1
|
Cui P, Wu D, Dong J, Chen Y, Feng J, Liu X, Chen Z, Ge J, Liu J. Effects of inhibiting the orexin system on nicotine reward memory and fear memory. Neuropharmacology 2025; 276:110523. [PMID: 40398734 DOI: 10.1016/j.neuropharm.2025.110523] [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: 03/05/2025] [Revised: 04/14/2025] [Accepted: 05/16/2025] [Indexed: 05/23/2025]
Abstract
The orexin system is associated with various brain functions, including reward processing and the stress response. Previous studies have suggested that orexin receptors (OXRs) antagonists could be beneficial in treating several mental disorders, such as drug addiction and post-traumatic stress disorder. However, the specific role of the orexin system in both appetitive (reward-related) and aversive (fear-related) memories remains unclear. In this study, we used nicotine-induced conditioned place preference (CPP) and fear conditioning models to evaluate the effects of suvorexant, a dual OXRs antagonist, and chemogenetic inhibition of orexin neurons in the lateral hypothalamus (LHA) on the retrieval, reconsolidation, and/or extinction of both nicotine reward memory and fear memory. Our findings indicated that suvorexant impaired the expression of nicotine CPP in a dose-dependent manner. These behavioral results aligned with neurochemical evidence from cFos staining, which showed activation of the LHA and orexin neurons in the LHA following the expression of nicotine CPP. Furthermore, using orexin-cre rats, we found that chemogenetic inhibition of orexin neurons in the LHA also reduced the expression of nicotine CPP. However, neither of these interventions affected the reconsolidation of nicotine reward memory. Moreover, neither suvorexant nor the chemogenetic inhibition of LHA orexin neurons affected the retrieval, reconsolidation, or extinction of fear memory, or anxiety-like behaviors, as evaluated by elevated plus maze and open field tests. In summary, our data suggest that the orexin system plays a crucial role in retrieving nicotine reward memory, and suggest that suvorexant may be a promising pharmacotherapy for treating nicotine addiction.
Collapse
Affiliation(s)
- Pan Cui
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, China
| | - Danping Wu
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, China
| | - Jing Dong
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, China
| | - Yun Chen
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, China
| | - Jialu Feng
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, China
| | - Xiaoliu Liu
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, China
| | - Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Jing Ge
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, China.
| | - Jianfeng Liu
- School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, China.
| |
Collapse
|
2
|
Wall EK, Virakorn EA, Baker KD, Cohen EM, Richardson R. Preclinical behavioral and pharmacological treatments for enhancing fear extinction in adolescence. Neurosci Biobehav Rev 2025; 172:106090. [PMID: 40049540 DOI: 10.1016/j.neubiorev.2025.106090] [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: 01/09/2025] [Revised: 02/17/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Adolescence is a window of vulnerability for the development of anxiety disorders but also a window of opportunity for treatments to minimize the long-term impact of such disorders. Current first-line treatments, primarily exposure-based cognitive-behavioral therapy (CBT), have limited long-term efficacy in adolescents. The urgent need for more effective interventions is underscored by the frequent reports of extinction impairments in adolescents as well as the rising anxiety rates in youth, particularly post-COVID-19. Preclinical research on the extinction of learned fear in adolescents may contribute to developing better treatment approaches to anxiety in this age group. Unfortunately, this is still a largely under-explored area. However, both pharmacological and behavioral augmentation strategies can be used to enhance extinction learning and consolidation. Here we describe work exploring such adjuncts, focusing on pre-clinical work with rodents. Much of the research to date shows striking developmental differences in response to various pharmacological treatments, with only a few shown to be effective in adolescents. Further, recent experience of stress reduces the efficacy of these treatments in adolescence. This review highlights the necessity for tailored strategies, especially when it comes to pharmacological adjuncts, that address developmental differences in drug responses as well as the impact of stressful experiences on treatment efficacy.
Collapse
Affiliation(s)
- Emily K Wall
- School of Psychology, UNSW Sydney, Sydney, NSW 2052, Australia
| | | | - Kathryn D Baker
- School of Psychology, UNSW Sydney, Sydney, NSW 2052, Australia; Department of Psychology, Counselling and Therapy, La Trobe University, Melbourne, VIC 3086, Australia
| | - E Myfanwy Cohen
- School of Psychology, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Rick Richardson
- School of Psychology, UNSW Sydney, Sydney, NSW 2052, Australia.
| |
Collapse
|
3
|
Messore F, Narayanan Therpurakal R, Dufour JP, Hoerder-Suabedissen A, Guidi L, Korrell K, Mueller M, Abuelem M, Lak A, Bannerman DM, Mann EO, Molnár Z. An orexin-sensitive subpopulation of layer 6 neurons regulates cortical excitability and anxiety behaviour. Transl Psychiatry 2025; 15:147. [PMID: 40229262 PMCID: PMC11997144 DOI: 10.1038/s41398-025-03350-2] [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: 10/11/2024] [Revised: 02/26/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025] Open
Abstract
Cortical layer 6 neurons are the only projection neuron population in the cortical mantle known to electrophysiologically respond to orexin-a neuropeptide involved in cortical arousal and emotive behaviour. These neurons exhibit extensive intercortical and thalamic projections, yet the exact mechanisms underlying these responses are not fully understood. We hypothesize that cortical circuits activated by orexin sensitive L6 neurons in the medial prefrontal cortex (mPFC) are responsible for detecting salient features of sensory stimuli and are therefore involved in regulating emotional states. Here, we show that Drd1a-Cre+ neurons in the mPFC are selectively sensitive to orexin and gate the activation of the prefrontal network in vivo. Moreover, we demonstrated that chronically "silencing" this subpopulation of L6 neurons (Drd1a-Cre+/+:Snap25fl/fl) across the cortical mantle from birth abolishes the orexin-induced prefrontal activation. Consequently, the chronic silencing of these neurons had strong anxiolytic effects on several anxiety-related behavioural paradigms, indicating that orexin-responsive L6 neurons modulate emotional states and may be a substrate for anxiety regulation.
Collapse
Affiliation(s)
- Fernando Messore
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
| | - Rajeevan Narayanan Therpurakal
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Jean-Philippe Dufour
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
| | | | - Luiz Guidi
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
| | - Kim Korrell
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
| | - Marissa Mueller
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
| | - Mohammed Abuelem
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Armin Lak
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Edward O Mann
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA.
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics University of Oxford, Oxford, USA.
| |
Collapse
|
4
|
Chaki S. Orexin receptors: possible therapeutic targets for psychiatric disorders. Psychopharmacology (Berl) 2025:10.1007/s00213-025-06767-1. [PMID: 40153060 DOI: 10.1007/s00213-025-06767-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/30/2025]
Abstract
RATIONALE Orexins, comprising orexin-A and orexin-B, are neuropeptides with extensive projections throughout the central nervous system. They are implicated in a variety of physiological processes through their receptors, orexin type 1 (OX1) and orexin type 2 (OX2) receptors. Among the physiological functions of orexins, their role in sleep/wake regulation has garnered significant attention. Consequently, three orexin receptor antagonists that block both OX1 and OX2 receptors (dual orexin receptor antagonist; DORA) are available on the market for the treatment of insomnia. Additionally, another DORA, vornorexant, has been submitted for approval. OBJECTIVE Beyond sleep disorders, the orexin system is deeply implicated in the pathophysiology of several psychiatric disorders, including depression, anxiety, and substance use disorders. RESULTS Accumulating evidence indicates that orexin receptor antagonists improve behavioral abnormalities that mimic certain psychiatric disorders in animal models and are effective in treating these disorders or their symptoms in humans. Moreover, orexin receptor antagonists are expected not only to alleviate core symptoms of psychiatric disorders but also to improve sleep disturbances, which are often comorbid with these conditions. CONCLUSION Drug discovery and development targeting orexin receptors should provide novel therapeutic options for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Shigeyuki Chaki
- Taisho Pharmaceutical Co., Ltd, Toshima-Ku, Tokyo, 170-8633, Japan.
- Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| |
Collapse
|
5
|
Oliveira GVM, Hernandes PM, Santos FHD, Soares VPMN, Falconi-Sobrinho LL, Coimbra NC, Wotjak CT, Almada RC. Orexin mechanisms in the prelimbic cortex modulate the expression of contextual conditioned fear. Psychopharmacology (Berl) 2025; 242:521-532. [PMID: 39387863 DOI: 10.1007/s00213-024-06701-x] [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: 12/28/2023] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
RATIONALE Despite the existing anatomical and physiological evidence pointing to the involvement of orexinergic projections from the lateral hypothalamus (LH) in regulating fear-related responses, little is known regarding the contribution of the orexin system in the prelimbic cortex (PL) on contextual fear. OBJECTIVES We investigated the role of orexin-A (OrxA) and orexin type 1 receptors (Orx1R) in the PL during the expression of contextual conditioned fear in mice. METHODS Neural tract tracing of the LH-PL pathway and Orx1R immunoreactivity in the PL of C57BL/6 male mice were performed. In a pharmacological approach, the animals were treated with either the Orx1R selective antagonist SB 334,867 (3, 30, and 300 nM/0.1 µL) or OrxA (28, 70, and 140 pmol/0.1 µL) in the PL before the test session of contextual fear conditioning. RESULTS Neural tract tracing deposits in the LH showed some perikarya, mainly axons and terminal buttons in the PL, suggesting LH-PL reciprocate pathways. Furthermore, we showed a profuse network comprised of Orx1R-labeled thin varicose fibers widely distributed in the same field of LH-PL pathways projection. The selective blockade of Orx1R with SB 334,867 at 30 and 300 nM in the PL caused a decrease in freezing response, whereas the treatment with OrxA at 140 pmol promoted an increase in freezing response. CONCLUSION In summary, these data confirmed an anatomical link between LH and PL, established the presence of Orx1R in the PL, and a modulatory role of the orexin system in such structure, possibly mainly via Orx1R, during contextual fear conditioning.
Collapse
Affiliation(s)
- Gabriela V M Oliveira
- Laboratory of Neurobiology and Neurobiotechnology, Department of Biological Sciences, School of Sciences, Humanities and Languages of the São Paulo State University (Unesp), Assis, São Paulo, 19806-900, Brazil
| | - Paloma M Hernandes
- Laboratory of Neurobiology and Neurobiotechnology, Department of Biological Sciences, School of Sciences, Humanities and Languages of the São Paulo State University (Unesp), Assis, São Paulo, 19806-900, Brazil
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Fábio H Dos Santos
- Laboratory of Neurobiology and Neurobiotechnology, Department of Biological Sciences, School of Sciences, Humanities and Languages of the São Paulo State University (Unesp), Assis, São Paulo, 19806-900, Brazil
| | - Victor P M N Soares
- Laboratory of Neurobiology and Neurobiotechnology, Department of Biological Sciences, School of Sciences, Humanities and Languages of the São Paulo State University (Unesp), Assis, São Paulo, 19806-900, Brazil
| | - Luiz Luciano Falconi-Sobrinho
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
- Behavioural Neuroscience Institute (INeC), Ribeirão Preto, São Paulo, Brazil
- NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto Medical School of the University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Norberto C Coimbra
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
- Behavioural Neuroscience Institute (INeC), Ribeirão Preto, São Paulo, Brazil
- NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto Medical School of the University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Carsten T Wotjak
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharmaceuticals Die Gesellschaft mit Beschränkter Haftung & Compagnie Kommanditgesellschaft, Biberach Riss, Germany
| | - Rafael Carvalho Almada
- Laboratory of Neurobiology and Neurobiotechnology, Department of Biological Sciences, School of Sciences, Humanities and Languages of the São Paulo State University (Unesp), Assis, São Paulo, 19806-900, Brazil.
- Behavioural Neuroscience Institute (INeC), Ribeirão Preto, São Paulo, Brazil.
- NAP-USP-Neurobiology of Emotions Research Centre (NuPNE), Ribeirão Preto Medical School of the University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
6
|
Kozłowska A, Ma WJ, Shyu BC, Huang ACW. Analysis of Anxiety Disorders and Post-Traumatic Stress Disorders for Screening Anxiolytic Drugs and Linking Preclinical and Clinical Research. Int J Mol Sci 2025; 26:1414. [PMID: 40003881 PMCID: PMC11855677 DOI: 10.3390/ijms26041414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/16/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
How to determine suitable animal models of anxiety disorders and post-traumatic stress disorders (PTSD) for screening anxiolytic drugs and linking preclinical and clinical research is a crucial issue. This review paper provides background knowledge and critical determination to the animal models for discussing this issue. Moreover, this article analyzes the characteristics, properties, advantages, and disadvantages of various animal models of anxiety disorders and PTSD. It offers an overview of the pathophysiology, treatments, prevalence, and symptoms of anxiety disorders in the clinics. Furthermore, it comprehensively discusses pharmacological treatments and neural mechanisms, as well as the types and properties of the animal models of anxiety disorders in shaping and testing anxiety behaviors. In light of the previous literature discussion, we can understand the different functions of the animal models of anxiety disorders and PTSD to help us link preclinical and clinical research. Animal models are used in advanced drug discovery programs, specifically in psychiatry and neuroscience research. The challenge for the future will be to keep pace with developing the appropriate animal models of anxiolytic drugs to improve the translation of large datasets obtained to clinics.
Collapse
Affiliation(s)
- Anna Kozłowska
- Department of Human Physiology and Pathophysiology, Collegium Medicum, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland;
| | - Wan-Jiun Ma
- Department of Psychology, Fo Guang University, Yilan County 26247, Taiwan;
| | - Bai-Chuang Shyu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan;
| | | |
Collapse
|
7
|
Zharova NV, Osadchiy AS, Lobanova AK, Isakova TA, Zharov NA, Zharikov YO, Pontes-Silva A, Zharikova TS. Functional Anatomy of the Structures of the Limbic System Involved in the Development of Neuropsychiatric Disorders: A Review. Curr Behav Neurosci Rep 2025; 12:1. [DOI: 10.1007/s40473-024-00291-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 05/03/2025]
|
8
|
Christensen J, Vlassopoulos E, Barlow CK, Schittenhelm RB, Li CN, Sgro M, Warren S, Semple BD, Yamakawa GR, Shultz SR, Mychasiuk R. The beneficial effects of modafinil administration on repeat mild traumatic brain injury (RmTBI) pathology in adolescent male rats are not dependent upon the orexinergic system. Exp Neurol 2024; 382:114969. [PMID: 39332798 DOI: 10.1016/j.expneurol.2024.114969] [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: 06/08/2024] [Revised: 08/22/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
Abstract
The sleep-wake cycle plays an influential role in the development and progression of repeat mild traumatic brain injury (RmTBI)-related pathology. Therefore, we first aimed to manipulate the sleep-wake cycle post-RmTBI using modafinil, a wake-promoting substance used for the treatment of narcolepsy. We hypothesized that modafinil would exacerbate RmTBI-induced deficits. Chronic behavioural analyses were completed along with a 27-plex serum cytokine array, metabolomic and proteomic analyses of cerebrospinal fluid (CSF), as well as immunohistochemical staining in structures important for sleep/wake cycles, to examine orexin, melanin-concentrating hormone, tyrosine hydroxylase, and choline acetyltransferase, in the lateral hypothalamus, locus coeruleus, and basal forebrain, respectively. Contrary to expectation, modafinil administration attenuated behavioural deficits, metabolomic changes, and neuropathological modifications. Therefore, the second aim was to determine if the beneficial effects of modafinil treatment were driven by the orexinergic system. The same experimental protocol was used; however, RmTBI rats received chronic orexin-A administration instead of modafinil. Orexin-A administration produced drastically different outcomes, exacerbating anxiety-related and motor deficits, while also significantly disrupting their metabolomic and neuropathological profiles. These results suggest that the beneficial effects of modafinil administration post-RmTBI, work independently of its wake-promoting properties, as activation of the orexinergic wake-promoting system with orexin-A was detrimental. Overall, these findings highlight the complexity of sleep-wake changes in the injured brain and showcase the potential of the arousal and sleep systems in its treatment.
Collapse
Affiliation(s)
- Jennaya Christensen
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Elaina Vlassopoulos
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Christopher K Barlow
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Crystal N Li
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Marissa Sgro
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Samantha Warren
- Monash Micro Imaging, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Bridgette D Semple
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Centre for Trauma and Mental Health Research, Vancouver Island University, Nanaimo, B.C., Canada
| | - Richelle Mychasiuk
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
9
|
Kukkonen JP, Jacobson LH, Hoyer D, Rinne MK, Borgland SL. International Union of Basic and Clinical Pharmacology CXIV: Orexin Receptor Function, Nomenclature and Pharmacology. Pharmacol Rev 2024; 76:625-688. [PMID: 38902035 DOI: 10.1124/pharmrev.123.000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
The orexin system consists of the peptide transmitters orexin-A and -B and the G protein-coupled orexin receptors OX1 and OX2 Orexin receptors are capable of coupling to all four families of heterotrimeric G proteins, and there are also other complex features of the orexin receptor signaling. The system was discovered 25 years ago and was immediately identified as a central regulator of sleep and wakefulness; this is exemplified by the symptomatology of the disorder narcolepsy with cataplexy, in which orexinergic neurons degenerate. Subsequent translation of these findings into drug discovery and development has resulted to date in three clinically used orexin receptor antagonists to treat insomnia. In addition to sleep and wakefulness, the orexin system appears to be a central player at least in addiction and reward, and has a role in depression, anxiety and pain gating. Additional antagonists and agonists are in development to treat, for instance, insomnia, narcolepsy with or without cataplexy and other disorders with excessive daytime sleepiness, depression with insomnia, anxiety, schizophrenia, as well as eating and substance use disorders. The orexin system has thus proved an important regulator of numerous neural functions and a valuable drug target. Orexin prepro-peptide and orexin receptors are also expressed outside the central nervous system, but their potential physiological roles there remain unknown. SIGNIFICANCE STATEMENT: The orexin system was discovered 25 years ago and immediately emerged as an essential sleep-wakefulness regulator. This discovery has tremendously increased the understanding of these processes and has thus far resulted in the market approval of three orexin receptor antagonists, which promote more physiological aspects of sleep than previous hypnotics. Further, orexin receptor agonists and antagonists with different pharmacodynamic properties are in development since research has revealed additional potential therapeutic indications. Orexin receptor signaling is complex and may represent novel features.
Collapse
Affiliation(s)
- Jyrki P Kukkonen
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Laura H Jacobson
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Daniel Hoyer
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Maiju K Rinne
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Stephanie L Borgland
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| |
Collapse
|
10
|
Jászberényi M, Thurzó B, Bagosi Z, Vécsei L, Tanaka M. The Orexin/Hypocretin System, the Peptidergic Regulator of Vigilance, Orchestrates Adaptation to Stress. Biomedicines 2024; 12:448. [PMID: 38398050 PMCID: PMC10886661 DOI: 10.3390/biomedicines12020448] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/10/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
The orexin/hypocretin neuropeptide family has emerged as a focal point of neuroscientific research following the discovery that this family plays a crucial role in a variety of physiological and behavioral processes. These neuropeptides serve as powerful neuromodulators, intricately shaping autonomic, endocrine, and behavioral responses across species. Notably, they serve as master regulators of vigilance and stress responses; however, their roles in food intake, metabolism, and thermoregulation appear complementary and warrant further investigation. This narrative review provides a journey through the evolution of our understanding of the orexin system, from its initial discovery to the promising progress made in developing orexin derivatives. It goes beyond conventional boundaries, striving to synthesize the multifaceted activities of orexins. Special emphasis is placed on domains such as stress response, fear, anxiety, and learning, in which the authors have contributed to the literature with original publications. This paper also overviews the advancement of orexin pharmacology, which has already yielded some promising successes, particularly in the treatment of sleep disorders.
Collapse
Affiliation(s)
- Miklós Jászberényi
- Department of Pathophysiology, University of Szeged, H-6701 Szeged, Hungary; (M.J.); (B.T.); (Z.B.)
| | - Balázs Thurzó
- Department of Pathophysiology, University of Szeged, H-6701 Szeged, Hungary; (M.J.); (B.T.); (Z.B.)
- Emergency Patient Care Unit, Albert Szent-Györgyi Health Centre, University of Szeged, H-6725 Szeged, Hungary
| | - Zsolt Bagosi
- Department of Pathophysiology, University of Szeged, H-6701 Szeged, Hungary; (M.J.); (B.T.); (Z.B.)
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, H-6725 Szeged, Hungary;
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Lee TY, Yi PL, Chang FC. Hypocretin role in posttraumatic stress disorder-like behaviors induced by a novel stress protocol in mice. Front Psychiatry 2023; 14:1196994. [PMID: 37457782 PMCID: PMC10343020 DOI: 10.3389/fpsyt.2023.1196994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Posttraumatic stress disorder (PTSD) is a psychiatric disorder developed in individuals who expose to traumatic events. These patients may experience symptoms, such as recurrent unwanted memory of the traumatic event, avoidance of reminders of the trauma, increased arousal, and cognitive difficulty. The hypocretinergic system originates from the lateral hypothalamic area (LHA) and projects diffusely to the whole brain, and hypocretin may be involved in the features of stress-related disorder, PTSD. Methods Our study aimed to investigate the role of basolateral amygdala (BLA) hypocretin signals in the pathophysiology of PTSD-like symptoms induced by the modified multiple-prolonged stress (MPS) protocol. The BLA, a brain region involved in fear-related behaviors, receives the hypocretin projections. In this study, TCS1102, a dual hypocretin receptor antagonist, was used to block the hypocretin signal in BLA. Results Our data indicated that the MPS protocol is a potential PTSD-like paradigm in mice. Meanwhile, the blockade of hypocretin signaling in the BLA relieved the MPS-induced fear response, and partially reduced PTSD-like anxiety behaviors performed by the open field test (OFT) and elevated plus maze (EPM) task. Discussion Our findings suggest that the hypocretinergic system is a potential therapeutic approach for PTSD treatment. With further research, the hypocretin-based medication can be a candidate for human PTSD treatment.
Collapse
Affiliation(s)
- Tung-Yen Lee
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Lu Yi
- Department of Sport Management, College of Tourism, Leisure and Sports, Aletheia University, Taipei, Taiwan
| | - Fang-Chia Chang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung City, Taiwan
- Department of Medicine, College of Medicine, China Medical University, Taichung City, Taiwan
| |
Collapse
|
14
|
Fagan HA, Baldwin DS. Pharmacological Treatment of Generalised Anxiety Disorder: Current Practice and Future Directions. Expert Rev Neurother 2023:1-14. [PMID: 37183813 DOI: 10.1080/14737175.2023.2211767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
INTRODUCTION Generalized Anxiety Disorder (GAD) is a common psychiatric condition, characterized by the presence of general apprehensiveness and excessive worry. Current management consists of a range of pharmacological and psychological treatments. However, many patients do not respond to first-line pharmacological treatments and novel anxiolytic drugs are being developed. AREAS COVERED In this review, the authors first discuss the diagnostic criteria and epidemiology of GAD. The effective pharmacological treatments for GAD and their tolerability are addressed. Current consensus guidelines for treatment of GAD are discussed, and maintenance treatment, the management of treatment resistance, and specific management of older adults and children/adolescents are considered. Finally, novel anxiolytics under development are discussed, with a focus on those which have entered clinical trials. EXPERT OPINION A range of effective treatments for GAD are available, particularly duloxetine, escitalopram, pregabalin, quetiapine, and venlafaxine. There is a limited evidence base to support the further pharmacological management of patients with GAD who have not responded to initial treatment. Although many novel anxiolytics have progressed to clinical trials, translation from animal models has been mostly unsuccessful. However, the potential of several compounds including certain psychedelics, ketamine, oxytocin, and agents modulating the orexin, endocannabinoid, and immune systems merits further study.
Collapse
Affiliation(s)
- Harry A Fagan
- Clinical and Experimental Sciences (CNS and Psychiatry), Faculty of Medicine, University of Southampton, Southampton, UK
- College Keep, Southern Health NHS Foundation Trust, Southampton, UK
| | - David S Baldwin
- Clinical and Experimental Sciences (CNS and Psychiatry), Faculty of Medicine, University of Southampton, Southampton, UK
- College Keep, Southern Health NHS Foundation Trust, Southampton, UK
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
15
|
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
|
16
|
On making (and turning adaptive to) maladaptive aversive memories in laboratory rodents. Neurosci Biobehav Rev 2023; 147:105101. [PMID: 36804263 DOI: 10.1016/j.neubiorev.2023.105101] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Fear conditioning and avoidance tasks usually elicit adaptive aversive memories. Traumatic memories are more intense, generalized, inflexible, and resistant to attenuation via extinction- and reconsolidation-based strategies. Inducing and assessing these dysfunctional, maladaptive features in the laboratory are crucial to interrogating posttraumatic stress disorder's neurobiology and exploring innovative treatments. Here we analyze over 350 studies addressing this question in adult rats and mice. There is a growing interest in modeling several qualitative and quantitative memory changes by exposing already stressed animals to freezing- and avoidance-related tests or using a relatively high aversive training magnitude. Other options combine aversive/fearful tasks with post-acquisition or post-retrieval administration of one or more drugs provoking neurochemical or epigenetic alterations reported in the trauma aftermath. It is potentially instructive to integrate these procedures and incorporate the measurement of autonomic and endocrine parameters. Factors to consider when defining the organismic and procedural variables, partially neglected aspects (sex-dependent differences and recent vs. remote data comparison) and suggestions for future research (identifying reliable individual risk and treatment-response predictors) are discussed.
Collapse
|
17
|
Ma C, Zhou N, Ma K, Niu J, Mi T, He Z, Wen Y, Liu C, He Z, Niu J. Neural pathways from hypothalamic orexin neurons to the ventrolateral preoptic area mediate sleep impairments induced by conditioned fear. Front Neurosci 2023; 17:1122803. [PMID: 36998723 PMCID: PMC10043189 DOI: 10.3389/fnins.2023.1122803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
IntroductionFear and sleep impairments common co-exist, but the underlying mechanisms remain unclear. Hypothalamic orexinergic neurons are involved in the regulation of sleep-wake and fear expression. The ventrolateral preoptic area (VLPO) is an essential brain region to promote sleep, and orexinergic axonal fibers projecting to the VLPO are involved in the maintenance of sleep-wake. Neural pathways from hypothalamic orexin neurons to the VLPO might mediate sleep impairments induced by conditioned fear.MethodsTo verify above hypothesis, electroencephalogram (EEG) and electromyogram (EMG) were recorded for analysis of sleep-wake states before and 24 h after conditioned fear training. The retrograde tracing technique and immunofluorescence staining was used to identify the projections from the hypothalamic orexin neurons to the VLPO and to observe their activation in mice with conditioned fear. Moreover, optogenetic activation or inhibition of hypothalamic orexin-VLPO pathways was performed to observe whether the sleep-wake can be regulated in mice with conditioned fear. Finally, orexin-A and orexin receptor antagonist was administered into the VLPO to certify the function of hypothalamic orexin-VLPO pathways on mediating sleep impairments induced by conditioned fear.ResultsIt was found that there was a significant decrease in the non-rapid eye movement (NREM) and rapid eye movement (REM) sleep time and a significant increase in the wakefulness time in mice with conditioned fear. The results of retrograde tracing technique and immunofluorescence staining showed that hypothalamic orexin neurons projected to the VLPO and observed the CTB labeled orexin neurons were significantly activated (c-Fos+) in the hypothalamus in mice with conditioned fear. Optogenetic activation of hypothalamic orexin to the VLPO neural pathways significantly decreased NREM and REM sleep time and increased wakefulness time in mice with conditioned fear. A significant decrease in NREM and REM sleep time and an increase in wakefulness time were observed after the injection of orexin-A into the VLPO, and the effects of orexin-A in the VLPO were blocked by a pre-administrated dual orexin antagonist (DORA).ConclusionThese findings suggest that the neural pathways from hypothalamic orexinergic neurons to the VLPO mediate sleep impairments induced by conditioned fear.
Collapse
Affiliation(s)
- Caifen Ma
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Ning Zhou
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Kang Ma
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Jiandong Niu
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Ting Mi
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Zhenquan He
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Yujun Wen
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Chunhong Liu
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
- Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zhongyi He
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- *Correspondence: Zhongyi He,
| | - Jianguo Niu
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
- Jianguo Niu,
| |
Collapse
|
18
|
Sex-dependent role of orexin deficiency in feeding behavior and affective state of mice following intermittent access to a Western diet - Implications for binge-like eating behavior. Physiol Behav 2023; 260:114069. [PMID: 36572152 DOI: 10.1016/j.physbeh.2022.114069] [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: 10/20/2022] [Revised: 12/02/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
Binge eating disorder is a debilitating disease characterized by recurrent episodes of excessive food consumption and associated with psychiatric comorbidities. Despite a growing body of research investigating the neurobiological underpinnings of eating disorders, specific treatments are lacking. Given its fundamental role in feeding behaviors, we investigated the role of the orexin (hypocretin) neuropeptide system in binge-like eating and associated phenotypes. Specifically, we submitted female and male orexin-deficient mice to a paradigm of intermittent access (once weekly for 24 h) to a Western diet (WD) to induce binge-like eating. Additionally, we measured their anxiety-like behavior and plasma corticosterone levels. All mice showed binge-like eating in response to the intermittent WD access, but females did so to a greater extent than males. While orexin deficiency did not affect binge-like eating in this paradigm, we found that female orexin-deficient mice generally weighed more, and they expressed increased hypophagia and stress levels compared to wild-type mice following binge-like eating episodes. These detrimental effects of orexin deficiency were marginal or absent in males. Moreover, male wild-type mice expressed post-binge anxiety, but orexin-deficient mice did not. In conclusion, these results extend our knowledge of orexin's role in dysregulated eating and associated negative affective states, and contribute to the growing body of evidence indicating a sexual dimorphism of the orexin system. Considering that many human disorders, and especially eating disorders, have a strong sex bias, our findings further emphasize the importance of testing both female and male subjects.
Collapse
|
19
|
Chronic orexin-1 receptor blockage attenuates depressive behaviors and provokes PSD-95 expression in a rat model of depression. Behav Brain Res 2023; 437:114123. [PMID: 36154849 DOI: 10.1016/j.bbr.2022.114123] [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: 01/30/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/21/2022]
Abstract
Depression is a devastating mood disorder affecting more than 300 million people worldwide. Almost 30 % of patients still suffer from treatment resistant depression. Although many reports support the involvement of orexin in the pathophysiology of depression, the precise role of orexin is still unclear. In this study, we evaluated the role of the orexin 1 receptor (Orx1R) on depressive behaviors and the alterations in postsynaptic density-95 (PSD-95) protein in the chronic mild stress (CMS) model of depression. Fifty-four male Wistar rats were randomly allocated to 6 groups; Control, CMS, acute SB-334867 (SB), CMS+SB, chronic SB (CSB) and CMS+CSB. Rats were exposed to one or two unpredictable stressors each day for three weeks for the induction of CMS. Intracerebroventricular (icv) injection of SB-334867, a selective Orx1R antagonist, was performed either 30 min before behavioral tests (acute) or once daily for 14 days (chronic). Behavioral despair was assessed by immobility time in the forced swim test (FST), sucrose consumption in sucrose preference test (SPT), and the number of crosses in the open field test (OFT) on days 1, 11, and 22 of the experiment. Finally, rats were decapitated, and brain tissue of the hippocampus (HPC) and prefrontal cortex (PFC) were collected, and the relative expression of PSD-95 was evaluated by western blotting. The CMS model rats showed a significant increase in FST immobility time (P = 0.001) and a decrease in locomotion (P = 0.04) and sucrose preference (P = 0.039). Chronic application of SB decreased immobility time to the control values (P = 0.001) and diminished locomotion (P = 0.047) and sucrose preference (P = 0.042) in comparison to the CMS group. Acute SB reversed just the immobility time (P ≤ 0.006). Chronic SB treatment increased the relative PSD-95 expression in PFC (P = 0.001). Hence, chronic antagonism of Orx1R alleviates depressive behaviors induced by CMS and improves PSD-95 expression in PFC.
Collapse
|
20
|
Orexin Receptor Antagonists in the Treatment of Depression: A Leading Article Summarising Pre-clinical and Clinical Studies. CNS Drugs 2023; 37:1-12. [PMID: 36436175 DOI: 10.1007/s40263-022-00974-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2022] [Indexed: 11/28/2022]
Abstract
The orexin (hypocretin) system comprises two neuropeptides (orexin-A and orexin-B) and two G-protein coupled receptors (the orexin type 1 and the orexin type 2 receptor). The system regulates several biological functions including appetite, the sleep-wake cycle, the stress response, and motivation and reward processing. Dysfunction of the orexin system has been implicated in the pathophysiology of depression in human and animal studies, although the exact nature of this dysfunction remains unclear. Orexin receptor antagonists (ORAs) are a class of compounds developed for the treatment of insomnia and have demonstrated efficacy in this area. Three dual orexin receptor antagonists (DORAs) have received licences for treatment of primary insomnia and some ORAs have since been investigated as potential treatments for major depressive disorder (MDD). In this leading article, we summarise the existing literature on use of ORAs in depression, in pre-clinical and clinical studies. In rodent models of depression, investigated ORAs have included the DORA almorexant and TCS1102, the selective orexin 1 receptor antagonists SB334867 and SB674042 and the selective orexin 2 receptor antagonists LSN2424100, MK-1064 and TCS-OX2-29. These pre-clinical studies suggest a possible antidepressant effect of systemic DORA treatment, however the evidence from selective ORAs is conflicting. To date, four published RCTs (one with the DORA filorexant and three with the selective orexin 2 receptor antagonist seltorexant), have compared an ORA with placebo in the treatment of MDD. Only one of these demonstrated a statistically significant difference relative to placebo.
Collapse
|
21
|
Smits JAJ, Monfils MH, Otto MW, Telch MJ, Shumake J, Feinstein JS, Khalsa SS, Cobb AR, Parsons EM, Long LJ, McSpadden B, Johnson D, Greenberg A, Exposure Therapy Consortium. CO 2 reactivity as a biomarker of exposure-based therapy non-response: study protocol. BMC Psychiatry 2022; 22:831. [PMID: 36575425 PMCID: PMC9793569 DOI: 10.1186/s12888-022-04478-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Exposure-based therapy is an effective first-line treatment for anxiety-, obsessive-compulsive, and trauma- and stressor-related disorders; however, many patients do not improve, resulting in prolonged suffering and poorly used resources. Basic research on fear extinction may inform the development of a biomarker for the selection of exposure-based therapy. Growing evidence links orexin system activity to deficits in fear extinction and we have demonstrated that reactivity to an inhaled carbon dioxide (CO2) challenge-a safe, affordable, and easy-to-implement procedure-can serve as a proxy for orexin system activity and predicts fear extinction deficits in rodents. Building upon this basic research, the goal for the proposed study is to validate CO2 reactivity as a biomarker of exposure-based therapy non-response. METHODS We will assess CO2 reactivity in 600 adults meeting criteria for one or more fear- or anxiety-related disorders prior to providing open exposure-based therapy. By incorporating CO2 reactivity into a multivariate model predicting treatment non-response that also includes reactivity to hyperventilation as well as a number of related predictor variables, we will establish the mechanistic specificity and the additive predictive utility of the potential CO2 reactivity biomarker. By developing models independently within two study sites (University of Texas at Austin and Boston University) and predicting the other site's data, we will validate that the results are likely to generalize to future clinical samples. DISCUSSION Representing a necessary stage in translating basic research, this investigation addresses an important public health issue by testing an accessible clinical assessment strategy that may lead to a more effective treatment selection (personalized medicine) for patients with anxiety- and fear-related disorders, and enhanced understanding of the mechanisms governing exposure-based therapy. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05467683 (20/07/2022).
Collapse
Affiliation(s)
- Jasper A. J. Smits
- grid.89336.370000 0004 1936 9924Department of Psychology and Institute for Mental Health Research, University of Texas at Austin, 1 University Station, Austin, TX 78712 USA
| | - Marie-H. Monfils
- grid.89336.370000 0004 1936 9924Department of Psychology and Institute for Mental Health Research, University of Texas at Austin, 1 University Station, Austin, TX 78712 USA
| | - Michael W. Otto
- grid.189504.10000 0004 1936 7558Department of Psychological and Brain Sciences, Boston University, 900 Commonwealth Avenue, Floor 2, Boston, MA 02215 USA
| | - Michael J. Telch
- grid.89336.370000 0004 1936 9924Department of Psychology and Institute for Mental Health Research, University of Texas at Austin, 1 University Station, Austin, TX 78712 USA
| | - Jason Shumake
- grid.89336.370000 0004 1936 9924Department of Psychology and Institute for Mental Health Research, University of Texas at Austin, 1 University Station, Austin, TX 78712 USA
| | - Justin S. Feinstein
- grid.417423.70000 0004 0512 88633The Laureate Institute for Brain Research, 6655 South Yale Ave., Tulsa, Oklahoma 74136 USA
| | - Sahib S. Khalsa
- grid.417423.70000 0004 0512 88633The Laureate Institute for Brain Research, 6655 South Yale Ave., Tulsa, Oklahoma 74136 USA
| | - Adam R. Cobb
- grid.89336.370000 0004 1936 9924Department of Psychology and Institute for Mental Health Research, University of Texas at Austin, 1 University Station, Austin, TX 78712 USA ,grid.259828.c0000 0001 2189 3475Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina and Ralph H. Johnson VAHCS, 67 President Street MSC 862, Charleston, SC 29425 USA
| | - E. Marie Parsons
- grid.189504.10000 0004 1936 7558Department of Psychological and Brain Sciences, Boston University, 900 Commonwealth Avenue, Floor 2, Boston, MA 02215 USA
| | - Laura J. Long
- grid.189504.10000 0004 1936 7558Department of Psychological and Brain Sciences, Boston University, 900 Commonwealth Avenue, Floor 2, Boston, MA 02215 USA
| | - Bryan McSpadden
- grid.89336.370000 0004 1936 9924Department of Psychology and Institute for Mental Health Research, University of Texas at Austin, 1 University Station, Austin, TX 78712 USA
| | - David Johnson
- grid.89336.370000 0004 1936 9924Department of Psychology and Institute for Mental Health Research, University of Texas at Austin, 1 University Station, Austin, TX 78712 USA
| | - Alma Greenberg
- grid.189504.10000 0004 1936 7558Department of Psychological and Brain Sciences, Boston University, 900 Commonwealth Avenue, Floor 2, Boston, MA 02215 USA
| | | |
Collapse
|
22
|
Swift KM, Thomas CL, Balkin TJ, Lowery-Gionta EG, Matson LM. Acute sleep interventions as an avenue for treatment of trauma-associated disorders. J Clin Sleep Med 2022; 18:2291-2312. [PMID: 35678060 PMCID: PMC9435330 DOI: 10.5664/jcsm.10074] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022]
Abstract
Scientific evidence that acute, posttrauma sleep disturbances (eg, nightmares and insomnia) can contribute significantly to the pathogenesis of trauma-induced disorders is compelling. Sleep disturbances precipitating from trauma are uniquely predictive of daytime posttrauma symptom occurrence and severity, as well as subsequent onset of mental health disorders, including post-traumatic stress disorder. Conversely, adequate sleep during the acute posttrauma period is associated with reduced likelihood of adverse mental health outcomes. These findings, which are broadly consistent with what is known about the role of sleep in the regulation of emotion, suggest that the acute posttrauma period constitutes a "window of opportunity" during which treatment of sleep disturbances may be especially effective for preventing or mitigating progression of aberrant psychophysiological processes. At this point, the weight of the scientific evidence supporting this possibility warrants initiation of clinical trials to confirm the benefits of targeted prophylactic sleep enhancement, and to establish treatment guidelines as appropriate. CITATION Swift KM, Thomas CL, Balkin TJ, Lowery-Gionta EG, Matson LM. Acute sleep interventions as an avenue for treatment of trauma-associated disorders. J Clin Sleep Med. 2022;18(9):2291-2312.
Collapse
Affiliation(s)
- Kevin M. Swift
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Connie L. Thomas
- Department of Sleep Medicine, Walter Reed National Military Medical Center, Bethesda, Maryland
- Department of Psychiatry, Uniformed Services University of Health Sciences, Bethesda, Maryland
| | - Thomas J. Balkin
- Behavioral Biology Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Emily G. Lowery-Gionta
- Behavioral Biology Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Liana M. Matson
- Behavioral Biology Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland
| |
Collapse
|
23
|
Demidova A, Kahl E, Fendt M. Orexin deficiency affects sensorimotor gating and its amphetamine-induced impairment. Prog Neuropsychopharmacol Biol Psychiatry 2022; 116:110517. [PMID: 35101602 DOI: 10.1016/j.pnpbp.2022.110517] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/17/2022]
Abstract
The orexin neuropeptides have an important role in the regulation of the sleep/wake cycle and foraging, as well as in reward processing and emotions. Furthermore, recent research implicates the orexin system in different behavioral endophenotypes of neuropsychiatric diseases such as social avoidance and cognitive flexibility. Utilizing orexin-deficient mice, the present study tested the hypothesis that orexin is involved in two further mouse behavioral endophenotypes of neuropsychiatric disorders, i.e., sensorimotor gating and amphetamine sensitivity. The data revealed that orexin-deficient mice expressed a deficit in sensorimotor gating, measured by prepulse inhibition of the startle response. Amphetamine treatment impaired prepulse inhibition in wildtype and heterozygous orexin-deficient mice, but had no effects in homozygous orexin-deficient mice. Furthermore, locomotor activity and center time in the open field was not affected by orexin deficiency but was similarly increased or decreased, respectively, by amphetamine treatment in all genotypes. These data indicate that the orexin system modulates prepulse inhibition and is involved in mediating amphetamine's effect on prepulse inhibition. Future studies should investigate whether pharmacological manipulations of the orexin system can be used to treat neuropsychiatric diseases associated with deficits in sensorimotor gating, such as schizophrenia or attention deficit hyperactivity disorder.
Collapse
Affiliation(s)
- Alexandrina Demidova
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany; Psychology Master Program, 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; Psychology Master Program, Otto-von-Guericke University Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke University Magdeburg, Germany.
| |
Collapse
|
24
|
Abstract
The hypocretins (Hcrts), also known as orexins, are two neuropeptides produced exclusively in the lateral hypothalamus. They act on two specific receptors that are widely distributed across the brain and involved in a myriad of neurophysiological functions that include sleep, arousal, feeding, reward, fear, anxiety and cognition. Hcrt cell loss in humans leads to narcolepsy with cataplexy (narcolepsy type 1), a disorder characterized by intrusions of sleep into wakefulness, demonstrating that the Hcrt system is nonredundant and essential for sleep/wake stability. The causal link between Hcrts and arousal/wakefulness stabilisation has led to the development of a new class of drugs, Hcrt receptor antagonists to treat insomnia, based on the assumption that blocking orexin-induced arousal will facilitate sleep. This has been clinically validated: currently, two Hcrt receptor antagonists are approved to treat insomnia (suvorexant and lemborexant), with a New Drug Application recently submitted to the US Food and Drug Administration for a third drug (daridorexant). Other therapeutic applications under investigation include reduction of cravings in substance-use disorders and prevention of neurodegenerative disorders such as Alzheimer's disease, given the apparent bidirectional relationship between poor sleep and worsening of the disease. Circuit neuroscience findings suggest that the Hcrt system is a hub that integrates diverse inputs modulating arousal (e.g., circadian rhythms, metabolic status, positive and negative emotions) and conveys this information to multiple output regions. This neuronal architecture explains the wealth of physiological functions associated with Hcrts and highlights the potential of the Hcrt system as a therapeutic target for a number of disorders. We discuss present and future possible applications of drugs targeting the Hcrt system for the treatment of circuit-related neuropsychiatric and neurodegenerative conditions.
Collapse
Affiliation(s)
- Laura H Jacobson
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.,Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia.,Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Daniel Hoyer
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.,Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
25
|
Ten-Blanco M, Flores Á, Pereda-Pérez I, Piscitelli F, Izquierdo-Luengo C, Cristino L, Romero J, Hillard CJ, Maldonado R, Di Marzo V, Berrendero F. Amygdalar CB2 cannabinoid receptor mediates fear extinction deficits promoted by orexin-A/hypocretin-1. Biomed Pharmacother 2022; 149:112925. [PMID: 35477218 DOI: 10.1016/j.biopha.2022.112925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/25/2022] [Accepted: 04/04/2022] [Indexed: 11/02/2022] Open
Abstract
Anxiety and stress disorders are often characterized by an inability to extinguish learned fear responses. Orexins/hypocretins are involved in the modulation of aversive memories, and dysregulation of this system may contribute to the aetiology of anxiety disorders characterized by pathological fear. The mechanisms by which orexins regulate fear are unknown. Here we investigated the role of the endogenous cannabinoid system in the impaired fear extinction induced by orexin-A (OXA) in male mice. The selective inhibitor of 2-arachidonoylglycerol (2-AG) biosynthesis O7460 abolished the fear extinction deficits induced by OXA. Accordingly, increased 2-AG levels were observed in the amygdala and hippocampus of mice treated with OXA that do not extinguish fear, suggesting that high levels of this endocannabinoid are related to poor extinction. Impairment of fear extinction induced by OXA was associated with increased expression of CB2 cannabinoid receptor (CB2R) in microglial cells of the basolateral amygdala. Consistently, the intra-amygdala infusion of the CB2R antagonist AM630 completely blocked the impaired extinction promoted by OXA. Microglial and CB2R expression depletion in the amygdala with PLX5622 chow also prevented these extinction deficits. These results show that overactivation of the orexin system leads to impaired fear extinction through 2-AG and amygdalar CB2R. This novel mechanism could be of relevance for the development of novel potential approaches to treat diseases associated with inappropriate retention of fear, such as post-traumatic stress disorder, panic anxiety and phobias.
Collapse
Affiliation(s)
- Marc Ten-Blanco
- Instituto de Investigaciones Biosanitarias, Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - África Flores
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, PRBB, 08003 Barcelona, Spain
| | - Inmaculada Pereda-Pérez
- Instituto de Investigaciones Biosanitarias, Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Cristina Izquierdo-Luengo
- Instituto de Investigaciones Biosanitarias, Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Luigia Cristino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Julián Romero
- Instituto de Investigaciones Biosanitarias, Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Cecilia J Hillard
- Department of Pharmacology and Toxicology and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, PRBB, 08003 Barcelona, Spain
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy; Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Faculty of Medicine and Faculty of Agriculture and Food Sciences, Hearth and Lung Research Institute (IUCPQ), Institute of Nutrition and Functional Foods (INAF) and NUTRISS Center, Université Laval, Quebec City, Canada
| | - Fernando Berrendero
- Instituto de Investigaciones Biosanitarias, Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain.
| |
Collapse
|
26
|
Yaeger JDW, Krupp KT, Jacobs BM, Onserio BO, Meyerink BL, Cain JT, Ronan PJ, Renner KJ, DiLeone RJ, Summers CH. Orexin 1 Receptor Antagonism in the Basolateral Amygdala Shifts the Balance From Pro- to Antistress Signaling and Behavior. Biol Psychiatry 2022; 91:841-852. [PMID: 35279280 PMCID: PMC9020795 DOI: 10.1016/j.biopsych.2021.12.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/18/2021] [Accepted: 12/29/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Stress produces differential behavioral responses through select molecular modifications to specific neurocircuitry elements. The orexin (Orx) system targets key components of this neurocircuitry in the basolateral amygdala (BLA). METHODS We assessed the contribution of intra-BLA Orx1 receptors (Orx1Rs) in the expression of stress-induced phenotypes of mice. Using the Stress Alternatives Model, a social stress paradigm that produces two behavioral phenotypes, we characterized the role of intra-BLA Orx1R using acute pharmacological inhibition (SB-674042) and genetic knockdown (AAV-U6-Orx1R-shRNA) strategies. RESULTS In the BLA, we observed that Orx1R (Hcrtr1) messenger RNA is predominantly expressed in CamKIIα+ glutamatergic neurons and rarely in GABAergic (gamma-aminobutyric acidergic) cells. While there is a slight overlap in Hcrtr1 and Orx2 receptor (Hcrtr2) messenger RNA expression in the BLA, we find that these receptors are most often expressed in separate cells. Antagonism of intra-BLA Orx1R after phenotype formation shifted behavioral expression from stress-sensitive (Stay) to stress-resilient (Escape) responses, an effect that was mimicked by genetic knockdown. Acute inhibition of Orx1R in the BLA also reduced contextual and cued fear freezing responses in Stay animals. This phenotype-specific behavioral change was accompanied by biased molecular transcription favoring Hcrtr2 over Hcrtr1 and Mapk3 over Plcb1 cell signaling cascades and enhanced Bdnf messenger RNA. CONCLUSIONS Functional reorganization of intra-BLA gene expression is produced by antagonism of Orx1R, which promotes elevated Hcrtr2, greater Mapk3, and increased Bdnf expression. Together, these results provide evidence for a receptor-driven mechanism that balances pro- and antistress responses within the BLA.
Collapse
Affiliation(s)
- Jazmine D W Yaeger
- Department of Biology, University of South Dakota, Vermillion, South Dakota; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, South Dakota
| | - Kevin T Krupp
- Department of Biology, University of South Dakota, Vermillion, South Dakota; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Benjamin M Jacobs
- Department of Biology, University of South Dakota, Vermillion, South Dakota; Texas Christian University School of Medicine, Fort Worth, Texas
| | - Benard O Onserio
- Department of Biology, University of South Dakota, Vermillion, South Dakota
| | - Brandon L Meyerink
- Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota; Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota
| | - Jacob T Cain
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota
| | - Patrick J Ronan
- Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, South Dakota
| | - Kenneth J Renner
- Department of Biology, University of South Dakota, Vermillion, South Dakota; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| | - Ralph J DiLeone
- Division of Molecular Psychiatry, Ribicoff Research Facilities, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Cliff H Summers
- Department of Biology, University of South Dakota, Vermillion, South Dakota; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, South Dakota.
| |
Collapse
|
27
|
Sanacora G, Yan Z, Popoli M. The stressed synapse 2.0: pathophysiological mechanisms in stress-related neuropsychiatric disorders. Nat Rev Neurosci 2022; 23:86-103. [PMID: 34893785 DOI: 10.1038/s41583-021-00540-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2021] [Indexed: 12/25/2022]
Abstract
Stress is a primary risk factor for several neuropsychiatric disorders. Evidence from preclinical models and clinical studies of depression have revealed an array of structural and functional maladaptive changes, whereby adverse environmental factors shape the brain. These changes, observed from the molecular and transcriptional levels through to large-scale brain networks, to the behaviours reveal a complex matrix of interrelated pathophysiological processes that differ between sexes, providing insight into the potential underpinnings of the sex bias of neuropsychiatric disorders. Although many preclinical studies use chronic stress protocols, long-term changes are also induced by acute exposure to traumatic stress, opening a path to identify determinants of resilient versus susceptible responses to both acute and chronic stress. Epigenetic regulation of gene expression has emerged as a key player underlying the persistent impact of stress on the brain. Indeed, histone modification, DNA methylation and microRNAs are closely involved in many aspects of the stress response and reveal the glutamate system as a key player. The success of ketamine has stimulated a whole line of research and development on drugs directly or indirectly targeting glutamate function. However, the challenge of translating the emerging understanding of stress pathophysiology into effective clinical treatments remains a major challenge.
Collapse
Affiliation(s)
- Gerard Sanacora
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Department of Pharmaceutical Sciences, University of Milano, Milan, Italy.
| |
Collapse
|
28
|
Durairaja A, Steinecke CS, Fendt M. Intracerebroventricular infusion of the selective orexin 1 receptor antagonist SB-334867 impairs cognitive flexibility in a sex-dependent manner. Behav Brain Res 2022; 424:113791. [DOI: 10.1016/j.bbr.2022.113791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 11/17/2022]
|
29
|
Wu G, Zhang X, Li S, Zhou D, Bai J, Wang H, Shu Q. Overexpression of ORX or MCH Protects Neurological Function Against Ischemic Stroke. Neurotox Res 2022; 40:44-55. [PMID: 35013906 DOI: 10.1007/s12640-021-00457-4] [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: 05/27/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 11/29/2022]
Abstract
In recent years, orexin (ORX) and melanin-concentrating hormone (MCH) have been demonstrated to exert neuroprotective roles in cerebral ischemia. Hence, this study investigated the regulatory function of ORX and MCH in neurological function following ischemic stroke and explored the molecular mechanism underlying these functions. A rat model of ischemic stroke was developed by middle cerebral artery occlusion (MCAO), and Longa scoring was employed to evaluate the degree of neurological function deficit. The expression patterns of ORX and MCH were examined by real-time polymerase chain reaction in the brain tissues of rats with ischemic stroke induced by middle cerebral artery occlusion (MCAO). Moreover, electroencephalography (EEG) analysis and high-performance liquid chromatography (HPLC) were respectively performed to detect rapid-eye movement (REM) sleep, the glutamate (Glu) uptake, and the expression of γ-aminobutyric acid B receptor (GABAB). Immunoblotting was performed to test the levels of autophagic markers LC3, BECLIN-1, and p62. Immunohistochemistry (IHC) staining and TUNEL assays were respectively used to assess the autophagy and neuronal apoptosis. Results demonstrated that ORX and MCH were lowly expressed in brain of rats with ischemic stroke. ORX or MCH overexpression decreased neuronal apoptosis and autophagy, and improved the sleep architecture of post-stroke rats, while rescuing Glu uptake and GABA expression. ORX or MCH upregulation exerted protective effects on neurological function. Taken together, ORX and/or MCH protect against ischemic stroke in a rat model, highlighting their value as targets for the clinical treatment of ischemic stroke.
Collapse
Affiliation(s)
- Gang Wu
- East Section of South Second Ring Road, The Second Affiliated Hospital of Xi'an Jiaotong University, No.151, Xi'an 710054, Shaanxi, China
| | - Xi'an Zhang
- Ninth Hospital of Xi'an Affiliated To Xi'an Jiaotong University, Xi'an 710054, China
| | - Shijun Li
- Department of Pharmacy, Wuhan Union Hospital, Wuhan, 430022, China
| | - Dan Zhou
- Ninth Hospital of Xi'an Affiliated To Xi'an Jiaotong University, Xi'an 710054, China
| | - Jie Bai
- East Section of South Second Ring Road, The Second Affiliated Hospital of Xi'an Jiaotong University, No.151, Xi'an 710054, Shaanxi, China
| | - Hanxiang Wang
- Department of Pharmacy, Wuhan Union Hospital, Wuhan, 430022, China
| | - Qing Shu
- Ninth Hospital of Xi'an Affiliated To Xi'an Jiaotong University, Xi'an 710054, China.
| |
Collapse
|
30
|
Insulin-like growth factor I mitigates post-traumatic stress by inhibiting AMP-kinase in orexin neurons. Mol Psychiatry 2022; 27:2182-2196. [PMID: 35115701 PMCID: PMC9126821 DOI: 10.1038/s41380-022-01442-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 12/14/2021] [Accepted: 01/11/2022] [Indexed: 12/21/2022]
Abstract
Maladaptive coping behaviors are probably involved in post-traumatic stress disorders (PTSD), but underlying mechanisms are incompletely understood. We now report that mice lacking functional insulin-like growth factor I (IGF-I) receptors in orexin neurons of the lateral hypothalamus (Firoc mice) are unresponsive to the anxiolytic actions of IGF-I and develop PTSD-like behavior that is ameliorated by inhibition of orexin neurons. Conversely, systemic IGF-I treatment ameliorated PTSD-like behavior in a wild-type mouse model of PTSD (PTSD mice). Further, systemic IGF-I modified the GABA/Glutamate synaptic structure in orexin neurons of naïve wild-type mice by increasing the dephosphorylation of GABA(B) receptor subunit through inhibition of AMP-kinase (AMPK). Significantly, pharmacological inhibition of AMPK mimicked IGF-I, normalizing fear behavior in PTSD mice. Thus, we suggest that IGF-I enables coping behaviors by balancing E/I input onto orexin neurons in a context-dependent manner. These observations provide a novel therapeutic approach to PTSD through modulation of AMPK.
Collapse
|
31
|
Lo Y, Yi PL, Hsiao YT, Chang FC. Hypocretin in locus coeruleus and dorsal raphe nucleus mediates inescapable footshock stimulation (IFS)-induced REM sleep alteration. Sleep 2021; 45:6490200. [PMID: 34969120 DOI: 10.1093/sleep/zsab301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/12/2021] [Indexed: 11/14/2022] Open
Abstract
Hypocretin (hcrt) is a stress-reacting neuropeptide mediating arousal and energy homeostasis. An inescapable footshock stimulation (IFS) could initiate the hcrt release from the lateral hypothalamus (LHA) and suppresses rapid eye movement (REM) sleep in rodents. However, the effects of the IFS-induced hcrts on REM-off nuclei, the locus coeruleus (LC) and dorsal raphe nucleus (DRN), remained unclear. We hypothesized that the hcrt projections from the LHA to LC or DRN mediate IFS-induced sleep disruption. Our results demonstrated that the IFS increased hcrt expression and the neuronal activities in the LHA, hypothalamus, brainstem, thalamus, and amygdala. Suppressions of REM sleep and slow wave activity during non-REM (NREM) sleep caused by the high expression of hcrts were blocked when a non-specific and dual hcrt receptor antagonist was administered into the LC or DRN. Furthermore, the IFS also caused an elevated innate anxiety, but was limitedly influenced by the hcrt antagonist. This result suggests that the increased hcrt concentrations in the LC and DRN mediate stress-induced sleep disruptions and might partially involve IFS-induced anxiety.
Collapse
Affiliation(s)
- Yun Lo
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan, University, Taipei, Taiwan
| | - Pei-Lu Yi
- Department of Sport Management, College of Tourism, Leisure and Sports, Aletheia, University, New Taipei City, Taiwan
| | - Yi-Tse Hsiao
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan, University, Taipei, Taiwan
| | - Fang-Chia Chang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan, University, Taipei, Taiwan.,Graduate Institute of Brain & Mind Sciences, College of Medicine, National Taiwan, University, Taipei, Taiwan.,Graduate Institute of Acupuncture Science, College of Chinese Medicine, China, Medical University, Taichung, Taiwan.,Department of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
32
|
Han D, Shi Y, Han F. The effects of orexin-A and orexin receptors on anxiety- and depression-related behaviors in a male rat model of post-traumatic stress disorder. J Comp Neurol 2021; 530:592-606. [PMID: 34387361 DOI: 10.1002/cne.25231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 08/08/2021] [Accepted: 08/09/2021] [Indexed: 12/14/2022]
Abstract
Orexin neurons play an important role in stress-related mental disorders including post-traumatic stress disorder (PTSD). Anxiety- and depression-related symptoms commonly occur in combination with PTSD. However, the role of the orexin system in mediating alterations in these affective symptoms remains unclear. The medial prefrontal cortex (mPFC) is implicated in both cognitive and emotional processing. In the present study, we investigated anxiety- and depression-related behavioral changes using the elevated plus maze, the sucrose preference test, and the open field test in male rats with single prolonged stress (SPS) induced-PTSD. The expression of orexin-A in the hypothalamus and orexin receptors (OX1R and OX2R) in the mPFC was detected and quantified by immunohistochemistry, western blotting, and real-time polymerase chain reaction. We found that the SPS rats exhibited enhanced levels of anxiety, reduced exploratory activities, and anhedonia. Furthermore, SPS resulted in reductions in the expression of orexin-A in the hypothalamus and the increased the expression of OX1R in the mPFC. The intracerebroventricular administration of orexin-A alleviated behavioral changes in SPS rats and partly restored the increased levels of OX1R in the mPFC. These results suggest that the orexin system plays a role in the anxiety- and depression-related symptoms observed in PTSD.
Collapse
Affiliation(s)
- Dan Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, Shenyang, China.,Department of Neonatology, The First Hospital of China Medical University, Shenyang, China
| | - Yuxiu Shi
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, Shenyang, China
| | - Fang Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, Shenyang, China
| |
Collapse
|
33
|
Souffi S, Nodal FR, Bajo VM, Edeline JM. When and How Does the Auditory Cortex Influence Subcortical Auditory Structures? New Insights About the Roles of Descending Cortical Projections. Front Neurosci 2021; 15:690223. [PMID: 34413722 PMCID: PMC8369261 DOI: 10.3389/fnins.2021.690223] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/14/2021] [Indexed: 12/28/2022] Open
Abstract
For decades, the corticofugal descending projections have been anatomically well described but their functional role remains a puzzling question. In this review, we will first describe the contributions of neuronal networks in representing communication sounds in various types of degraded acoustic conditions from the cochlear nucleus to the primary and secondary auditory cortex. In such situations, the discrimination abilities of collicular and thalamic neurons are clearly better than those of cortical neurons although the latter remain very little affected by degraded acoustic conditions. Second, we will report the functional effects resulting from activating or inactivating corticofugal projections on functional properties of subcortical neurons. In general, modest effects have been observed in anesthetized and in awake, passively listening, animals. In contrast, in behavioral tasks including challenging conditions, behavioral performance was severely reduced by removing or transiently silencing the corticofugal descending projections. This suggests that the discriminative abilities of subcortical neurons may be sufficient in many acoustic situations. It is only in particularly challenging situations, either due to the task difficulties and/or to the degraded acoustic conditions that the corticofugal descending connections bring additional abilities. Here, we propose that it is both the top-down influences from the prefrontal cortex, and those from the neuromodulatory systems, which allow the cortical descending projections to impact behavioral performance in reshaping the functional circuitry of subcortical structures. We aim at proposing potential scenarios to explain how, and under which circumstances, these projections impact on subcortical processing and on behavioral responses.
Collapse
Affiliation(s)
- Samira Souffi
- Department of Integrative and Computational Neurosciences, Paris-Saclay Institute of Neuroscience (NeuroPSI), UMR CNRS 9197, Paris-Saclay University, Orsay, France
| | - Fernando R. Nodal
- Department of Physiology, Anatomy and Genetics, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Victoria M. Bajo
- Department of Physiology, Anatomy and Genetics, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Jean-Marc Edeline
- Department of Integrative and Computational Neurosciences, Paris-Saclay Institute of Neuroscience (NeuroPSI), UMR CNRS 9197, Paris-Saclay University, Orsay, France
| |
Collapse
|
34
|
Yamashita A, Moriya S, Nishi R, Kaminosono J, Yamanaka A, Kuwaki T. Aversive emotion rapidly activates orexin neurons and increases heart rate in freely moving mice. Mol Brain 2021; 14:104. [PMID: 34193206 PMCID: PMC8247171 DOI: 10.1186/s13041-021-00818-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/22/2021] [Indexed: 01/28/2023] Open
Abstract
The perifornical area of the hypothalamus has been known as the center for the defense response, or fight-or-flight response, which is characterized by a concomitant rise in arterial blood pressure, heart rate, and respiratory frequency. It is well established that orexin neurons, which are located in this region, play a critical role in this response. In this study, we further examined this role by recording orexin neuronal activity and heart rate in freely moving mice using an original dual-channel fiber photometry system in vivo. Analysis of orexin neuron activity in relation to autonomic responses to aversive stimuli revealed a rapid increase in neuronal activity just prior to changes in heart rate. In addition, we examined whether orexin neurons would be activated by a conditioned neutral sound that was previously associated with aversive stimulus. We show that the memory of the aversive stimulus activated orexin neurons and increased heart rate. Our data suggest that orexin neurons are a key component linking aversive emotions to autonomic defense response. Our data also suggest that targeting orexin neurons may enable treatment of psychiatric disorders associated with chronic stress and traumatic memories.
Collapse
Affiliation(s)
- Akira Yamashita
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, 890-8544, Japan
| | - Shunpei Moriya
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, 890-8544, Japan
| | - Ryusei Nishi
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, 890-8544, Japan
| | - Jun Kaminosono
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, 890-8544, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan
| | - Tomoyuki Kuwaki
- Department of Physiology, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, 890-8544, Japan.
| |
Collapse
|
35
|
Gupta PR, Prabhavalkar K. Combination therapy with neuropeptides for the treatment of anxiety disorder. Neuropeptides 2021; 86:102127. [PMID: 33607407 DOI: 10.1016/j.npep.2021.102127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/29/2020] [Accepted: 01/26/2021] [Indexed: 11/29/2022]
Abstract
Anxiety is a neurological disorder that is characterized by excessive, persistent, and unreasonable worry about everyday things like family, work, money, and relationships. The current therapy used for the treatment has many disadvantages like higher cost, severe adverse reactions, and has suboptimal efficiency. There is a need to look for more innovative approaches for the treatment of anxiety disorder which overcomes the disadvantages of conventional treatment. Recent findings suggest a strong correlation of glutamate with anxiety. Some promising drugs which have a novel mechanism for anxiolytic action are currently under clinical development for generalized anxiety disorder, social anxiety disorder, panic disorder, obsessive-compulsive disorder, or post-traumatic stress disorder. Similarly, an interrelation of oxytocin with neuropeptide S or glutamate or vasopressin can also be considered for further evaluation for the development of new drugs for anxiety treatment. Anxiolytic drug development is a multi-target approach, with the idea of more efficiently equilibrating perturbed circuits. This review focuses on targeting unconventional targets like the glutamate system, voltage-gated ion channels, and neuropeptides system either alone or in combination for the treatment of anxiety disorder.
Collapse
Affiliation(s)
- Priti Ramakant Gupta
- Department of Pharmacology, SVKM'S Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400 056, India
| | - Kedar Prabhavalkar
- Department of Pharmacology, SVKM'S Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400 056, India.
| |
Collapse
|
36
|
Clark JW, Daykin H, Metha JA, Allocca G, Hoyer D, Drummond SPA, Jacobson LH. Manipulation of REM sleep via orexin and GABAA receptor modulators differentially affects fear extinction in mice: effect of stable versus disrupted circadian rhythm. Sleep 2021; 44:6171207. [PMID: 33720375 DOI: 10.1093/sleep/zsab068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/22/2021] [Indexed: 12/18/2022] Open
Abstract
Sleep disruption, and especially REM sleep disruption, is associated with fear inhibition impairment in animals and humans. The REM sleep-fear inhibition relationship raises concern for individuals with PTSD, whose sleep disturbance is commonly treated with hypnotics which disrupt and/or decrease REM sleep, such as benzodiazepines or 'Z-drugs'. Here, we examined the effects of the Z-drug zolpidem, a GABAA receptor positive allosteric modulator, as well as suvorexant, an orexin receptor antagonist (hypnotics which decrease and increase REM sleep, respectively) in the context of circadian disruption in murine models of fear inhibition-related processes (i.e., fear extinction and safety learning). Adult male C57Bl/6J mice completed fear and safety conditioning before undergoing shifts in the light-dark (LD) cycle or maintaining a consistent LD schedule. Fear extinction and recall of conditioned safety were thereafter tested daily. Immediately prior to onset of the light phase between testing sessions, mice were treated with zolpidem, suvorexant, or vehicle (methylcellulose). EEG/EMG analysis showed temporal distribution of REM sleep was misaligned during LD cycle-shifts, while REM sleep duration was preserved. Suvorexant increased REM sleep and improved fear extinction rate, relative to zolpidem, which decreased REM sleep. Survival analysis demonstrated LD shifted mice treated with suvorexant were faster to achieve complete extinction than vehicle and zolpidem-treated mice in the LD shifted condition. By contrast, retention of conditioned safety memory was not influenced by either treatment. This study thus provides preclinical evidence for the potential clinical utility of hypnotics which increase REM sleep for fear extinction after PTSD-relevant sleep disturbance.
Collapse
Affiliation(s)
- Jacob W Clark
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, VIC, Australia.,Department of Pharmacology and Therapeutics, The University of Melbourne, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, VIC, Australia
| | - Heather Daykin
- Department of Pharmacology and Therapeutics, The University of Melbourne, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, VIC, Australia
| | - Jeremy A Metha
- Department of Pharmacology and Therapeutics, The University of Melbourne, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, VIC, Australia.,Brain, Mind and Markets Laboratory, Department of Finance, The University of Melbourne, VIC, Australia
| | - Giancarlo Allocca
- Department of Pharmacology and Therapeutics, The University of Melbourne, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, VIC, Australia.,Somnivore Pty. Ltd., Bacchus Marsh, Victoria, Australia
| | - Daniel Hoyer
- Department of Pharmacology and Therapeutics, The University of Melbourne, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, VIC, Australia.,Department of Molecular Medicine, The Scripps Research Institute, CA, The United States of America
| | - Sean P A Drummond
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, VIC, Australia
| | - Laura H Jacobson
- Department of Pharmacology and Therapeutics, The University of Melbourne, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, VIC, Australia
| |
Collapse
|
37
|
Noritake A, Ninomiya T, Isoda M. Subcortical encoding of agent-relevant associative signals for adaptive social behavior in the macaque. Neurosci Biobehav Rev 2021; 125:78-87. [PMID: 33609569 DOI: 10.1016/j.neubiorev.2021.02.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/24/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
Primates are group-living creatures that constantly face the challenges posed by complex social demands. To date, the cortical mechanisms underlying social information processing have been the major focus of attention. However, emerging evidence suggests that subcortical regions also mediate the collection and processing of information from other agents. Here, we review the literature supporting the hypothesis that behavioral variables important for decision-making, i.e., stimulus, action, and outcome, are associated with agent information (self and other) in subcortical regions, such as the amygdala, striatum, lateral hypothalamus, and dopaminergic midbrain nuclei. Such self-relevant and other-relevant associative signals are then integrated into a social utility signal, presumably at the level of midbrain dopamine neurons. This social utility signal allows decision makers to organize their optimal behavior in accordance with social demands. Determining how self-relevant and other-relevant signals might be altered in psychiatric and neurodevelopmental disorders will be fundamental to better understand how social behaviors are dysregulated in disease conditions.
Collapse
Affiliation(s)
- Atsushi Noritake
- Division of Behavioral Development, Department of System Neuroscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, 38 Myodaiji, Okazaki, Aichi, 444-8585, Japan; Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Hayama, Kanagawa, 240-0193, Japan
| | - Taihei Ninomiya
- Division of Behavioral Development, Department of System Neuroscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, 38 Myodaiji, Okazaki, Aichi, 444-8585, Japan; Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Hayama, Kanagawa, 240-0193, Japan
| | - Masaki Isoda
- Division of Behavioral Development, Department of System Neuroscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, 38 Myodaiji, Okazaki, Aichi, 444-8585, Japan; Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Hayama, Kanagawa, 240-0193, Japan.
| |
Collapse
|
38
|
Berteotti C, Liguori C, Pace M. Dysregulation of the orexin/hypocretin system is not limited to narcolepsy but has far-reaching implications for neurological disorders. Eur J Neurosci 2020; 53:1136-1154. [PMID: 33290595 DOI: 10.1111/ejn.15077] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 12/23/2022]
Abstract
Neuropeptides orexin A and B (OX-A/B, also called hypocretin 1 and 2) are released selectively by a population of neurons which projects widely into the entire central nervous system but is localized in a restricted area of the tuberal region of the hypothalamus, caudal to the paraventricular nucleus. The OX system prominently targets brain structures involved in the regulation of wake-sleep state switching, and also orchestrates multiple physiological functions. The degeneration and dysregulation of the OX system promotes narcoleptic phenotypes both in humans and animals. Hence, this review begins with the already proven involvement of OX in narcolepsy, but it mainly discusses the new pre-clinical and clinical insights of the role of OX in three major neurological disorders characterized by sleep impairment which have been recently associated with OX dysfunction, such as Alzheimer's disease, stroke and Prader Willi syndrome, and have been emerged over the past 10 years to be strongly associated with the OX dysfunction and should be more considered in the future. In the light of the impairment of the OX system in these neurological disorders, it is conceivable to speculate that the integrity of the OX system is necessary for a healthy functioning body.
Collapse
Affiliation(s)
- Chiara Berteotti
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Claudio Liguori
- Sleep Medicine Centre, Neurology Unit, University Hospital Tor Vergata, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Marta Pace
- Genetics and Epigenetics of Behaviour Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
39
|
Prajapati SK, Krishnamurthy S. Non-selective orexin-receptor antagonist attenuates stress-re-stress-induced core PTSD-like symptoms in rats: Behavioural and neurochemical analyses. Behav Brain Res 2020; 399:113015. [PMID: 33212086 DOI: 10.1016/j.bbr.2020.113015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 01/21/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a psychological disorder affecting many around the world. Growing evidence suggests that orexin-A is involved in the pathophysiology of depression and panic anxiety disorder. However, the role of orexin-A in PTSD remains unclear. Therefore, pharmacological manipulation of orexin-A can be a potential approach for the treatment of PTSD. Male Wistar rats were subjected to stress re-stress (SRS) by restraining them for 2 h followed by foot shock (FS) and halothane exposure on day-2 (D-2). Then the rats were weekly exposed to FS as re-stress cue . Suvorexant, an orexin antagonist (10, 20 and 30 mg/kg p.o.) and paroxetine (10 mg/kg p.o.) were administered from D-8 to D-32. Plasma and cerebrospinal fluid (CSF) were collected for corticosterone and orexin-A measurement. The analysis of serotonin and corticotropin-releasing factor receptor-1 (CRF-R1) were performed in the amygdalar tissue. SRS-induced PTSD-like symptoms like fear response, anxiety-like behaviour and hypocorticosteronism were attenuated by suvorexant and paroxetine. Interestingly, SRS exposed rats showed activation of orexin-A and serotonergic systems, which were also attenuated by suvorexant. Additionally, suvorexant ameliorated the extrahypothalamic induced upregulation of CRH-R1 in SRS-exposed rats. Therefore, orexin-A may be considered as a neurochemical-marker for PTSD and suvorexant alleviated PTSD-like symptoms through modulating orexinergic, serotonergic and neuroendocrine systems.
Collapse
Affiliation(s)
- Santosh Kumar Prajapati
- Neurotherapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi- 221 005, U.P., India
| | - Sairam Krishnamurthy
- Neurotherapeutics Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi- 221 005, U.P., India.
| |
Collapse
|
40
|
Tenorio-Lopes L, Fournier S, Henry MS, Bretzner F, Kinkead R. Disruption of estradiol regulation of orexin neurons: a novel mechanism in excessive ventilatory response to CO 2 inhalation in a female rat model of panic disorder. Transl Psychiatry 2020; 10:394. [PMID: 33173029 PMCID: PMC7656265 DOI: 10.1038/s41398-020-01076-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/01/2020] [Accepted: 10/20/2020] [Indexed: 12/27/2022] Open
Abstract
Panic disorder (PD) is ~2 times more frequent in women. An excessive ventilatory response to CO2 inhalation is more likely during the premenstrual phase. While ovarian hormones appear important in the pathophysiology of PD, their role remains poorly understood as female animals are rarely used in pre-clinical studies. Using neonatal maternal separation (NMS) to induce a "PD-like" respiratory phenotype, we tested the hypothesis that NMS disrupts hormonal regulation of the ventilatory response to CO2 in female rats. We then determined whether NMS attenuates the inhibitory actions of 17-β estradiol (E2) on orexin neurons (ORX). Pups were exposed to NMS (3 h/day; postnatal day 3-12). The ventilatory response to CO2-inhalation was tested before puberty, across the estrus cycle, and following ovariectomy. Plasma E2 and hypothalamic ORXA were measured. The effect of an ORX1 antagonist (SB334867; 15 mg/kg) on the CO2 response was tested. Excitatory postsynaptic currents (EPSCs) were recorded from ORX neurons using whole-cell patch-clamp. NMS-related increase in the CO2 response was observed only when ovaries were functional; the largest ventilation was observed during proestrus. SB334867 blocked this effect. NMS augmented levels of ORXA in hypothalamus extracts. EPSC frequency varied according to basal plasma E2 levels across the estrus cycle in controls but not NMS. NMS reproduces developmental and cyclic changes of respiratory manifestations of PD. NMS disrupts the inhibitory actions of E2 on the respiratory network. Impaired E2-related inhibition of ORX neurons during proestrus is a novel mechanism in respiratory manifestations of PD in females.
Collapse
Affiliation(s)
- Luana Tenorio-Lopes
- Hotchkiss Brain Institute; Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Stéphanie Fournier
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec. Département de Pédiatrie. Université Laval, Québec, QC, Canada
| | - Mathilde S Henry
- INRAE, Université de Bordeaux, Bordeaux INP, Nutrineuro, UMR 1286, F-33000, Bordeaux, France
| | - Frédéric Bretzner
- Centre de Recherche du CHU de Québec-Université Laval, Axe Neurosciences. Département de Psychiatrie et de Neurosciences, Université Laval, Québec, QC, Canada
| | - Richard Kinkead
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec. Département de Pédiatrie. Université Laval, Québec, QC, Canada.
| |
Collapse
|
41
|
Faesel N, Kolodziejczyk MH, Koch M, Fendt M. Orexin deficiency affects sociability and the acquisition, expression, and extinction of conditioned social fear. Brain Res 2020; 1751:147199. [PMID: 33160959 DOI: 10.1016/j.brainres.2020.147199] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/06/2020] [Accepted: 11/01/2020] [Indexed: 12/19/2022]
Abstract
Accumulating evidence indicates that the central orexin (hypocretin) system plays an important role in regulating emotional processes in both humans and rodents. Thus, the orexin system has been repeatedly implicated in the pathophysiology of several neuropsychiatric disorders, such as anxiety disorders. Among others, symptoms like social fear and social withdrawal are frequently observed in these disorders. Based on this, we investigated the role of orexin deficiency in social (fear) behavior. For that, female and male orexin-deficient mice were tested for (1) sociability and social novelty, and (2) acquisition, expression, and extinction of conditioned social fear. We found that female orexin-deficient mice displayed reduced sociability and decreased preference for social novelty compared to their wild-type littermates. These effects of orexin deficiency were not observed in males. Moreover, orexin deficiency facilitated the acquisition and/or expression of conditioned social fear and impaired the extinction of social fear in both sexes. Taken together, our results indicate an important, partly sex-dependent, regulatory role of the orexin system in social (fear) behavior. Our findings support the hypothesis of orexin being an integrator of motivation, affect, and emotion.
Collapse
Affiliation(s)
- Nadine Faesel
- Institute for Pharmacology and Toxicology, Otto von Guericke University Magdeburg, Leipziger Straße 44, D-39120 Magdeburg, Germany; Department of Neuropharmacology, Brain Research Institute, University of Bremen, Hochschulring 18, D-28359 Bremen, Germany.
| | - Malgorzata H Kolodziejczyk
- Institute for Pharmacology and Toxicology, Otto von Guericke University Magdeburg, Leipziger Straße 44, D-39120 Magdeburg, Germany
| | - Michael Koch
- Department of Neuropharmacology, Brain Research Institute, University of Bremen, Hochschulring 18, D-28359 Bremen, Germany
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Otto von Guericke University Magdeburg, Leipziger Straße 44, D-39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University Magdeburg, Leipziger Straße 44, D-39120 Magdeburg, Germany
| |
Collapse
|
42
|
James MH, Fragale JE, O'Connor SL, Zimmer BA, Aston-Jones G. The orexin (hypocretin) neuropeptide system is a target for novel therapeutics to treat cocaine use disorder with alcohol coabuse. Neuropharmacology 2020; 183:108359. [PMID: 33091458 DOI: 10.1016/j.neuropharm.2020.108359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/05/2020] [Accepted: 10/15/2020] [Indexed: 12/14/2022]
Abstract
An estimated 50-90% of individuals with cocaine use disorder (CUD) also report using alcohol. Cocaine users report coabusing alcohol to 'self-medicate' against the negative emotional side effects of the cocaine 'crash', including the onset of anxiety. Thus, pharmaceutical strategies to treat CUD would ideally reduce the motivational properties of cocaine, alcohol, and their combination, as well as reduce the onset of anxiety during drug withdrawal. The hypothalamic orexin (hypocretin) neuropeptide system offers a promising target, as orexin neurons are critically involved in activating behavioral and physiological states to respond to both positive and negative motivators. Here, we seek to describe studies demonstrating efficacy of orexin receptor antagonists in reducing cocaine, alcohol- and stress-related behaviors, but note that these studies have largely focused on each of these phenomena in isolation. For orexin-based compounds to be viable in the clinical setting, we argue that it is imperative that their efficacy be tested in animal models that account for polysubstance use patterns. To begin to examine this, we present new data showing that rats' preferred level of cocaine intake is significantly increased following chronic homecage access to alcohol. We also report that cocaine intake and motivation are reduced by a selective orexin-1 receptor antagonist when rats have a history of cocaine + alcohol, but not a limited history of cocaine alone. In light of these proof-of-principle data, we outline what we believe to be the key priorities going forward with respect to further examining the orexin system in models of polysubstance use. This article is part of the special issue on Neurocircuitry Modulating Drug and Alcohol Abuse.
Collapse
Affiliation(s)
- Morgan H James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA; Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA; Florey Institute of Neuroscience and Mental Health, University of Melbourne, VIC, Australia
| | - Jennifer E Fragale
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Shayna L O'Connor
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA; Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Benjamin A Zimmer
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Gary Aston-Jones
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA; Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA.
| |
Collapse
|
43
|
Salvadore G, Bonaventure P, Shekhar A, Johnson PL, Lord B, Shireman BT, Lebold TP, Nepomuceno D, Dugovic C, Brooks S, Zuiker R, Bleys C, Tatikola K, Remmerie B, Jacobs GE, Schruers K, Moyer J, Nash A, Van Nueten LGM, Drevets WC. Translational evaluation of novel selective orexin-1 receptor antagonist JNJ-61393215 in an experimental model for panic in rodents and humans. Transl Psychiatry 2020; 10:308. [PMID: 32895369 PMCID: PMC7477545 DOI: 10.1038/s41398-020-00937-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/08/2020] [Accepted: 07/14/2020] [Indexed: 11/24/2022] Open
Abstract
Orexin neurons originating in the perifornical and lateral hypothalamic area project to anxiety- and panic-associated neural circuitry, and are highly reactive to anxiogenic stimuli. Preclinical evidence suggests that the orexin system, and particularly the orexin-1 receptor (OX1R), may be involved in the pathophysiology of panic and anxiety. Selective OX1R antagonists thus may constitute a potential new treatment strategy for panic- and anxiety-related disorders. Here, we characterized a novel selective OX1R antagonist, JNJ-61393215, and determined its affinity and potency for human and rat OX1R in vitro. We also evaluated the safety, pharmacokinetic, and pharmacodynamic properties of JNJ-61393215 in first-in-human single- and multiple-ascending dose studies conducted. Finally, the potential anxiolytic effects of JNJ-61393215 were evaluated both in rats and in healthy men using 35% CO2 inhalation challenge to induce panic symptoms. In the rat CO2 model of panic anxiety, JNJ-61393215 demonstrated dose-dependent attenuation of CO2-induced panic-like behavior without altering baseline locomotor or autonomic activity, and had minimal effect on spontaneous sleep. In phase-1 human studies, JNJ-61393215 at 90 mg demonstrated significant reduction (P < 0.02) in CO2-induced fear and anxiety symptoms that were comparable to those obtained using alprazolam. The most frequently reported adverse events were somnolence and headache, and all events were mild in severity. These results support the safety, tolerability, and anxiolytic effects of JNJ-61393215, and validate CO2 exposure as a translational cross-species experimental model to evaluate the therapeutic potential of novel anxiolytic drugs.
Collapse
Affiliation(s)
- Giacomo Salvadore
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, Titusville, NJ USA
| | | | - Anantha Shekhar
- grid.257413.60000 0001 2287 3919Departments of Psychiatry, and Pharmacology, Indiana University, School of Medicine, Indianapolis, IN USA
| | - Philip L. Johnson
- grid.257413.60000 0001 2287 3919Department of Anatomy, Physiology and Cell Biology, Indiana University, School of Medicine, Indianapolis, IN USA
| | - Brian Lord
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, San Diego, CA USA
| | - Brock T. Shireman
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, San Diego, CA USA
| | - Terry P. Lebold
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, San Diego, CA USA
| | - Diane Nepomuceno
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, San Diego, CA USA
| | - Christine Dugovic
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, San Diego, CA USA
| | - Sander Brooks
- grid.418011.d0000 0004 0646 7664Centre for Human Drug Research, Leiden, The Netherlands ,grid.10419.3d0000000089452978Leiden University Medical Center, Leiden, The Netherlands
| | - Rob Zuiker
- grid.418011.d0000 0004 0646 7664Centre for Human Drug Research, Leiden, The Netherlands
| | - Cathy Bleys
- grid.419619.20000 0004 0623 0341Janssen Research & Development, LLC, Beerse, Belgium
| | - Kanaka Tatikola
- grid.497530.c0000 0004 0389 4927Janssen Scientific Affairs, LLC, Titusville, NJ USA
| | - Bart Remmerie
- grid.419619.20000 0004 0623 0341Janssen Research & Development, LLC, Beerse, Belgium
| | - Gabriel E. Jacobs
- grid.418011.d0000 0004 0646 7664Centre for Human Drug Research, Leiden, The Netherlands ,grid.10419.3d0000000089452978Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
| | - Koen Schruers
- grid.5012.60000 0001 0481 6099Research School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - John Moyer
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, Titusville, NJ USA
| | - Abigail Nash
- grid.497530.c0000 0004 0389 4927Janssen Scientific Affairs, LLC, Titusville, NJ USA
| | - Luc G. M. Van Nueten
- grid.419619.20000 0004 0623 0341Janssen Research & Development, LLC, Beerse, Belgium
| | - Wayne C. Drevets
- grid.497530.c0000 0004 0389 4927Janssen Research & Development, LLC, San Diego, CA USA
| |
Collapse
|
44
|
Burdakov D, Peleg-Raibstein D. The hypothalamus as a primary coordinator of memory updating. Physiol Behav 2020; 223:112988. [DOI: 10.1016/j.physbeh.2020.112988] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/05/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022]
|
45
|
Pulver A, Kiive E, Kanarik M, Harro J. Association of orexin/hypocretin receptor gene (HCRTR1) with reward sensitivity, and interaction with gender. Brain Res 2020; 1746:147013. [PMID: 32652147 DOI: 10.1016/j.brainres.2020.147013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022]
Abstract
Orexins/hypocretins maintain wakefulness, increase appetite and participate in the coordination of stress response. We have recently provided evidence on the role of orexins in aggression, showing the association of the HCRTR1 genotype. (rs2271933 G > A; leading to amino acid substitution Ile408Val) with aggressiveness or breach of law in four independent cohorts. Aggressive behaviour can be reward driven and hence we have examined the association of HCRTR1 rs2271933 genotype with different aspects of reward sensitivity in the birth cohort representative Estonian Children Personality Behaviour and Health Study. HCRTR1 genotype was associated with reward sensitivity in a gender dependent manner. Male HCRTR1 A/A homozygotes had higher Openness to Rewards and the overall reward sensitivity score while, in contrast, female A/A homozygotes scored lower than G-allele carriers in Openness to Rewards. In the total sample, aggressiveness correlated positively with reward sensitivity, but this was on account of Insatiability by Reward. In contrast, the HCRTR1 A/A homozygotes had a positive association of aggressiveness and Openness to Rewards. Experience of stressful life events had a small but significant increasing effect on both aspects of reward sensitivity, and correlated in an anomalous way with reward sensitivity in the HCRTR1 A/A homozygotes. Conclusively, the higher aggressiveness of HCRTR1 A/A homozygotes appears based on a qualitative difference in sensitivity to rewards, in the form that suggests their lower ability to prevent responses to challenges being converted into overt aggression.
Collapse
Affiliation(s)
- Aleksander Pulver
- School of Natural Sciences and Health, Tallinn University, Narva Road 29, Astra Building, 10120 Tallinn, Estonia
| | - Evelyn Kiive
- Division of Special Education, Department of Education, University of Tartu, Näituse 2, 50409 Tartu, Estonia
| | - Margus Kanarik
- Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Ravila 14A Chemicum, 50411 Tartu, Estonia
| | - Jaanus Harro
- School of Natural Sciences and Health, Tallinn University, Narva Road 29, Astra Building, 10120 Tallinn, Estonia; Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Ravila 14A Chemicum, 50411 Tartu, Estonia.
| |
Collapse
|
46
|
Abstract
Objectives: The relationships between orexins and stress-related conditions have been well documented in animal studies. However, human studies confirming this relationship are limited. The aim of this study was to investigate the association between orexin-A and anxiety disorders in adolescents. Additionally, we aimed to examine the relationship between orexin-A and cortisol levels in those with anxiety disorders.Methods: A total of 56 medication-free adolescents diagnosed with any anxiety disorder, except for specific phobias, and 32 healthy controls were included in this study. Depression, state and trait anxiety levels of the participants were measured using self-report scales. Orexin-A and cortisol levels were measured by an enzyme-linked immunosorbent assay (ELISA).Results: Analysis of covariance (ANCOVA) indicated that serum orexin-A levels were significantly higher in the anxiety disorder group than in the control group while controlling for age, sex and depression levels. After controlling for age and sex, orexin-A levels were positively and negatively correlated to depression and cortisol levels, respectively. In addition, a positive correlation trend between trait anxiety and orexin-A was found.Conclusions: Orexin-A levels are higher in adolescents with anxiety disorder; however, depressive symptoms should be considered when investigating this relationship.
Collapse
Affiliation(s)
- Ömer Faruk Akça
- Department of Child and Adolescent Psychiatry, Necmettin Erbakan University Meram School of Medicine, Konya, Turkey
| | - Necati Uzun
- Department of Child and Adolescent Psychiatry, Dr. Ali Kemal Belviranlı Children Hospital, Konya, Turkey
| | - İbrahim Kılınç
- Department of Biochemistry, Necmettin Erbakan University Meram School of Medicine, Konya, Turkey
| |
Collapse
|
47
|
Salehabadi S, Abrari K, Elahdadi Salmani M, Nasiri M, Lashkarbolouki T. Investigating the role of the amygdala orexin receptor 1 in memory acquisition and extinction in a rat model of PTSD. Behav Brain Res 2020; 384:112455. [DOI: 10.1016/j.bbr.2019.112455] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 11/15/2022]
|
48
|
Han Y, Yuan K, Zheng Y, Lu L. Orexin Receptor Antagonists as Emerging Treatments for Psychiatric Disorders. Neurosci Bull 2020; 36:432-448. [PMID: 31782044 PMCID: PMC7142186 DOI: 10.1007/s12264-019-00447-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 09/27/2019] [Indexed: 12/12/2022] Open
Abstract
Orexins comprise two neuropeptides produced by orexin neurons in the lateral hypothalamus and are released by extensive projections of these neurons throughout the central nervous system. Orexins bind and activate their associated G protein-coupled orexin type 1 receptors (OX1Rs) and OX2Rs and act on numerous physiological processes, such as sleep-wake regulation, feeding, reward, emotion, and motivation. Research on the development of orexin receptor antagonists has dramatically increased with the approval of suvorexant for the treatment of primary insomnia. In the present review, we discuss recent findings on the involvement of the orexin system in the pathophysiology of psychiatric disorders, including sleep disorders, depression, anxiety, and drug addiction. We discuss the actions of orexin receptor antagonists, including selective OX1R antagonists (SORA1s), selective OX2R antagonists (SORA2s), and dual OX1/2R antagonists (DORAs), in the treatment of these disorders based on both preclinical and clinical evidence. SORA2s and DORAs have more pronounced efficacy in the treatment of sleep disorders, whereas SORA1s may be promising for the treatment of anxiety and drug addiction. We also discuss potential challenges and opportunities for the application of orexin receptor antagonists to clinical interventions.
Collapse
Affiliation(s)
- Ying Han
- National Institute of Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Yongbo Zheng
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Lin Lu
- National Institute of Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China.
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China.
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
49
|
Representation of distinct reward variables for self and other in primate lateral hypothalamus. Proc Natl Acad Sci U S A 2020; 117:5516-5524. [PMID: 32094192 PMCID: PMC7071915 DOI: 10.1073/pnas.1917156117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Motivation is affected by rewards to both oneself and others. Which brain regions separately monitor self-rewards and other-rewards? It has been thought that higher-order, neocortical regions, such as the medial prefrontal cortex, monitor behavioral information in agent-selective manners. Here, we show that a subcortical region called the lateral hypothalamus (LH), an evolutionarily old structure in the vertebrate brain, also contains agent-specific reward information and further integrates it into a subjective reward value. This other-reward–dependent value signal is causally used for adaptive behavior, because deactivation of LH cells totally eliminates the motivational impact of other-rewards. Our findings indicate that the LH is an integral component of social brain networks and shapes socially motivated behavior via functional coordination with neocortical regions. The lateral hypothalamus (LH) has long been implicated in maintaining behavioral homeostasis essential for the survival of an individual. However, recent evidence suggests its more widespread roles in behavioral coordination, extending to the social domain. The neuronal and circuit mechanisms behind the LH processing of social information are unknown. Here, we show that the LH represents distinct reward variables for “self” and “other” and is causally involved in shaping socially motivated behavior. During a Pavlovian conditioning procedure incorporating ubiquitous social experiences where rewards to others affect one’s motivation, LH cells encoded the subjective value of self-rewards, as well as the likelihood of self- or other-rewards. The other-reward coding was not a general consequence of other’s existence, but a specific effect of other’s reward availability. Coherent activity with and top-down information flow from the medial prefrontal cortex, a hub of social brain networks, contributed to signal encoding in the LH. Furthermore, deactivation of LH cells eliminated the motivational impact of other-rewards. These results indicate that the LH constitutes a subcortical node in social brain networks and shapes one’s motivation by integrating cortically derived, agent-specific reward information.
Collapse
|
50
|
Cohen S, Matar MA, Vainer E, Zohar J, Kaplan Z, Cohen H. Significance of the orexinergic system in modulating stress-related responses in an animal model of post-traumatic stress disorder. Transl Psychiatry 2020; 10:10. [PMID: 32066707 PMCID: PMC7026175 DOI: 10.1038/s41398-020-0698-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 10/02/2019] [Accepted: 10/20/2019] [Indexed: 01/23/2023] Open
Abstract
Converging evidence indicates that orexins (ORXs), the regulatory neuropeptides, are implicated in anxiety- and depression-related behaviors via the modulation of neuroendocrine, serotonergic, and noradrenergic systems. This study evaluated the role of the orexinergic system in stress-associated physiological responses in a controlled prospective animal model. The pattern and time course of activation of hypothalamic ORX neurons in response to predator-scent stress (PSS) were examined using c-Fos as a marker for neuronal activity. The relationship between the behavioral response pattern 7 days post-exposure and expressions of ORXs was evaluated. We also investigated the effects of intracerebroventricular microinfusion of ORX-A or almorexant (ORX-A/B receptor antagonist) on behavioral responses 7 days following PSS exposure. Hypothalamic levels of ORX-A, neuropeptide Y (NPY), and brain-derived neurotrophic factor (BDNF) were assessed. Compared with rats whose behaviors were extremely disrupted (post-traumatic stress disorder [PTSD]-phenotype), those whose behaviors were minimally selectively disrupted displayed significantly upregulated ORX-A and ORX-B levels in the hypothalamic nuclei. Intracerebroventricular microinfusion of ORX-A before PSS reduced the prevalence of the PTSD phenotype compared with that of artificial cerebrospinal fluid or almorexant, and rats treated with almorexant displayed a higher prevalence of the PTSD phenotype than did untreated rats. Activated ORX neurons led to upregulated expressions of BDNF and NPY, which might provide an additional regulatory mechanism for the modulation of adaptive stress responses. The study indicates that the activated ORX system might promote adaptive responses to PSS probably via stimulation of BDNF and NPY secretion, and early intervention with ORX-A reduces the prevalence of the PTSD phenotype and increases the prevalence of adaptive phenotypes. The findings provide some insights into the mechanisms underlying the involvement of the ORX system in stress-related disorders.
Collapse
Affiliation(s)
- Shlomi Cohen
- Ministry of Health, Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer- Sheva, Israel
- Department of Psychology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Michael A Matar
- Ministry of Health, Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer- Sheva, Israel
| | - Ella Vainer
- Ministry of Health, Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer- Sheva, Israel
| | - Joseph Zohar
- Division of Psychiatry, The Chaim Sheba Medical Center, Ramat-Gan, Israel
- Sackler Medical School, Tel-Aviv University, Tel-Aviv, Israel
| | - Zeev Kaplan
- Ministry of Health, Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer- Sheva, Israel
| | - Hagit Cohen
- Ministry of Health, Beer-Sheva Mental Health Center, Anxiety and Stress Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer- Sheva, Israel.
- Department of Psychology, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|