo
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Luciana Benedetto
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
222
|
Yoshida-Tanaka K, Shimada M, Honda Y, Fujimoto A, Tokunaga K, Honda M, Miyagawa T. Narcolepsy type I-associated DNA methylation and gene expression changes in the human leukocyte antigen region. Sci Rep 2023; 13:10464. [PMID: 37380713 DOI: 10.1038/s41598-023-37511-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023] Open
Abstract
Narcolepsy type 1 (NT1) is caused by a loss of hypothalamic orexin-producing cells, and autoreactive CD4+ and CD8+ T cells have been suggested to play a role in the autoimmune mechanism. Although NT1 showed a strong association with human leukocyte antigen (HLA)-DQB1*06:02, the responsible antigens remain unidentified. We analyzed array-based DNA methylation and gene expression data for the HLA region in CD4+ and CD8+ T cells that were separated from the peripheral blood mononuclear cells of Japanese subjects (NT1, N = 42; control, N = 42). As the large number of SNPs in the HLA region might interfere with the affinity of the array probes, we conducted a comprehensive assessment of the reliability of each probe. The criteria were based on a previous study reporting that the presence of frequent SNPs, especially on the 3' side of the probe, makes the probe unreliable. We confirmed that 90.3% of the probes after general filtering in the HLA region do not include frequent SNPs, and are thus suitable for analysis, particularly in Japanese subjects. We then performed an association analysis, and found that several CpG sites in the HLA class II region of the patients were significantly hypomethylated in CD4+ and CD8+ T cells. This association was not detected when the effect of HLA-DQB1*06:02 was considered, suggesting that the hypomethylation was possibly derived from HLA-DQB1*06:02. Further RNA sequencing revealed reduced expression levels of HLA-DQB1 alleles other than HLA-DQB1*06:02 in the patients with NT1. Our results suggest the involvement of epigenetic and expressional changes in HLA-DQB1 in the pathogenesis of NT1.
Collapse
Affiliation(s)
- Kugui Yoshida-Tanaka
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mihoko Shimada
- Genome Medical Science Project (Toyama), National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan.
| | - Yoshiko Honda
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Akihiro Fujimoto
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsushi Tokunaga
- Genome Medical Science Project (Toyama), National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Makoto Honda
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Seiwa Hospital, Institute of Neuropsychiatry, Tokyo, Japan
| | - Taku Miyagawa
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
223
|
Khouma A, Moeini MM, Plamondon J, Richard D, Caron A, Michael NJ. Histaminergic regulation of food intake. Front Endocrinol (Lausanne) 2023; 14:1202089. [PMID: 37448468 PMCID: PMC10338010 DOI: 10.3389/fendo.2023.1202089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/06/2023] [Indexed: 07/15/2023] Open
Abstract
Histamine is a biogenic amine that acts as a neuromodulator within the brain. In the hypothalamus, histaminergic signaling contributes to the regulation of numerous physiological and homeostatic processes, including the regulation of energy balance. Histaminergic neurons project extensively throughout the hypothalamus and two histamine receptors (H1R, H3R) are strongly expressed in key hypothalamic nuclei known to regulate energy homeostasis, including the paraventricular (PVH), ventromedial (VMH), dorsomedial (DMH), and arcuate (ARC) nuclei. The activation of different histamine receptors is associated with differential effects on neuronal activity, mediated by their different G protein-coupling. Consequently, activation of H1R has opposing effects on food intake to that of H3R: H1R activation suppresses food intake, while H3R activation mediates an orexigenic response. The central histaminergic system has been implicated in atypical antipsychotic-induced weight gain and has been proposed as a potential therapeutic target for the treatment of obesity. It has also been demonstrated to interact with other major regulators of energy homeostasis, including the central melanocortin system and the adipose-derived hormone leptin. However, the exact mechanisms by which the histaminergic system contributes to the modification of these satiety signals remain underexplored. The present review focuses on recent advances in our understanding of the central histaminergic system's role in regulating feeding and highlights unanswered questions remaining in our knowledge of the functionality of this system.
Collapse
Affiliation(s)
- Axelle Khouma
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Moein Minbashi Moeini
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Julie Plamondon
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
| | - Denis Richard
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Medicine, Université Laval, Québec, QC, Canada
| | - Alexandre Caron
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
- Montreal Diabetes Research Center, Montreal, QC, Canada
| | - Natalie Jane Michael
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| |
Collapse
|
224
|
Elhosainy A, Suzuki-Abe H, Kaushik MK, Kim SJ, Saitoh T, Ishikawa Y, Hotta-Hirashima N, Miyoshi C, Funato H, Yanagisawa M. Face validation and pharmacologic analysis of Sik3 Sleepy mutant mouse as a possible model of idiopathic hypersomnia. Eur J Pharmacol 2023:175877. [PMID: 37356786 DOI: 10.1016/j.ejphar.2023.175877] [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: 01/19/2023] [Revised: 06/09/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023]
Abstract
Idiopathic hypersomnia (IH) is a chronic neurologic disorder with unknown mechanisms that result in long night-time sleep, daytime sleepiness, long non-refreshing naps, and difficult awakening presenting as sleep drunkenness. IH patients are typically diagnosed by shorter sleep latency on multiple sleep latency test (MSLT) along with long sleep time. Only symptomatic drug treatments are currently available for IH and no animal model to study it. Sleepy mice carry a splicing mutation in the Sik3 gene, leading to increased sleep time and sleep need. Here we used a mouse version of MSLT and a decay analysis of wake EEG delta power to validate the Sleepy mutant mouse as an animal model for IH. Sleepy mice had shorter sleep latency in the dark (active) phase than wild-type mice. They also showed lower decay of EEG delta density during wakefulness, possibly reflecting increased sleep inertia. These data indicate that the Sleepy mouse may have partial face validity as a mouse model for idiopathic hypersomnia. We then investigated the effect of orexin-A and the orexin receptor 2-selective agonist YNT-185 on the sleepiness symptoms of the Sleepy mouse. Intracerebroventricular orexin-A promoted wakefulness for 3 h and decreased wake EEG delta density after injection in Sleepy mice and wild-type mice. Moreover, Sleepy mice but not wild-type mice showed a sleep rebound after the orexin-A-induced wakefulness. Intraperitoneal YNT-185 promoted wakefulness for 3 h after injection in Sleepy mice, indicating the potential of using orexin agonists to treat not only orexin deficiency but hypersomnolence of various etiologies.
Collapse
Affiliation(s)
- Asmaa Elhosainy
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Haruka Suzuki-Abe
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Mahesh K Kaushik
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Staci J Kim
- 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
| | - Yukiko Ishikawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Noriko Hotta-Hirashima
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Chika Miyoshi
- 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, Graduate School of Medicine, Toho University, Tokyo, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan; Life Science Centre for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA; R&D Center for Frontiers of Mirai in Policy and Technology (F-MIRAI), University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
225
|
Grujic N, Tesmer A, Bracey E, Peleg-Raibstein D, Burdakov D. Control and coding of pupil size by hypothalamic orexin neurons. Nat Neurosci 2023:10.1038/s41593-023-01365-w. [PMID: 37336973 DOI: 10.1038/s41593-023-01365-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/17/2023] [Indexed: 06/21/2023]
Abstract
Brain orexin (hypocretin) neurons are implicated in sleep-wake switching and reward-seeking but their roles in rapid arousal dynamics and reward perception are unclear. Here, cell-specific stimulation, deletion and in vivo recordings revealed strong correlative and causal links between pupil dilation-a quantitative arousal marker-and orexin cell activity. Coding of arousal and reward was distributed across orexin cells, indicating that they specialize in rapid, multiplexed communication of momentary arousal and reward states.
Collapse
Affiliation(s)
- Nikola Grujic
- Neurobehavioural Dynamics Laboratory, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Alexander Tesmer
- Neurobehavioural Dynamics Laboratory, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Ed Bracey
- Neurobehavioural Dynamics Laboratory, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Daria Peleg-Raibstein
- Neurobehavioural Dynamics Laboratory, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Denis Burdakov
- Neurobehavioural Dynamics Laboratory, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
226
|
Pai SK. Mechanisms underlying fragmented sleep in aging. AGING BRAIN 2023; 3:100077. [PMID: 37304173 PMCID: PMC10248856 DOI: 10.1016/j.nbas.2023.100077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 06/13/2023] Open
|
227
|
Kukkonen JP. The G protein preference of orexin receptors is currently an unresolved issue. Nat Commun 2023; 14:3162. [PMID: 37264034 PMCID: PMC10235037 DOI: 10.1038/s41467-023-38764-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 05/15/2023] [Indexed: 06/03/2023] Open
Affiliation(s)
- Jyrki P Kukkonen
- Department of Pharmacology, Institute of Biomedicine, Faculty of Medicine, University of Helsinki, POB 63, FI-00014, Helsinki, Finland.
| |
Collapse
|
228
|
Ishikawa T, Hara H, Kawano A, Tohyama K, Kajita Y, Miyanohana Y, Koike T, Kimura H. TAK-994, a Novel Orally Available Brain-Penetrant Orexin 2 Receptor-Selective Agonist, Suppresses Fragmentation of Wakefulness and Cataplexy-Like Episodes in Mouse Models of Narcolepsy. J Pharmacol Exp Ther 2023; 385:193-204. [PMID: 37001988 DOI: 10.1124/jpet.122.001449] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/23/2022] [Accepted: 02/28/2023] [Indexed: 04/03/2023] Open
Abstract
Loss of orexin neurons is associated with narcolepsy type 1 (NT1), which is characterized by multiple symptoms including excessive daytime sleepiness and cataplexy. Orexin 2 receptor (OX2R) knockout (KO) mice, but not orexin 1 receptor (OX1R) KO mice, show narcolepsy-like phenotypes, thus OX2R agonists are potentially promising for treating NT1. In fact, in early proof-of-concept studies, intravenous infusion of danavorexton, an OX2R-selective agonist, significantly increased wakefulness in individuals with NT1. However, danavorexton has limited oral availability. Here, we report pharmacological characteristics of a novel OX2R agonist, TAK-994 [N-{(2S,3S)-1-(2-hydroxy-2-methylpropanoyl)-2-[(2,3',5'-trifluorobiphenyl-3-yl)methyl]pyrrolidin-3-yl}methanesulfonamide sesquihydrate]. TAK-994 activated recombinant human OX2R (EC50 value of 19 nM) with > 700-fold selectivity against OX1R and activated OX2R-downstream signaling similar to those by orexin peptides in vitro. Oral administration of TAK-994 promoted wakefulness in normal mice but not in OX2R KO mice. TAK-994 also ameliorated narcolepsy-like symptoms in two mouse models of narcolepsy: orexin/ataxin-3 mice and orexin-tTA;TetO diphtheria toxin A mice. The wake-promoting effects of TAK-994 in orexin/ataxin-3 mice were maintained after chronic dosing for 14 days. These data suggest that overall in vitro and in vivo properties, except oral availability, are very similar between TAK-994 and danavorexton. Preclinical characteristics of TAK-994 shown here, together with upcoming clinical study results, can improve our understanding for orally available OX2R agonists as new therapeutic drugs for NT1 and other hypersomnia disorders. SIGNIFICANCE STATEMENT: Narcolepsy type 1 (NT1) is caused by a loss of orexin neurons, and thus an orexin 2 receptor (OX2R) agonist is considered to address the underlying pathophysiology of NT1. Oral administration of TAK-994, a novel OX2R agonist, promoted wakefulness in normal mice, but not in OX2R knockout mice, and ameliorated fragmentation of wakefulness and cataplexy-like episodes in mouse models of narcolepsy. These findings indicate that TAK-994 is an orally available brain-penetrant OX2R-selective agonist with potential to improve narcolepsy-like symptoms.
Collapse
Affiliation(s)
- Takashi Ishikawa
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hiroe Hara
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Ayumi Kawano
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Kimio Tohyama
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yuichi Kajita
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yuhei Miyanohana
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Tatsuki Koike
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Haruhide Kimura
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
229
|
Sun Y, Tisdale RK, Yamashita A, Kilduff TS. Peripheral vs. core body temperature as hypocretin/orexin neurons degenerate: Exercise mitigates increased heat loss. Peptides 2023; 164:171002. [PMID: 36963505 PMCID: PMC10337601 DOI: 10.1016/j.peptides.2023.171002] [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: 12/17/2022] [Revised: 03/01/2023] [Accepted: 03/21/2023] [Indexed: 03/26/2023]
Abstract
Hypocretins/Orexins (Hcrt/Ox) are hypothalamic neuropeptides implicated in diverse functions, including body temperature regulation through modulation of sympathetic vasoconstrictor tone. In the current study, we measured subcutaneous (Tsc) and core (Tb) body temperature as well as activity in a conditional transgenic mouse strain that allows the inducible ablation of Hcrt/Ox-containing neurons by removal of doxycycline (DOX) from their diet (orexin-DTA mice). Measurements were made during a baseline, when mice were being maintained on food containing DOX, and over 42 days while the mice were fed normal chow which resulted in Hcrt/Ox neuron degeneration. The home cages of the orexin-DTA mice were equipped with running wheels that were either locked or unlocked. In the presence of a locked running wheel, Tsc progressively decreased on days 28 and 42 in the DOX(-) condition, primarily during the dark phase (the major active period for rodents). This nocturnal reduction in Tsc was mitigated when mice had access to unlocked running wheels. In contrast to Tsc, Tb was largely maintained until day 42 in the DOX(-) condition even when the running wheel was locked. Acute changes in both Tsc and Tb were observed preceding, during, and following cataplexy. Our results suggest that ablation of Hcrt/Ox-containing neurons results in elevated heat loss, likely through reduced sympathetic vasoconstrictor tone, and that exercise may have some therapeutic benefit to patients with narcolepsy, a disorder caused by Hcrt/Ox deficiency. Acute changes in body temperature may facilitate prediction of cataplexy onset and lead to interventions to mitigate its occurrence.
Collapse
Affiliation(s)
- Yu Sun
- Biosciences Division, SRI International, 333 Ravenswood Ave, Menlo Park, CA 94025, USA.
| | - Ryan K Tisdale
- Biosciences Division, SRI International, 333 Ravenswood Ave, Menlo Park, CA 94025, USA
| | - Akira Yamashita
- Biosciences Division, SRI International, 333 Ravenswood Ave, Menlo Park, CA 94025, USA; Department of Physiology, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8544, Japan
| | - Thomas S Kilduff
- Biosciences Division, SRI International, 333 Ravenswood Ave, Menlo Park, CA 94025, USA
| |
Collapse
|
230
|
St-Onge MP, Cherta-Murillo A, Darimont C, Mantantzis K, Martin FP, Owen L. The interrelationship between sleep, diet, and glucose metabolism. Sleep Med Rev 2023; 69:101788. [PMID: 37156196 PMCID: PMC10247426 DOI: 10.1016/j.smrv.2023.101788] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/28/2023] [Accepted: 04/23/2023] [Indexed: 05/10/2023]
Abstract
Obesity and type 2 diabetes (T2D) are increasingly common worldwide. While these disorders have increased in prevalence over the past several decades, there has been a concomitant reduction in sleep duration. Short sleep duration has been associated with higher rates of obesity and T2D, and the causality of these associations and their directionality, continue to necessitate evaluation. In this review we consider the evidence that sleep is an intrinsic factor in the development of obesity and chronic metabolic disorders, such as insulin resistance and T2D, while evaluating a potential bi-directional association. We consider the evidence that diet and meal composition, which are known to impact glycemic control, may have both chronic and acute impact upon sleep. Moreover, we consider that postprandial nocturnal metabolism and peripheral glycemia may affect sleep quality. We propose putative mechanisms whereby acute effects of nighttime glucose excursions may lead to increased sleep fragmentation. We conclude that dietary manipulations, particularly with respect to carbohydrate quality, may confer sleep benefits. Future research may seek to evaluate the effectiveness of synergistic nutrient strategies to promote sleep quality, with particular attention to carbohydrate quality, quantity, and availability as well as carbohydrate to protein ratio.
Collapse
Affiliation(s)
- Marie-Pierre St-Onge
- Division of General Medicine and Center of Excellence for Sleep & Circadian Research, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | | | - Christian Darimont
- Nestlé Research, Nestlé Institute of Health Sciences, Lausanne, Switzerland
| | | | | | - Lauren Owen
- Nestlé Research, Nestlé Institute of Health Sciences, Lausanne, Switzerland
| |
Collapse
|
231
|
Kniazkina M, Dyachuk V. Does EGFR Signaling Mediate Orexin System Activity in Sleep Initiation? Int J Mol Sci 2023; 24:ijms24119505. [PMID: 37298454 DOI: 10.3390/ijms24119505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/21/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Sleep-wake cycle disorders are an important symptom of many neurological diseases, including Parkinson's disease, Alzheimer's disease, and multiple sclerosis. Circadian rhythms and sleep-wake cycles play a key role in maintaining the health of organisms. To date, these processes are still poorly understood and, therefore, need more detailed elucidation. The sleep process has been extensively studied in vertebrates, such as mammals and, to a lesser extent, in invertebrates. A complex, multi-step interaction of homeostatic processes and neurotransmitters provides the sleep-wake cycle. Many other regulatory molecules are also involved in the cycle regulation, but their functions remain largely unclear. One of these signaling systems is epidermal growth factor receptor (EGFR), which regulates the activity of neurons in the modulation of the sleep-wake cycle in vertebrates. We have evaluated the possible role of the EGFR signaling pathway in the molecular regulation of sleep. Understanding the molecular mechanisms that underlie sleep-wake regulation will provide critical insight into the fundamental regulatory functions of the brain. New findings of sleep-regulatory pathways may provide new drug targets and approaches for the treatment of sleep-related diseases.
Collapse
Affiliation(s)
- Marina Kniazkina
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Vyacheslav Dyachuk
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| |
Collapse
|
232
|
Jin R, Sun S, Hu Y, Zhang H, Sun X. Neuropeptides Modulate Feeding via the Dopamine Reward Pathway. Neurochem Res 2023:10.1007/s11064-023-03954-4. [PMID: 37233918 DOI: 10.1007/s11064-023-03954-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Dopamine (DA) is a catecholamine neurotransmitter widely distributed in the central nervous system. It participates in various physiological functions, such as feeding, anxiety, fear, sleeping and arousal. The regulation of feeding is exceptionally complex, involving energy homeostasis and reward motivation. The reward system comprises the ventral tegmental area (VTA), nucleus accumbens (NAc), hypothalamus, and limbic system. This paper illustrates the detailed mechanisms of eight typical orexigenic and anorexic neuropeptides that regulate food intake through the reward system. According to recent literature, neuropeptides released from the hypothalamus and other brain regions regulate reward feeding predominantly through dopaminergic neurons projecting from the VTA to the NAc. In addition, their effect on the dopaminergic system is mediated by the prefrontal cortex, paraventricular thalamus, laterodorsal tegmental area, amygdala, and complex neural circuits. Research on neuropeptides involved in reward feeding can help identify more targets to treat diseases with metabolic disorders, such as obesity.
Collapse
Affiliation(s)
- Ruijie Jin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Shanbin Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Yang Hu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Hongfei Zhang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xiangrong Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
233
|
Willmore L, Minerva AR, Engelhard B, Murugan M, McMannon B, Oak N, Thiberge SY, Peña CJ, Witten IB. Overlapping representations of food and social stimuli in VTA dopamine neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541104. [PMID: 37293057 PMCID: PMC10245666 DOI: 10.1101/2023.05.17.541104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Dopamine neurons of the ventral tegmental area (VTA DA ) respond to food and social stimuli and contribute to both forms of motivation. However, it is unclear if the same or different VTA DA neurons encode these different stimuli. To address this question, we performed 2-photon calcium imaging in mice presented with food and conspecifics, and found statistically significant overlap in the populations responsive to both stimuli. Both hunger and opposite-sex social experience further increased the proportion of neurons that respond to both stimuli, implying that modifying motivation for one stimulus affects responses to both stimuli. In addition, single-nucleus RNA sequencing revealed significant co-expression of feeding- and social-hormone related genes in individual VTA DA neurons. Taken together, our functional and transcriptional data suggest overlapping VTA DA populations underlie food and social motivation.
Collapse
Affiliation(s)
- Lindsay Willmore
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Adelaide R. Minerva
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Ben Engelhard
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
- Department of Medicine, Technion, Haifa, 3525433, Israel
| | - Malavika Murugan
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Brenna McMannon
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Nirja Oak
- Department of Medicine, Technion, Haifa, 3525433, Israel
| | - Stephan Y. Thiberge
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Catherine J. Peña
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| | - Ilana B. Witten
- Princeton Neuroscience Institute, Princeton University, Princeton NJ 08544 USA
| |
Collapse
|
234
|
Bogan RK, Maynard JP, Neuwirth R, Faessel H, Swick T, Olsson T. Safety and pharmacodynamics of a single infusion of danavorexton in adults with obstructive sleep apnea experiencing excessive daytime sleepiness despite adequate use of CPAP. Sleep Med 2023; 107:229-235. [PMID: 37244138 DOI: 10.1016/j.sleep.2023.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/06/2023] [Accepted: 05/01/2023] [Indexed: 05/29/2023]
Abstract
BACKGROUND Sleep disruptions experienced by patients with obstructive sleep apnea (OSA) can lead to excessive daytime sleepiness (EDS) and significantly impact patients' quality of life. EDS may persist despite use of continuous positive airway pressure (CPAP) therapy. Small molecules that target the orexin system, which has a known role in sleep-wake regulation, show therapeutic potential for the treatment of EDS in patients with hypersomnia. This randomized, placebo-controlled, phase 1b study aimed to investigate the safety of danavorexton, a small-molecule orexin-2 receptor agonist, and its effects on residual EDS in patients with OSA. METHODS Adults with OSA aged 18-67 years with adequate CPAP use were randomized to one of six treatment sequences of single IV infusions of danavorexton 44 mg, danavorexton 112 mg, and placebo. Adverse events were monitored throughout the study. Pharmacodynamic assessments included maintenance of wakefulness test (MWT), Karolinska Sleepiness Scale (KSS), and the psychomotor vigilance test (PVT). RESULTS AND CONCLUSION Among 25 randomized patients, 16 (64.0%) had treatment-emergent adverse events (TEAEs) and 12 (48.0%) had TEAEs considered related to treatment, all mild or moderate. Seven patients (28.0%) had urinary TEAEs: three, seven, and none while taking danavorexton 44 mg, danavorexton 112 mg, and placebo, respectively. There were no deaths or TEAEs leading to discontinuation. Improvements in mean MWT, KSS, and PVT scores were observed with danavorexton 44 mg and 112 mg vs placebo. These findings show that danavorexton can improve subjective and objective measures of EDS in patients with OSA and residual EDS despite adequate CPAP use.
Collapse
Affiliation(s)
- Richard K Bogan
- University of South Carolina School of Medicine, Columbia, SC, USA.
| | | | - Rachel Neuwirth
- Takeda Development Center Americas, Inc., Lexington, MA, USA.
| | - Hélène Faessel
- Takeda Development Center Americas, Inc., Lexington, MA, USA.
| | - Todd Swick
- Takeda Development Center Americas, Inc., Lexington, MA, USA.
| | - Tina Olsson
- Takeda Development Center Americas, Inc., Lexington, MA, USA.
| |
Collapse
|
235
|
Durairaja A, Pandey S, Kahl E, Fendt M. Nasal administration of orexin A partially rescues dizocilpine-induced cognitive impairments in female C57BL/6J mice. Behav Brain Res 2023; 450:114491. [PMID: 37172740 DOI: 10.1016/j.bbr.2023.114491] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Sex difference has been reported in several behavioural endophenotypes of neuropsychiatric disorder in both rodents and humans. However, sex difference in cognitive symptoms associated with neuropsychiatric disorders has not been studied in detail. In this study, we induced cognitive impairment using the NMDA receptor antagonist, dizocilpine (MK-801), in male and female C57BL/6J mice and performed a visual discrimination task in an automated touchscreen system. We found that discrimination performance decreased with increased doses of MK-801 in both sexes. However, female mice showed stronger deficit in discrimination performance than the male mice especially after administration of low (0.01mg/kg) and high (0.15mg/kg) doses of MK-801. Furthermore, we tested if administration of orexin A, orexin-1 receptor antagonist SB-334867 or orexin-2 receptor antagonist EMPA rescued MK-801 (0.15mg/kg) induced cognitive impairment in visual discrimination. We found that nasal administration of orexin A partially rescued the cognitive impairment induced by MK-801 in females but not in males. Taken together, our data show that female C57BL/6J mice are more sensitive compared to males to some doses of MK-801 in discrimination learning task and that orexin A partially rescues this cognitive impairment in females.
Collapse
Affiliation(s)
- Archana Durairaja
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany.
| | - Samiksha Pandey
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany; Integrative Neuroscience Programme, Otto-von-Guericke University, Magdeburg, Germany
| | - Evelyn Kahl
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany; Center of Behavioral Brain Sciences, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
236
|
Leino TO, Turku A, Urvas L, Adhikari K, Oksanen J, Steynen Y, Yli-Kauhaluoma J, Xhaard H, Kukkonen JP, Wallén EAA. Azulene as a biphenyl mimetic in orexin/hypocretin receptor agonists. Bioorg Med Chem 2023; 88-89:117325. [PMID: 37209639 DOI: 10.1016/j.bmc.2023.117325] [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: 02/02/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/22/2023]
Abstract
Azulene is a rare ring structure in drugs, and we investigated whether it could be used as a biphenyl mimetic in known orexin receptor agonist Nag 26, which is binding to both orexin receptors OX1 and OX2 with preference towards OX2. The most potent azulene-based compound was identified as an OX1 orexin receptor agonist (pEC50 = 5.79 ± 0.07, maximum response = 81 ± 8% (s.e.m. of five independent experiments) of the maximum response to orexin-A in Ca2+ elevation assay). However, the azulene ring and the biphenyl scaffold are not identical in their spatial shape and electron distribution, and their derivatives may adopt different binding modes in the binding site.
Collapse
Affiliation(s)
- Teppo O Leino
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland; Department of Chemistry and NanoScience Center, University of Jyväskylä, P.O. Box 35, FI-40014 University of Jyväskylä, Finland.
| | - Ainoleena Turku
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland; Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 66, FI-00014 University of Helsinki, Finland
| | - Lauri Urvas
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland; Department of Pharmacology, Faculty of Medicine, University of Helsinki, P.O. Box 63, FI-00014 University of Helsinki, Finland
| | - Karuna Adhikari
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Jouni Oksanen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Yana Steynen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Henri Xhaard
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Jyrki P Kukkonen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 66, FI-00014 University of Helsinki, Finland; Department of Pharmacology, Faculty of Medicine, University of Helsinki, P.O. Box 63, FI-00014 University of Helsinki, Finland
| | - Erik A A Wallén
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| |
Collapse
|
237
|
Alrouji M, Al-Kuraishy HM, Al-Gareeb AI, Zaafar D, Batiha GES. Orexin pathway in Parkinson's disease: a review. Mol Biol Rep 2023:10.1007/s11033-023-08459-5. [PMID: 37155018 DOI: 10.1007/s11033-023-08459-5] [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: 02/23/2023] [Accepted: 04/13/2023] [Indexed: 05/10/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease (NDD) caused by dopaminergic neuron degeneration in the substantia nigra (SN). Orexin is a neuropeptide that plays a role in the pathogenesis of PD. Orexin has neuroprotective properties in dopaminergic neurons. In PD neuropathology, there is also degeneration of orexinergic neurons in the hypothalamus, in addition to dopaminergic neurons. However, the loss of orexinergic neurons in PD began after the degeneration of dopaminergic neurons. Reduced activity of orexinergic neurons has been linked to developing and progressing motor and non-motor symptoms in PD. In addition, the dysregulation of the orexin pathway is linked to the development of sleep disorders. The hypothalamic orexin pathway regulates various aspects of PD neuropathology at the cellular, subcellular, and molecular levels. Finally, non-motor symptoms, particularly insomnia and disturbed sleep, promote neuroinflammation and the accumulation of neurotoxic proteins as a result of defects in autophagy, endoplasmic reticulum (ER) stress, and the glymphatic system. As a result, this review aimed to highlight the potential role of orexin in PD neuropathology.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra, 11961, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of clinical pharmacology and therapeutic medicine, college of medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of clinical pharmacology and therapeutic medicine, college of medicine, Mustansiriyah University, Baghdad, Iraq
| | - Dalia Zaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Al Beheira, Egypt.
| |
Collapse
|
238
|
Wright CJ, Milosavljevic S, Pocivavsek A. The stress of losing sleep: Sex-specific neurobiological outcomes. Neurobiol Stress 2023; 24:100543. [PMID: 37252645 PMCID: PMC10209346 DOI: 10.1016/j.ynstr.2023.100543] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/20/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023] Open
Abstract
Sleep is a vital and evolutionarily conserved process, critical to daily functioning and homeostatic balance. Losing sleep is inherently stressful and leads to numerous detrimental physiological outcomes. Despite sleep disturbances affecting everyone, women and female rodents are often excluded or underrepresented in clinical and pre-clinical studies. Advancing our understanding of the role of biological sex in the responses to sleep loss stands to greatly improve our ability to understand and treat health consequences of insufficient sleep. As such, this review discusses sex differences in response to sleep deprivation, with a focus on the sympathetic nervous system stress response and activation of the hypothalamic-pituitary-adrenal (HPA) axis. We review sex differences in several stress-related consequences of sleep loss, including inflammation, learning and memory deficits, and mood related changes. Focusing on women's health, we discuss the effects of sleep deprivation during the peripartum period. In closing, we present neurobiological mechanisms, including the contribution of sex hormones, orexins, circadian timing systems, and astrocytic neuromodulation, that may underlie potential sex differences in sleep deprivation responses.
Collapse
Affiliation(s)
- Courtney J. Wright
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Snezana Milosavljevic
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
239
|
Modaberi S, Amirteymori H, Mesgar S, Eskandari K, Haghparast A. The blockade of orexin receptors within the dentate gyrus of the hippocampus attenuated methamphetamine-induced reward learning during conditioning place preference. Pharmacol Biochem Behav 2023; 226:173559. [PMID: 37100179 DOI: 10.1016/j.pbb.2023.173559] [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: 12/06/2022] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/28/2023]
Abstract
Orexins and orexinergic receptors have been shown to play a critical role in reward processing and drug addiction. Previous studies showed that the orexinergic system in the dentate gyrus (DG) region of the hippocampus affects the conditioning (acquisition) and post-conditioning (expression) phases of morphine-induced conditioned place preference (CPP). The action of each orexin receptor within the DG during conditioning and expression phases for methamphetamine (METH)-induced CPP remains unclear. The present study aimed to determine the role of orexin-1 and -2 receptors in the hippocampal DG in METH CPP acquisition and expression. During the 5-day conditioning phase, rats received an intra-DG microinjection of SB334867, a selective orexin-1 receptor (OX1R) antagonist, or TCS OX2-29, a selective orexin-2 receptor (OX2R) antagonist, before injection of METH (1 mg/kg; sc). In different sets of animals on the expression day, rats received each antagonist before the CPP test. The results showed that SB334867 (3, 10, and 30 nmol) and TCS OX2-29 (3, 10, and 30 nmol) significantly decreased the acquisition of METH CPP during the conditioning phase. Furthermore, administration of SB 334867 (10 and 30 nmol) and TCS OX2-29 (3 and 10 nmol) on the post-conditioning day significantly reduced METH-induced CPP expression. The results also indicated that orexin receptors play a more critical role in the conditioning phase than in the expression phase. In summary, the orexin receptors in the DG play a crucial role in drug learning and memory and are essential for METH reward acquisition and expression.
Collapse
Affiliation(s)
- Shaghayegh Modaberi
- Department of Sport Sciences, Faculty of Social Sciences, Imam Khomeini International University, Qazvin, Iran
| | - Haleh Amirteymori
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somaye Mesgar
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kiarash Eskandari
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran; Department of Basic Sciences, Iranian Academy of Medical Sciences, Tehran, Iran.
| |
Collapse
|
240
|
Reddy I, Yadav Y, Dey CS. Cellular and Molecular Regulation of Exercise-A Neuronal Perspective. Cell Mol Neurobiol 2023; 43:1551-1571. [PMID: 35986789 PMCID: PMC11412429 DOI: 10.1007/s10571-022-01272-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022]
Abstract
The beneficial effects of exercise on the proper functioning of the body have been firmly established. Multi-systemic metabolic regulation of exercise is the consequence of multitudinous changes that occur at the cellular level. The exercise responsome comprises all molecular entities including exerkines, miRNA species, growth factors, signaling proteins that are elevated and activated by physical exercise. Exerkines are secretory molecules released by organs such as skeletal muscle, adipose tissue, liver, and gut as a function of acute/chronic exercise. Exerkines such as FNDC5/irisin, Cathepsin B, Adiponectin, and IL-6 circulate through the bloodstream, cross the blood-brain barrier, and modulate the expression of important signaling molecules such as AMPK, SIRT1, PGC1α, BDNF, IGF-1, and VEGF which further contribute to improved energy metabolism, glucose homeostasis, insulin sensitivity, neurogenesis, synaptic plasticity, and overall well-being of the body and brain. These molecules are also responsible for neuroprotective adaptations that exercise confers on the brain and potentially ameliorate neurodegeneration. This review aims to detail important cellular and molecular species that directly or indirectly mediate exercise-induced benefits in the body, with an emphasis on the central nervous system.
Collapse
Affiliation(s)
- Ishitha Reddy
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India
| | - Yamini Yadav
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India.
| |
Collapse
|
241
|
Abstract
Narcolepsy is a sleep disorder caused by insufficient levels of orexins, with current treatment options addressing symptoms rather than etiology. New research reveals that transplantation of orexin cells in a mouse model of narcolepsy reduces cataplexy.
Collapse
Affiliation(s)
- Giancarlo Vanini
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48109, USA; Center for Consciousness Science, University of Michigan, Ann Arbor, MI 48109, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA. gvanini,@,umich.edu
| |
Collapse
|
242
|
Maruyama T, Ueta Y. Internal and external modulation factors of the orexin system (REVIEW). Peptides 2023; 165:171009. [PMID: 37054895 DOI: 10.1016/j.peptides.2023.171009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/15/2023]
Abstract
Orexin-A and -B (identical to hypocretin-1 and -2) are neuropeptides synthesized in the lateral hypothalamus and perifornical area, and orexin neurons project their axon terminals broadly throughout the entire central nervous system (CNS). The activity of orexins is mediated by two specific G protein-coupled receptors (GPCRs), termed orexin type1 receptor (OX1R) and orexin type2 receptor (OX2R). The orexin system plays a relevant role in various physiological functions, including arousal, feeding, reward, and thermogenesis, and is key to human health. Orexin neurons receive various signals related to environmental, physiological, and emotional stimuli. Previous studies have reported that several neurotransmitters and neuromodulators influence the activation or inhibition of orexin neuron activity. In this review, we summarize the modulating factors of orexin neurons in the sleep/wake rhythm and feeding behavior, particularly in the context of the modulation of appetite, body fluids, and circadian signaling. We also describe the effects of life activity, behavior, and diet on the orexin system. Some studies have observed phenomena that have been verified in animal experiments, revealing the detailed mechanism and neural pathway, while their applications to humans is expected in future research.
Collapse
Affiliation(s)
- Takashi Maruyama
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Japan.
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Japan
| |
Collapse
|
243
|
Abstract
The recently uncovered key role of the peripheral and central nervous systems in controlling tumorigenesis and metastasis has opened a new area of research to identify innovative approaches against cancer. Although the 'neural addiction' of cancer is only partially understood, in this Perspective we discuss the current knowledge and perspectives on peripheral and central nerve circuitries and brain areas that can support tumorigenesis and metastasis and the possible reciprocal influence that the brain and peripheral tumours exert on one another. Tumours can build up local autonomic and sensory nerve networks and are able to develop a long-distance relationship with the brain through circulating adipokines, inflammatory cytokines, neurotrophic factors or afferent nerve inputs, to promote cancer initiation, growth and dissemination. In turn, the central nervous system can affect tumour development and metastasis through the activation or dysregulation of specific central neural areas or circuits, as well as neuroendocrine, neuroimmune or neurovascular systems. Studying neural circuitries in the brain and tumours, as well as understanding how the brain communicates with the tumour or how intratumour nerves interplay with the tumour microenvironment, can reveal unrecognized mechanisms that promote cancer development and progression and open up opportunities for the development of novel therapeutic strategies. Targeting the dysregulated peripheral and central nervous systems might represent a novel strategy for next-generation cancer treatment that could, in part, be achieved through the repurposing of neuropsychiatric drugs in oncology.
Collapse
Affiliation(s)
- Claire Magnon
- Laboratory of Cancer and Microenvironment-National Institute of Health and Medical Research (INSERM), Institute of Biology François Jacob-Atomic Energy Commission (CEA), University of Paris Cité, University of Paris-Saclay, Paris, France.
| | - Hubert Hondermarck
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
244
|
Ramser A, Dridi S. Hormonal regulation of visfatin and adiponectin system in quail muscle cells. Comp Biochem Physiol A Mol Integr Physiol 2023; 281:111425. [PMID: 37044369 DOI: 10.1016/j.cbpa.2023.111425] [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: 03/23/2023] [Revised: 04/08/2023] [Accepted: 04/08/2023] [Indexed: 04/14/2023]
Abstract
Visfatin and adiponectin are two adipokines known to regulate energy homeostasis and stress response within different peripheral tissues. Their role and regulation in highly metabolically active tissue such as the muscle is of particular interest. As modern poultry exhibit insulin resistance, obesity, and hyperglycemia along with a lack of insight into the regulation of these avian adipokines, we undertook the present work to determine the regulation of visfatin and adiponectin system by cytokines and obesity-related hormones in a relevant in vitro model of avian muscle, quail muscle (QM7) cells. Cells were treated with pro-inflammatory cytokine IL-6 (5 and 10 ng/mL) and TNFα (5 and 10 ng/mL), as well as leptin (10 and 100 ng/mL) and both orexin-A and orexin-B (ORX-A/B) (5 and 10 ng/mL). Results showed significant increases in visfatin mRNA abundance under both cytokines (IL-6 and TNFα), and down regulation with ORX-B treatment. Adiponectin expression was also upregulated by pro-inflammatory cytokines (IL-6 and TNFα), but down regulated by leptin, ORX-A, and ORXB. High doses of IL-6 and TNFα up regulated the expression of adiponectin receptors AdipoR1 and AdipoR2, respectively. Leptin and orexin treatments also down regulated both AdipoR1 and AdipoR2 expression. Taken together, this is the first report showing a direct response of visfatin and the adiponectin system to pro-inflammatory and obesity-related hormones in avian muscle cells.
Collapse
Affiliation(s)
- Alison Ramser
- University of Arkansas, Center of Excellence for Poultry Science, Fayetteville, AR 72701, USA
| | - Sami Dridi
- University of Arkansas, Center of Excellence for Poultry Science, Fayetteville, AR 72701, USA.
| |
Collapse
|
245
|
Pintwala SK, Fraigne JJ, Belsham DD, Peever JH. Immortal orexin cell transplants restore motor-arousal synchrony during cataplexy. Curr Biol 2023; 33:1550-1564.e5. [PMID: 37044089 DOI: 10.1016/j.cub.2023.03.077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/11/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023]
Abstract
Waking behaviors such as sitting or standing require suitable levels of muscle tone. But it is unclear how arousal and motor circuits communicate with one another so that appropriate motor tone occurs during wakefulness. Cataplexy is a peculiar condition in which muscle tone is involuntarily lost during normal periods of wakefulness. Cataplexy therefore provides a unique opportunity for identifying the signaling mechanisms that synchronize motor and arousal behaviors. Cataplexy occurs when hypothalamic orexin neurons are lost in narcolepsy; however, it is unclear if motor-arousal decoupling in cataplexy is directly or indirectly caused by orexin cell loss. Here, we used genomic, proteomic, chemogenetic, electrophysiological, and behavioral assays to determine if grafting orexin cells into the brain of cataplectic (i.e., orexin-/-) mice restores normal motor-arousal behaviors by preventing cataplexy. First, we engineered immortalized orexin cells and found that they not only produce and release orexin but also exhibit a gene profile that mimics native orexin neurons. Second, we show that engineered orexin cells thrive and integrate into host tissue when transplanted into the brain of mice. Next, we found that grafting only 200-300 orexin cells into the dorsal raphe nucleus-a region densely innervated by native orexin neurons-reduces cataplexy. Last, we show that real-time chemogenetic activation of orexin cells restores motor-arousal synchrony by preventing cataplexy. We suggest that orexin signaling is critical for arousal-motor synchrony during wakefulness and that the dorsal raphe plays a pivotal role in coupling arousal and motor behaviors.
Collapse
Affiliation(s)
- Sara K Pintwala
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jimmy J Fraigne
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Denise D Belsham
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - John H Peever
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
246
|
Hung C, Yamanaka A. The role of orexin neuron activity in sleep/wakefulness regulation. Peptides 2023; 165:171007. [PMID: 37030519 DOI: 10.1016/j.peptides.2023.171007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 04/10/2023]
Abstract
Orexin (also known as hypocretin) is a neuropeptide exclusively synthesized in the neurons of the lateral hypothalamus (LH). Initially orexin was thought to be involved in the regulation of feeding behavior. However, it is now known to also be a critical regulator of sleep/wakefulness, especially the maintenance of wakefulness. Although the somas of orexin neurons are exclusively located in the LH, these neurons send axons throughout the brain and spinal cord. Orexin neurons integrate inputs from various brain regions and project to neurons that are involved in the regulation of sleep/wakefulness. Orexin knockout mice have a fragmentation of sleep/wakefulness and cataplexy-like behavior arrest, which is similar to the sleep disorder narcolepsy. Recent progress with manipulation of neural activity of targeted neurons, using experimental tools such as optogenetics and chemogenetics, has emphasized the role of orexin neuron activity on the regulation of sleep/wakefulness. Recording of orexin neuron activity in vivo using electrophysiological and gene-encoded calcium indicator proteins revealed that these cells have specific activity patterns across sleep/wakefulness state changes. Here, we also discuss not only the role of the orexin peptide, but also the role of other co-transmitters that are synthesized and released from orexin neurons and involved in sleep/wakefulness regulation.
Collapse
Affiliation(s)
- Chijung Hung
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Akihiro Yamanaka
- Chinese Institute for Brain Research, Beijing (CIBR), Beijing, 102206, China; National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8585 Japan; Division of Brain Sciences Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| |
Collapse
|
247
|
He J, Fang J, Wang Y, Ge C, Liu S, Jiang Y. Discovery of Small-Molecule Antagonists of Orexin 1/2 Receptors from Traditional Chinese Medicinal Plants with a Hypnotic Effect. Pharmaceuticals (Basel) 2023; 16:ph16040542. [PMID: 37111298 PMCID: PMC10146837 DOI: 10.3390/ph16040542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/25/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
Insomnia is an important public health problem. The currently available treatments for insomnia can cause some adverse effects. Orexin receptors 1 (OX1R) and 2 (OX2R) are burgeoning targets for insomnia treatment. It is an effective approach to screening OX1R and OX2R antagonists from traditional Chinese medicine, which contains abundant and diverse chemical components. This study established an in-home ligand library of small-molecule compounds from medicinal plants with a definite hypnotic effect, as described in the Chinese Pharmacopoeia. Molecular docking was applied to virtually screen potential orexin receptor antagonists using molecular operating environment software, and surface plasmon resonance (SPR) technology was used to detect the binding affinity between potential active compounds and orexin receptors. Finally, the results of virtual screening and SPR analysis were verified through in vitro assays. We successfully screened one potential lead compound (neferine) as an orexin receptor antagonist from the in-home ligand library, which contained more than 1000 compounds. The screened compound was validated as a potential agent for insomnia treatment through comprehensive biological assays. This research enabled the discovery of a potential small-molecule antagonist of orexin receptors for the treatment of insomnia, providing a novel screening approach for the detection of potential candidate compounds for corresponding targets.
Collapse
Affiliation(s)
- Jia He
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jing Fang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yuxin Wang
- College of Pharmacy, Dali University, Dali 671000, China
| | - Chengyu Ge
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shao Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yueping Jiang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
248
|
Ten-Blanco M, Flores Á, Cristino L, Pereda-Pérez I, Berrendero F. Targeting the orexin/hypocretin system for the treatment of neuropsychiatric and neurodegenerative diseases: from animal to clinical studies. Front Neuroendocrinol 2023; 69:101066. [PMID: 37015302 DOI: 10.1016/j.yfrne.2023.101066] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/15/2023] [Accepted: 03/30/2023] [Indexed: 04/06/2023]
Abstract
Orexins (also known as hypocretins) are neuropeptides located exclusively in hypothalamic neurons that have extensive projections throughout the central nervous system and bind two different G protein-coupled receptors (OX1R and OX2R). Since its discovery in 1998, the orexin system has gained the interest of the scientific community as a potential therapeutic target for the treatment of different pathological conditions. Considering previous basic science research, a dual orexin receptor antagonist, suvorexant, was the first orexin agent to be approved by the US Food and Drug Administration to treat insomnia. In this review, we discuss and update the main preclinical and human studies involving the orexin system with several psychiatric and neurodegenerative diseases. This system constitutes a nice example of how basic scientific research driven by curiosity can be the best route to the generation of new and powerful pharmacological treatments.
Collapse
Affiliation(s)
- Marc Ten-Blanco
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - África Flores
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Neurosciences Institute, University of Barcelona and Bellvitge University Hospital-IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Luigia Cristino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Inmaculada Pereda-Pérez
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Fernando Berrendero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain.
| |
Collapse
|
249
|
Moteshakereh SM, Nikoohemmat M, Farmani D, Khosrowabadi E, Salehi S, Haghparast A. The stress-induced antinociceptive responses to the persistent inflammatory pain involve the orexin receptors in the nucleus accumbens. Neuropeptides 2023; 98:102323. [PMID: 36736068 DOI: 10.1016/j.npep.2023.102323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/16/2022] [Accepted: 01/26/2023] [Indexed: 01/30/2023]
Abstract
Stress suppresses the sense of pain, a physiological phenomenon known as stress-induced analgesia (SIA). Brain orexin peptides regulate many physiological functions, including wakefulness and nociception. The contribution of the orexinergic system within the nucleus accumbens (NAc) in the modulation of antinociception induced by forced swim stress (FSS) remains unclear. The present study addressed the role of intra-accumbal orexin receptors in the antinociceptive responses induced by FSS during the persistent inflammatory pain model in the rat. Stereotaxic surgery was performed unilaterally on 106 adult male Wistar rats weighing 250-305 g. Different doses (1, 3, 10, and 30 nmol/ 0.5 μl DMSO) of orexin-1 receptor (OX1r) antagonist (SB334867) or OX2 receptor antagonist (TCS OX2 29) were administered into the NAc five minutes before exposure to FSS for a 6-min period. The formalin test was carried out using formalin injection (50 μl; 2.5%) into the rat's hind paw plantar surface, which induces biphasic pain-related responses. The first phase begins immediately after formalin infusion and takes 3-5 min. Subsequently, the late phase begins 15-20 min after formalin injection and takes 20-40 min. The findings demonstrated that intra-accumbal microinjection of SB334867 or TCS OX2 29 attenuated the FSS-induced antinociception in both phases of the formalin test, with the TCS OX2 29 showing higher potency. Moreover, the effect of TCS OX2 29 was more significant during the early phase of the formalin test. The results suggest that OX1 and OX2 receptors in the NAc might modulate the antinociceptive responses induced by the FSS.
Collapse
Affiliation(s)
| | - Mohammad Nikoohemmat
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Danial Farmani
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Khosrowabadi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sakineh Salehi
- epartment of Medicine, Ardabil Medical Sciences Branch, Islamic Azad University, Ardabil, Iran.
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
250
|
Bateman JT, Saunders SE, Levitt ES. Understanding and countering opioid-induced respiratory depression. Br J Pharmacol 2023; 180:813-828. [PMID: 34089181 PMCID: PMC8997313 DOI: 10.1111/bph.15580] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/06/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
Respiratory depression is the proximal cause of death in opioid overdose, yet the mechanisms underlying this potentially fatal outcome are not well understood. The goal of this review is to provide a comprehensive understanding of the pharmacological mechanisms of opioid-induced respiratory depression, which could lead to improved therapeutic options to counter opioid overdose, as well as other detrimental effects of opioids on breathing. The development of tolerance in the respiratory system is also discussed, as are differences in the degree of respiratory depression caused by various opioid agonists. Finally, potential future therapeutic agents aimed at reversing or avoiding opioid-induced respiratory depression through non-opioid receptor targets are in development and could provide certain advantages over naloxone. By providing an overview of mechanisms and effects of opioids in the respiratory network, this review will benefit future research on countering opioid-induced respiratory depression. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Affiliation(s)
- Jordan T Bateman
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, Florida, USA
| | - Sandy E Saunders
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, Florida, USA
| | - Erica S Levitt
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, Florida, USA
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA
| |
Collapse
|