1
|
Peter-Derex L, Fort E, Putois B, Martel N, Ricordeau F, Bastuji H, Arnulf I, Barateau L, Bourgin P, Dauvilliers Y, Debs R, Dodet P, Dudoignon B, Franco P, Hartley S, Lambert I, Lecendreux M, Leclair-Visonneau L, Léger D, Lemesle-Martin M, Léotard A, Leu-Semenescu S, Limousin N, Lopez R, Meslier N, Micoulaud-Franchi JA, Charley-Mocana C, d'Ortho MP, Philip P, Ruppert E, de La Tullaye S, Brigandet M, Charbotel B, Mazza S, Rolland B. Determinants of substance use patterns in patients with narcolepsy type 1: A multi-center comparative cross-sectional study. Sleep Med 2025; 129:148-166. [PMID: 40024108 DOI: 10.1016/j.sleep.2025.02.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/16/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVES In this multi-center cross-sectional study, we compared substance use patterns (SUPs) between patients with narcolepsy type 1 (NT1) and controls, and investigated, among patients, factors associated with the consumption of the main psychoactive substances. METHODS Adult patients with NT1 and controls completed questionnaires about tobacco, alcohol, and cannabis use patterns. Unadjusted bivariable then multivariate analyses (adjusted for sex, age, education, family status, and depression) were performed to compare SUPs between controls and patients, and to explore socio-demographic, psycho-behavioral, and clinical determinants of consumptions. RESULTS We included 235 patients (63.8 % women, 36.4 ± 14.7 years) and 166 controls (69.9 % women, 40.3 ± 14.4 years). Substances co-consumptions were frequent in both groups. Patients with NT1 were more frequently current smokers (32.3 % vs. 20.1 %, p < 0.01) or e-cigarettes users (12.1 % vs 2.4 %, p < 0.001) than controls, while no difference was observed for cannabis use and alcohol misuse. Only the increased likelihood of vaping remained significant in adjusted analysis. Among NT1 patients, smoking was associated with disrupted nighttime sleep (OR[95%CI] = 2.28[1.02-5.12], p < 0.05) and less obesity (OR = 0.24[0.09-0.59], p < 0.05). Alcohol misuse was associated with sleep paralysis (OR = 2.11[1.13-3.91], p < 0.05) and treatments (modafinil: OR = 2.14[1.15-4.01], p < 0.05; sodium oxybate: OR = 0.41[0.17-0.97], p < 0.05). Tobacco and cannabis consumptions were associated with lower physical activity (OR = 0.46 [0.24-0.87], p < 0.05 and OR = 0.25[0.10-0.66], p < 0.01). Alcohol misuse and cannabis use were associated with rule breaking behaviors (OR = 5.89[1.61-21.60], p < 0.05 and OR = 8.52[1.79-40.48], p = 0.01). CONCLUSION Patients with NT1 do not seem less vulnerable to psychoactive substance use/misuse. Consumptions patterns are associated with multiple dimensions of the disease including sleep-related symptoms, comorbidities, treatments, and psycho-behavioral factors.
Collapse
Affiliation(s)
- Laure Peter-Derex
- Centre for Sleep Medicine and Respiratory Diseases, National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon 1 University, Lyon, France; Lyon Neuroscience Research Centre, INSERM U1028, CNRS UMR 5292, Lyon, France.
| | - Emmanuel Fort
- Transport Work and Environmental Epidemiology Research and Surveillance Unit - UMRESTTE (UMR T9405), University Lyon 1, Lyon, France
| | - Benjamin Putois
- Lyon Neuroscience Research Centre, INSERM U1028, CNRS UMR 5292, Lyon, France; Swiss Distance Learning University, Faculty of Psychology, Brig, Switzerland
| | - Nora Martel
- Clinical Research Center, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France
| | - François Ricordeau
- Centre for Sleep Medicine and Respiratory Diseases, National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon 1 University, Lyon, France; Lyon Neuroscience Research Centre, INSERM U1028, CNRS UMR 5292, Lyon, France
| | - Hélène Bastuji
- Centre for Sleep Medicine and Respiratory Diseases, National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon 1 University, Lyon, France; Lyon Neuroscience Research Centre, INSERM U1028, CNRS UMR 5292, Lyon, France
| | - Isabelle Arnulf
- National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Sleep Disorders Clinic, Pitié-Salpêtrière Hospital, APHP-Sorbonne University, Paris, France
| | - Lucie Barateau
- National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Sleep Unit, Department of Neurology, CHU Montpellier, Univ Montpellier, Montpellier, France
| | - Patrice Bourgin
- CIRCSom (International Research Center for ChronoSomnology), Sleep Disorders Center, National Competence Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Strasbourg University Hospital, Strasbourg, France
| | - Yves Dauvilliers
- National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Sleep Unit, Department of Neurology, CHU Montpellier, Univ Montpellier, Montpellier, France
| | - Rachel Debs
- Sleep Unit, Department of Neurology, National Competence Centre for Orphan Diseases, Narcolepsy- Rare Hypersomnias, Pierre-Paul Riquet/Purpan University Hospital, Toulouse, France
| | - Pauline Dodet
- National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Sleep Disorders Clinic, Pitié-Salpêtrière Hospital, APHP-Sorbonne University, Paris, France
| | - Benjamin Dudoignon
- Pediatric Sleep Disorders Center, National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Robert Debre Hospital, APHP, Paris, France
| | - Patricia Franco
- Lyon Neuroscience Research Centre, INSERM U1028, CNRS UMR 5292, Lyon, France; Pediatric Sleep Unit and National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Mother-Children's Hospital, Hospices Civils de Lyon, Lyon, France
| | - Sarah Hartley
- Sleep Unit, Physiology Department, National Competence Centre for Orphan Diseases, Narcolepsy- Rare Hypersomnias, GHU Paris-Saclay, APHP, Raymond Poincaré Hospital, Garches, France
| | - Isabelle Lambert
- Sleep Unit, Epileptology and Cerebral Rhythmology, National Competence Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Timone Hospital, APHM, Marseille, France
| | - Michel Lecendreux
- Pediatric Sleep Disorders Center, National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Robert Debre Hospital, APHP, Paris, France
| | - Laurene Leclair-Visonneau
- Department of Clinical Neurophysiology, National Competence Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, CHU de Nantes, Nantes, France
| | - Damien Léger
- APHP, Hôtel-Dieu, Centre Du Sommeil et de La Vigilance, National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Université Paris Cité, VIFASOM, Paris, France
| | - Martine Lemesle-Martin
- Department of Clinical Neurophysiology, National Competence Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, University Hospital of Dijon, Dijon, France
| | - Antoine Léotard
- Sleep Unit, Physiology Department, National Competence Centre for Orphan Diseases, Narcolepsy- Rare Hypersomnias, GHU Paris-Saclay, APHP, Raymond Poincaré Hospital, Garches, France
| | - Smaranda Leu-Semenescu
- National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Sleep Disorders Clinic, Pitié-Salpêtrière Hospital, APHP-Sorbonne University, Paris, France
| | - Nadège Limousin
- Department of Neurology and Clinical Neurophysiology, National Competence Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, University Hospital Bretonneau, Tours, France
| | - Régis Lopez
- National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Sleep Unit, Department of Neurology, CHU Montpellier, Univ Montpellier, Montpellier, France
| | - Nicole Meslier
- Department of Respiratory and Sleep Medicine, National Competence Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias Angers University Hospital, Angers, France
| | - Jean-Arthur Micoulaud-Franchi
- Sleep Disorders Department, National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, CHU Bordeaux, Bordeaux, France
| | - Christelle Charley-Mocana
- Neurophysiologie Clinique, National Competence Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Hôpital Roger Salengro, Lille, France
| | - Marie-Pia d'Ortho
- Service de Physiologie - Explorations Fonctionnelles, National Competence Centre for Orphan Diseases, Narcolepsy- Rare Hypersomnias, Hôpital Bichat, AP-HP, Université Paris Cité, Inserm, NeuroDiderot, Paris, France
| | - Pierre Philip
- Sleep Disorders Department, National Reference Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, CHU Bordeaux, Bordeaux, France
| | - Elisabeth Ruppert
- CIRCSom (International Research Center for ChronoSomnology), Sleep Disorders Center, National Competence Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, Strasbourg University Hospital, Strasbourg, France
| | - Sylvie de La Tullaye
- Department of Clinical Neurophysiology, National Competence Centre for Orphan Diseases, Narcolepsy-Rare Hypersomnias, CHU de Nantes, Nantes, France
| | - Manon Brigandet
- ANC, Association for Patients with Narcolepsy Cataplexy and Rare Hypersomnias, France
| | - Barbara Charbotel
- Transport Work and Environmental Epidemiology Research and Surveillance Unit - UMRESTTE (UMR T9405), University Lyon 1, Lyon, France
| | - Stéphanie Mazza
- Lyon Neuroscience Research Centre, INSERM U1028, CNRS UMR 5292, Lyon, France
| | - Benjamin Rolland
- Lyon Neuroscience Research Centre, INSERM U1028, CNRS UMR 5292, Lyon, France; Service Universitaire D'Addictologie de Lyon, Centre Hospitalier Le Vinatier, Hospices Civils de Lyon, Lyon 1 University, Lyon, France
| |
Collapse
|
2
|
Duske J, D'Souza N, Mayer D, Dieterich DC, Fendt M. Orexinergic modulation of chronic jet lag-induced deficits in mouse cognitive flexibility. Neuropsychopharmacology 2025; 50:762-771. [PMID: 39478089 PMCID: PMC11914050 DOI: 10.1038/s41386-024-02017-8] [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: 07/09/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 03/19/2025]
Abstract
Cognitive flexibility and working memory are important executive functions mediated by the prefrontal cortex and can be impaired by circadian rhythm disturbances such as chronic jet lag (CJL) or shift work. In the present study, we used mice to investigate whether (1) simulated CJL impairs cognitive flexibility, (2) the orexin system is involved in such impairment, and (3) nasal administration of orexin A is able to reverse CJL-induced deficits in cognitive flexibility and working memory. Mice were exposed to either standard light-dark conditions or simulated CJL consisting of series of advance time shifts. Experiment (1) investigated the effects of a mild CJL protocol on cognitive flexibility using the attentional set shifting task. Experiment (2) used a stronger CJL protocol and examined CJL effects on the orexin system utilizing c-Fos and orexin immunohistochemistry. Experiment (3) tested whether nasal orexin application can rescue CJL-induced deficits in cognitive flexibility and working memory, the latter by measuring spontaneous alternation in the Y-maze. The present data show that CJL (1) impairs cognitive flexibility and (2) reduces the activity of orexin neurons in the lateral hypothalamus. (3) Nasal administration of orexin A rescued CJL-induced deficits in working memory and cognitive flexibility. These findings suggest that executive function impairments by circadian rhythm disturbances such as CJL are caused by dysregulation of orexinergic input to the prefrontal cortex. Compensation of decreased orexinergic input by nasal administration of orexin A could be a potential therapy for CJL- or shift work-induced human deficits in executive functions.
Collapse
Affiliation(s)
- Julius Duske
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Nicole D'Souza
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Institute of Neurophysiology, Goethe University, Frankfurt, Germany
| | - Dana Mayer
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniela C Dieterich
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center of Behavioural Brain Sciences, Otto-von-Guericke University, Magdeburg, Germany
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
- Center of Behavioural Brain Sciences, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
3
|
Raymond JS, Athanasopoulos AG, Badolato CJ, Doolan TJ, Scicluna RL, Everett NA, Bowen MT, James MH. Emerging medications and pharmacological treatment approaches for substance use disorders. Pharmacol Biochem Behav 2025; 248:173952. [PMID: 39719161 DOI: 10.1016/j.pbb.2024.173952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Medications to treat substance use disorders (SUDs) remain suboptimal or, in the case of stimulants and cannabis, non-existent. Many factors have contributed to this paucity, including the biological complexity of addiction, regulatory challenges, and a historical lack of enthusiasm among pharmaceutical companies to commit resources to this disease space. Despite these headwinds, the recent opioid crisis has highlighted the devastating consequences of SUDs for both individuals and society, stimulating urgent efforts to identify novel treatment approaches. In addition, several neurobiological systems have been recently implicated in unique aspects of drug reward, opening the door to candidate medications with novel mechanisms of action. Here, we provide an overview of efforts to target several of these new systems, with a focus on those that are the subject of ongoing clinical trials as well as being areas of interest among the authors' research groups (MHJ, MTB, NAE). Specifically, we discuss new classes of medications targeting the serotonin 2A receptor (i.e., psychedelics), glucagon-like peptide 1 receptor, cannabidiol, dynorphin/kappa opioid receptor, orexin/hypocretin, and oxytocin receptor systems, as well as emergent approaches for modulating the more canonical dopaminergic system via agonist therapies for stimulant use disorders. Collectively, innovations in this space give reason for optimism for an improved therapeutic landscape for substance use disorders in the near future.
Collapse
Affiliation(s)
- Joel S Raymond
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA; Rutgers Addiction Research Center, Brain Health Institute, Rutgers Health, Piscataway, NJ, USA
| | - Alexander G Athanasopoulos
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Connie J Badolato
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Tylah J Doolan
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Rhianne L Scicluna
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Nicholas A Everett
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Michael T Bowen
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Morgan H James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA; Rutgers Addiction Research Center, Brain Health Institute, Rutgers Health, Piscataway, NJ, USA; School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
4
|
Aliev F, De Sa Nogueira D, Aston-Jones G, Dick DM. Genetic associations between orexin genes and phenotypes related to behavioral regulation in humans, including substance use. Mol Psychiatry 2025:10.1038/s41380-025-02895-4. [PMID: 39880903 DOI: 10.1038/s41380-025-02895-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/23/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025]
Abstract
The hypothalamic neuropeptide system of orexin (hypocretin) neurons provides projections throughout the neuraxis and has been linked to sleep regulation, feeding and motivation for salient rewards including drugs of abuse. However, relatively little has been done to examine genes associated with orexin signaling and specific behavioral phenotypes in humans. Here, we tested for association of twenty-seven genes involved in orexin signaling with behavioral phenotypes in humans. We tested the full gene set, functional subsets, and individual genes involved in orexin signaling. Our primary phenotype of interest was Externalizing, a composite factor comprised of behaviors and disorders associated with reward-seeking, motivation, and behavioral regulation. We also tested for association with additional phenotypes that have been related to orexin regulation in model organism studies, including alcohol consumption, problematic alcohol use, daytime sleepiness, insomnia, cigarettes per day, smoking initiation, and body mass index. The composite set of 27 genes corresponding to orexin function was highly associated with Externalizing, as well as with alcohol consumption, insomnia, cigarettes per day, smoking initiation and BMI. In addition, all gene subsets (except the OXR2/HCRTR2 subset) were associated with Externalizing. BMI was significantly associated with all gene subsets. The "validated factors for PPOX/HCRT" and "PPOX/HCRT upregulation" gene subsets also were associated with alcohol consumption. Individually, 8 genes showed a strong association with Externalizing, 12 with BMI, 7 with smoking initiation, 3 with alcohol consumption, and 2 with problematic alcohol use, after correction for multiple testing. This study indicates that orexin genes are associated with multiple behaviors and disorders related to self-regulation in humans. This is consistent with prior work in animals that implicated orexin signaling in motivational activation induced by salient stimuli, and supports the hypothesis that orexin signaling is an important potential therapeutic target for numerous behavioral disorders.
Collapse
Affiliation(s)
- Fazil Aliev
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - David De Sa Nogueira
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - Gary Aston-Jones
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - Danielle M Dick
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA.
| |
Collapse
|
5
|
Moreira-Junior EDC, Rowlett JK, Berro LF. Suvorexant enhances oxycodone-induced respiratory depression in male rats. Drug Alcohol Depend 2024; 264:112434. [PMID: 39255740 PMCID: PMC11527583 DOI: 10.1016/j.drugalcdep.2024.112434] [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: 06/21/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Recent studies have proposed the use of dual orexin receptor antagonists, such as suvorexant, for the treatment of opioid use disorder (OUD) and opioid-related sleep disturbances because of orexin's role in sleep-wake regulation and addiction. Accumulating evidence suggests that orexin is also an important modulator of respiratory function, raising the possibility of adverse respiratory events when combining orexin antagonists and opioids. The aim of the present study was to investigate the effects of suvorexant, alone or in combination with the opioid oxycodone, on pulmonary ventilation in male rats. METHODS Adult, male Sprague Dawley rats received treatments with vehicle, oxycodone (3 and 10mg/kg, i.p.) or suvorexant (10 and 18mg/kg, i.p.), and respiratory measures were obtained using whole-body plethysmography. We then tested the effects of a combination of suvorexant (10 and 18mg/kg, i.p.) and the highest dose of oxycodone that did not suppress respiration alone (3mg/kg, i.p). RESULTS Oxycodone induced respiratory depression at 10mg/kg, but not 3.0mg/kg; as evident by significant decreases in minute volume (mls/min) and tidal volume (mls). Suvorexant alone did not alter any respiratory measures at the doses tested. When combined, 18mg/kg (but not 10mg/kg) suvorexant plus an ineffective dose of oxycodone significantly decreased minute and tidal volume compared with vehicle and either drug alone, whereas respiratory frequency was significantly decreased compared with vehicle. CONCLUSIONS Our findings show that suvorexant, at a dose associated with sleep promotion and blockade of oxycodone self-administration, robustly enhanced oxycodone-induced respiratory depression in male rats.
Collapse
Affiliation(s)
- Eliseu D C Moreira-Junior
- Department of Psychiatry and Human Behavior, Center for Innovation and Discovery in Addictions, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - James K Rowlett
- Department of Psychiatry and Human Behavior, Center for Innovation and Discovery in Addictions, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Lais F Berro
- Department of Psychiatry and Human Behavior, Center for Innovation and Discovery in Addictions, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
6
|
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
|
7
|
Steiner MA, Botticelli L, Bergamini G, Micioni Di Bonaventura E, Gatfield J, Williams JT, Treiber A, Vaillant C, Cifani C, Micioni Di Bonaventura MV. Evaluating the efficacy of the selective orexin 1 receptor antagonist nivasorexant in an animal model of binge-eating disorder. Int J Eat Disord 2024; 57:1418-1432. [PMID: 38456603 DOI: 10.1002/eat.24181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/09/2024]
Abstract
OBJECTIVE Test the efficacy of the selective orexin 1 receptor (OX1R) antagonist (SO1RA) nivasorexant in an animal model of binge-eating disorder (BED) and study its dose-response relationship considering free brain concentrations and calculated OX1R occupancy. Compare nivasorexant's profile to that of other, structurally diverse SO1RAs. Gain understanding of potential changes in orexin-A (OXA) neuropeptide and deltaFosB (ΔFosB) protein expression possibly underlying the development of the binge-eating phenotype in the rat model used. METHOD Binge-like eating of highly palatable food (HPF) in rats was induced through priming by intermittent, repeated periods of dieting and access to HPF, followed by an additional challenge with acute stress. Effects of nivasorexant were compared to the SO1RAs ACT-335827 and IDOR-1104-2408. OXA expression in neurons and neuronal fibers as well as ΔFosB and OXA-ΔFosB co-expression was studied in relevant brain regions using immuno- or immunofluorescent histochemistry. RESULTS All SO1RAs dose-dependently reduced binge-like eating with effect sizes comparable to the positive control topiramate, at unbound drug concentrations selectively blocking brain OX1Rs. Nivasorexant's efficacy was maintained upon chronic dosing and under conditions involving more frequent stress exposure. Priming for binge-like eating or nivasorexant treatment resulted in only minor changes in OXA or ΔFosB expression in few brain areas. DISCUSSION Selective OX1R blockade reduced binge-like eating in rats. Neither ΔFosB nor OXA expression proved to be a useful classifier for their binge-eating phenotype. The current results formed the basis for a clinical phase II trial in BED, in which nivasorexant was unfortunately not efficacious compared with placebo. PUBLIC SIGNIFICANCE Nivasorexant is a new investigational drug for the treatment of binge-eating disorder (BED). It underwent clinical testing in a phase II proof of concept trial in humans but was not efficacious compared with placebo. The current manuscript investigated the drug's efficacy in reducing binge-like eating behavior of a highly palatable sweet and fat diet in a rat model of BED, which initially laid the foundation for the clinical trial.
Collapse
Affiliation(s)
| | - Luca Botticelli
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Giorgio Bergamini
- CNS Pharmacology, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | | | - John Gatfield
- CNS Pharmacology, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Jodi T Williams
- CNS Pharmacology, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Alexander Treiber
- CNS Pharmacology, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | | | - Carlo Cifani
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | | |
Collapse
|
8
|
White AM, Eglovitch M, Parlier-Ahmad AB, Dzierzewski JM, James M, Bjork JM, Moeller FG, Martin CE. Insomnia symptoms and neurofunctional correlates among adults receiving buprenorphine for opioid use disorder. PLoS One 2024; 19:e0304461. [PMID: 38870144 PMCID: PMC11175529 DOI: 10.1371/journal.pone.0304461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 05/14/2024] [Indexed: 06/15/2024] Open
Abstract
OBJECTIVES Insomnia symptoms are negatively related to opioid use disorder (OUD) treatment outcomes, possibly reflecting the influence of sleep on neurofunctional domains implicated in addiction. Moreover, the intersection between OUD recovery and sleep represents an area well-suited for the development of novel, personalized treatment strategies. This study assessed the prevalence of clinically significant insomnia symptoms and characterized its neurofunctional correlates among a clinical sample of adults with OUD receiving buprenorphine. METHODS Adults (N = 129) receiving buprenorphine for OUD from an outpatient clinic participated in a cross-sectional survey. Participants completed an abbreviated version of NIDA's Phenotyping Assessment Battery, which assessed 6 neurofunctional domains: sleep, negative emotionality, metacognition, interoception, cognition, and reward. Bivariate descriptive statistics compared those with evidence of clinically significant insomnia symptoms (Insomnia Severity Index [ISI] score of ≥11) to those with minimal evidence of clinically significant insomnia symptoms (ISI score of ≤10) across each of the neurofunctional domains. RESULTS Roughly 60% of participants reported clinically significant insomnia symptoms (ISI score of ≥11). Experiencing clinically significant insomnia symptoms was associated with reporting greater levels of depression, anxiety, post-traumatic stress, stress intolerance, unhelpful metacognition, and interoceptive awareness (ps<0.05). Participants with evidence of clinically significant insomnia were more likely to report that poor sleep was interfering with their OUD treatment and that improved sleep would assist with their treatment (ps<0.05). CONCLUSIONS Insomnia was prevalent among adults receiving buprenorphine for OUD. Insomnia was associated with neurofunctional performance, which may impact OUD treatment trajectories. Our findings indicate potential targets in the development of personalized treatment plans for patients with co-morbid insomnia and OUD. To inform the development of novel treatment strategies, more research is needed to understand the potential mechanistic links between sleep disturbances and substance use.
Collapse
Affiliation(s)
- Augustus M. White
- School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Michelle Eglovitch
- School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Anna Beth Parlier-Ahmad
- School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | | | - Morgan James
- Department of Psychiatry, Rutgers University, Newark, New Jersey, United States of America
| | - James M. Bjork
- School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - F. Gerard Moeller
- School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Caitlin E. Martin
- School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
9
|
Zhang VY, O’Connor SL, Welsh WJ, James MH. Machine learning models to predict ligand binding affinity for the orexin 1 receptor. ARTIFICIAL INTELLIGENCE CHEMISTRY 2024; 2:100040. [PMID: 38476266 PMCID: PMC10927255 DOI: 10.1016/j.aichem.2023.100040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
The orexin 1 receptor (OX1R) is a G-protein coupled receptor that regulates a variety of physiological processes through interactions with the neuropeptides orexin A and B. Selective OX1R antagonists exhibit therapeutic effects in preclinical models of several behavioral disorders, including drug seeking and overeating. However, currently there are no selective OX1R antagonists approved for clinical use, fueling demand for novel compounds that act at this target. In this study, we meticulously curated a dataset comprising over 1300 OX1R ligands using a stringent filter and criteria cascade. Subsequently, we developed highly predictive quantitative structure-activity relationship (QSAR) models employing the optimized hyper-parameters for the random forest machine learning algorithm and twelve 2D molecular descriptors selected by recursive feature elimination with a 5-fold cross-validation process. The predictive capacity of the QSAR model was further assessed using an external test set and enrichment study, confirming its high predictivity. The practical applicability of our final QSAR model was demonstrated through virtual screening of the DrugBank database. This revealed two FDA-approved drugs (isavuconazole and cabozantinib) as potential OX1R ligands, confirmed by radiolabeled OX1R binding assays. To our best knowledge, this study represents the first report of highly predictive QSAR models on a large comprehensive dataset of diverse OX1R ligands, which should prove useful for the discovery and design of new compounds targeting this receptor.
Collapse
Affiliation(s)
- Vanessa Y. Zhang
- 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
- West Windsor-Plainsboro High School South, West Windsor, 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
| | - William J. Welsh
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA
| | - 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
| |
Collapse
|
10
|
Shen L, Zeng X, Zhang H. The protective effects of orexin-A in alleviating cell senescence against interleukin-1β (IL-1β) in chondrocytes. Aging (Albany NY) 2024; 16:9558-9568. [PMID: 38829778 PMCID: PMC11210258 DOI: 10.18632/aging.205884] [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: 09/22/2023] [Accepted: 03/03/2024] [Indexed: 06/05/2024]
Abstract
Osteoarthritis (OA) is one of the most important causes of global disability, and dysfunction of chondrocytes is an important risk factor. The treatment of OA is still a challenge. Orexin-A is a hypothalamic peptide, and its effects in OA are unknown. In this study, we found that exposure to interleukin-1β (IL-1β) reduced the expression of orexin-2R, the receptor of orexin-A in TC-28a2 chondrocytes. Importantly, the senescence-associated β-galactosidase (SA-β-gal) staining assay demonstrated that orexin-A treatment ameliorates IL-1β-induced cellular senescence. Importantly, the presence of IL-1β significantly reduced the telomerase activity of TC-28a2 chondrocytes, which was rescued by orexin-A. We also found that orexin-A prevented IL-1β-induced increase in the levels of Acetyl-p53 and the expression of p21. It is shown that orexin-A mitigates IL-1β-induced reduction of sirtuin 3 (SIRT3). Silencing of SIRT3 abolished the protective effects of orexin-A against IL-1β-induced cellular senescence. These results imply that orexin-A might serve as a promising therapeutic agent for OA.
Collapse
Affiliation(s)
- Lin Shen
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, China
| | - Xiantie Zeng
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, China
| | - Haiying Zhang
- Department of Orthopedics, Dongfang Hospital, Beijing University of Traditional Chinese Medicine, Beijing 100078, China
| |
Collapse
|
11
|
Esmaili-Shahzade-Ali-Akbari P, Ghaderi A, Sadeghi A, Nejat F, Mehramiz A. The Role of Orexin Receptor Antagonists in Inhibiting Drug Addiction: A Review Article. ADDICTION & HEALTH 2024; 16:130-139. [PMID: 39051042 PMCID: PMC11264478 DOI: 10.34172/ahj.2024.1491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 04/15/2024] [Indexed: 07/27/2024]
Abstract
The orexinergic system and its receptors are involved in many physiological processes. Their functions in energy homeostasis, arousal, cognition, stress processing, endocrine functions, and pain modulation have been investigated. Many studies have shown that the orexinergic system cooperates with the dopaminergic system in the addiction process. Emerging evidence suggests that the orexinergic system can be effective in the induction of drug dependence and tolerance. Therefore, several researches have been conducted on the effect of orexin receptor (OXR) antagonists on reducing tolerance and dependence caused by drug abuse. Due to the significant growth of the studies on the orexinergic system, the current literature was conducted to collect the findings of previous studies on orexin and its receptors in the induction of drug addiction. In addition, cellular and molecular mechanisms of the possible role of orexin in drug tolerance and dependence are discussed. The findings indicate that the administration of OXR antagonists reduces drug dependence. OXR blockers seem to counteract the addictive effects of drugs through multiple mechanisms, such as preventing neuronal adaptation. This review proposes the potential clinical use of OXR antagonists in the treatment of drug dependence.
Collapse
Affiliation(s)
- Peyman Esmaili-Shahzade-Ali-Akbari
- Department of Addiction Studies, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Ghaderi
- Department of Addiction Studies, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Atena Sadeghi
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Fatemeh Nejat
- Department of Biology and Health Sciences, Meredith College, Raleigh, North Carolina, USA
| | - Alireza Mehramiz
- Department of Physical Therapy, Faculty of Paramedical and Rehabilitation Science, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
12
|
Hasler BP, Schulz CT, Pedersen SL. Sleep-Related Predictors of Risk for Alcohol Use and Related Problems in Adolescents and Young Adults. Alcohol Res 2024; 44:02. [PMID: 38500552 PMCID: PMC10948113 DOI: 10.35946/arcr.v44.1.02] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024] Open
Abstract
PURPOSE Growing evidence supports sleep and circadian rhythms as influencing alcohol use and the course of alcohol use disorder (AUD). Studying sleep/circadian-alcohol associations during adolescence and young adulthood may be valuable for identifying sleep/circadian-related approaches to preventing and/or treating AUD. This paper reviews current evidence for prospective associations between sleep/circadian factors and alcohol involvement during adolescence and young adulthood with an emphasis on the effects of sleep/circadian factors on alcohol use. SEARCH METHODS The authors conducted a literature search in PsycInfo, PubMed, and Web of Science using the search terms "sleep" and "alcohol" paired with "adolescent" or "adolescence" or "young adult" or "emerging adult," focusing on the title/abstract fields, and restricting to English-language articles. Next, the search was narrowed to articles with a prospective/longitudinal or experimental design, a sleep-related measure as a predictor, an alcohol-related measure as an outcome, and confirming a primarily adolescent and/or young adult sample. This step was completed by a joint review of candidate article abstracts by two of the authors. SEARCH RESULTS The initial search resulted in 720 articles. After review of the abstracts, the list was narrowed to 27 articles reporting on observational longitudinal studies and three articles reporting on intervention trials. Noted for potential inclusion were 35 additional articles that reported on studies with alcohol-related predictors and sleep-related outcomes, and/or reported on candidate moderators or mediators of sleep-alcohol associations. Additional articles were identified via review of relevant article reference lists and prior exposure based on the authors' previous work in this area. DISCUSSION AND CONCLUSIONS Overall, the review supports a range of sleep/circadian characteristics during adolescence and young adulthood predicting the development of alcohol use and/or alcohol-related problems. Although sleep treatment studies in adolescents and young adults engaging in regular and/or heavy drinking show that sleep can be improved in those individuals, as well as potentially reducing alcohol craving and alcohol-related consequences, no studies in any age group have yet demonstrated that improving sleep reduces drinking behavior. Notable limitations include relatively few longitudinal studies and only two experimental studies, insufficient consideration of different assessment timescales (e.g., day-to-day vs. years), insufficient consideration of the multidimensional nature of sleep, a paucity of objective measures of sleep and circadian rhythms, and insufficient consideration of how demographic variables may influence sleep/circadian-alcohol associations. Examining such moderators, particularly those related to minoritized identities, as well as further investigation of putative mechanistic pathways linking sleep/circadian characteristics to alcohol outcomes, are important next steps.
Collapse
Affiliation(s)
- Brant P Hasler
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Christina T Schulz
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sarah L Pedersen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
13
|
McGregor R, Wu MF, Thannickal TC, Siegel JM. Opiate anticipation, opiate induced anatomical changes in hypocretin (Hcrt, orexin) neurons and opiate induced microglial activation are blocked by the dual Hcrt receptor antagonist suvorexant, while opiate analgesia is maintained. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.22.559044. [PMID: 37790444 PMCID: PMC10542511 DOI: 10.1101/2023.09.22.559044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
We previously found that heroin addiction in humans is accompanied by an increase in the number of detected Hcrt neurons and a decrease in their soma size. We now show that the increased number of Hcrt cells visible after morphine treatment is likely the result of increased Hcrt production in neurons having sub-detection levels of the peptides. We find that morphine increases Hcrt projections to the ventral tegmental area (VTA), the level of tyrosine hydroxylase enzyme (TH) and the number of TH positive cells in VTA, with no changes in the adjacent substantia nigra. We find that the dual Hcrt receptor antagonist suvorexant prevents morphine-induced changes in the number and size of Hcrt neurons, microglial activation and morphine anticipatory behavior, but does not diminish morphine analgesia. These findings suggest that combined administration of opiates and suvorexant may be a less addictive way of administering opiates for pain relief in humans.
Collapse
|
14
|
Brotschi C, Bolli MH, Gatfield J, Roch C, Sifferlen T, Treiber A, Williams JT, Boss C. Pyrazole derivatives as selective orexin-2 receptor antagonists (2-SORA): synthesis, structure-activity-relationship, and sleep-promoting properties in rats. RSC Med Chem 2024; 15:344-354. [PMID: 38283232 PMCID: PMC10809354 DOI: 10.1039/d3md00573a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 11/20/2023] [Indexed: 01/30/2024] Open
Abstract
Selective orexin 2 receptor antagonists (2-SORA) such as seltorexant (15) are in clinical development for the treatment of insomnia and other conditions such as depression. Herein, we report our structure-activity-relationship (SAR) optimization efforts starting from an HTS hit (1) (N-(1-((5-acetylfuran-2-yl)methyl)-1H-pyrazol-4-yl)-5-(m-tolyl)oxazole-4-carboxamide) that was derived from an unrelated in-house GPCR-agonist program. Medicinal chemistry efforts focused on the optimization of orexin 2 receptor (OX2R) antagonistic activity, stability in liver microsomes, time dependent CYP3A4 inhibition, and aqueous solubility. Compounds were assessed for their brain-penetrating potential in in vivo experiments to select the most promising compounds for our in vivo sleep model. Our lead optimization efforts led to the discovery of the potent, brain penetrating and orally active, 2-SORA (N-(1-(2-(5-methoxy-1H-pyrrolo[3,2-b]pyridin-3-yl)ethyl)-1H-pyrazol-4-yl)-5-(m-tolyl)oxazole-4-carboxamide) 43 with efficacy in a sleep model in rats comparable to 15.
Collapse
Affiliation(s)
- Christine Brotschi
- Idorsia Pharmaceuticals Ltd, Drug Discovery and Preclinical Development Hegenheimermattweg 91 4123 Allschwil Basel-Landschaft Switzerland
| | - Martin H Bolli
- Idorsia Pharmaceuticals Ltd, Drug Discovery and Preclinical Development Hegenheimermattweg 91 4123 Allschwil Basel-Landschaft Switzerland
| | - John Gatfield
- Idorsia Pharmaceuticals Ltd, Drug Discovery and Preclinical Development Hegenheimermattweg 91 4123 Allschwil Basel-Landschaft Switzerland
| | - Catherine Roch
- Idorsia Pharmaceuticals Ltd, Drug Discovery and Preclinical Development Hegenheimermattweg 91 4123 Allschwil Basel-Landschaft Switzerland
| | - Thierry Sifferlen
- Idorsia Pharmaceuticals Ltd, Drug Discovery and Preclinical Development Hegenheimermattweg 91 4123 Allschwil Basel-Landschaft Switzerland
| | - Alexander Treiber
- Idorsia Pharmaceuticals Ltd, Drug Discovery and Preclinical Development Hegenheimermattweg 91 4123 Allschwil Basel-Landschaft Switzerland
| | - Jodi T Williams
- Idorsia Pharmaceuticals Ltd, Drug Discovery and Preclinical Development Hegenheimermattweg 91 4123 Allschwil Basel-Landschaft Switzerland
| | - Christoph Boss
- Idorsia Pharmaceuticals Ltd, Drug Discovery and Preclinical Development Hegenheimermattweg 91 4123 Allschwil Basel-Landschaft Switzerland
| |
Collapse
|
15
|
Knowles LG, Armanious AJ, Peng Y, Welsh WJ, James MH. Recent advances in drug discovery efforts targeting the sigma 1 receptor system: Implications for novel medications designed to reduce excessive drug and food seeking. ADDICTION NEUROSCIENCE 2023; 8:100126. [PMID: 37753198 PMCID: PMC10519676 DOI: 10.1016/j.addicn.2023.100126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Psychiatric disorders characterized by uncontrolled reward seeking, such as substance use disorders (SUDs), alcohol use disorder (AUD) and some eating disorders, impose a significant burden on individuals and society. Despite their high prevalence and substantial morbidity and mortality rates, treatment options for these disorders remain limited. Over the past two decades, there has been a gradual accumulation of evidence pointing to the sigma-1 receptor (S1R) system as a promising target for therapeutic interventions designed to treat these disorders. S1R is a chaperone protein that resides in the endoplasmic reticulum, but under certain conditions translocates to the plasma membrane. In the brain, S1Rs are expressed in several regions important for reward, and following translocation, they physically associate with several reward-related GPCRs, including dopamine receptors 1 and 2 (D1R and D2R). Psychostimulants, alcohol, as well as palatable foods, all alter expression of S1R in regions important for motivated behavior, and S1R antagonists generally decrease behavioral responses to these rewards. Recent advances in structural modeling have permitted the development of highly-selective S1R antagonists with favorable pharmacokinetic profiles, thus providing a therapeutic avenue for S1R-based medications. Here, we provide an up-to-date overview of work linking S1R with motivated behavior for drugs of abuse and food, as well as evidence supporting the clinical utility of S1R antagonists to reduce their excessive consumption. We also highlight potential challenges associated with targeting the S1R system, including the need for a more comprehensive understanding of the underlying neurobiology and careful consideration of the pharmacological properties of S1R-based drugs.
Collapse
Affiliation(s)
- Liam G. Knowles
- Harpur School of Arts and Sciences, Binghamton University, Vestal, NY, USA
| | - Abanoub J. Armanious
- 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
| | - Youyi Peng
- Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - William J. Welsh
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA
| | - 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
| |
Collapse
|
16
|
Elhadi K, Daiwile AP, Cadet JL. Modeling methamphetamine use disorder and relapse in animals: short- and long-term epigenetic, transcriptional., and biochemical consequences in the rat brain. Neurosci Biobehav Rev 2023; 155:105440. [PMID: 38707245 PMCID: PMC11068368 DOI: 10.1016/j.neubiorev.2023.105440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 05/07/2024]
Abstract
Methamphetamine use disorder (MUD) is a neuropsychiatric disorder characterized by binge drug taking episodes, intervals of abstinence, and relapses to drug use even during treatment. MUD has been modeled in rodents and investigators are attempting to identify its molecular bases. Preclinical experiments have shown that different schedules of methamphetamine self-administration can cause diverse transcriptional changes in the dorsal striatum of Sprague-Dawley rats. In the present review, we present data on differentially expressed genes (DEGs) identified in the rat striatum following methamphetamine intake. These include genes involved in transcription regulation, potassium channel function, and neuroinflammation. We then use the striatal data to discuss the potential significance of the molecular changes induced by methamphetamine by reviewing concordant or discordant data from the literature. This review identified potential molecular targets for pharmacological interventions. Nevertheless, there is a need for more research on methamphetamine-induced transcriptional consequences in various brain regions. These data should provide a more detailed neuroanatomical map of methamphetamine-induced changes and should better inform therapeutic interventions against MUD.
Collapse
Affiliation(s)
- Khalid Elhadi
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| | - Atul P. Daiwile
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| |
Collapse
|
17
|
Uğurlu M. Orexin Receptor Antagonists as Adjunct Drugs for the Treatment of Depression: A Mini Meta-Analysis. Noro Psikiyatr Ars 2023; 61:77-84. [PMID: 38496221 PMCID: PMC10943935 DOI: 10.29399/npa.28383] [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: 12/18/2022] [Accepted: 04/25/2023] [Indexed: 03/19/2024] Open
Abstract
Introduction There is growing interest in the efficacy of orexin receptor antagonists (ORA), one of the new psychopharmacological agents used in the treatment of insomnia, in other psychiatric disorders such as depression. Methods This meta-analysis was conducted in accordance with PRISMA requirements. Literature searches were conducted using PubMed, Scopus and EBSCO (Medline) databases. Search words were (depression OR mood disorder OR affective disorder) AND (orexin OR orx OR hypocretin OR orx1 OR orx2 OR orexin receptor antagonist OR almorexant OR suvorexant OR lemborexant OR daridorexant OR seltorexant OR vornorexant OR filorexant). No date restrictions were used. The random effects model was used for analyses with I2 values above 50% and fixed effects model was used for analyses with I2 values below 50%. Results In the acute phase, ORAs had no significant effect on core, sleep-adjusted and total symptoms of depression respectively; Standardized Mean Difference (SMD) for random effect -0.422, 95% CI [-0.90; 0.06], p=0.089, I2=62.4%; SMD for random effect -0.375, 95% CI [-1.24; 0.49], p=0.400; I2=66.6% and SMD for random effect -0.477, 95% CI [-0.97; 0.01], p=0.059; I2=83.1%). However, they had a significant effect on core and total symptoms of depression in the early period respectively; SMD for fixed effect=-0.228, 95% CI [-0.44; -0.01], p=0.036, I2=9.1%; and SMD for fixed effect=-0.186, 95% CI [-0.37; -0.001], p=0.048, I2=0.0%, respectively). Conclusion The results of this meta-analysis suggest that ORAs may provide direct antidepressant efficacy when added to existing antidepressant treatment and may also have indirect antidepressant effects through improvement in sleep symptoms. Considering the physiological effects of orexin on behaviors, ORAs may be promising new treatment modalities in the treatment of many psychiatric disorders other than insomnia. However, these results are preliminary and further studies with different ORAs at different doses and with different samples are needed.
Collapse
Affiliation(s)
- Mustafa Uğurlu
- Ankara Yıldırım Beyazıt University, Faculty of Medicine, Psychiatry Department, Ankara, Turkey
| |
Collapse
|
18
|
Zhou M, Li Y. Effect of different doses of almorexant on learning and memory in 8-month-old APP/PS1 (AD) mice. Peptides 2023; 167:171044. [PMID: 37330110 DOI: 10.1016/j.peptides.2023.171044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 06/19/2023]
Abstract
OBJECTIVE To explore the effects of different doses of almorexant (an dual orexin receptor antagonist) on learning and memory in Alzheimer's disease (AD) mice. METHODS Forty-four APP/PS1 (model of Alzheimer's disease; AD) mice were randomly divided into 4 groups: the control group (CON) and those that received 10mg/kg almorexant (low dose; LOW), 30mg/kg almorexant (medium dose; MED) and 60mg/kg almorexant (high dose; HIGH). During the 28-day intervention period, mice received an intraperitoneal injection at the beginning of the light period (6:00 am). The effects of different doses of almorexant on learning and memory and 24-hour sleep-wake behaviour were assessed by immunohistochemical staining. The above continuous variables are expressed as the mean ± standard deviation (SD), and then univariate regression analysis and generalized estimating equations were performed to compare the groups; these results are expressed as the mean difference (MD) and 95% confidence interval (CI). The statistical software used STATA 17.0 MP. RESULTS Forty-one mice completed the experiment (3 died: 2 mice in the HIGH group and 1 mouse in the CON group). Compared with the CON group, the LOW group (MD=6,803s, 95% CI: 4,470 to 9,137s), MED group (MD=14,473s, 95% CI: 12,140 to 16,806s) and the HIGH group (MD=24,505s, 95% CI: 22,052 to 26,959s) had significantly longer sleep durations. The Y maze results showed that LOW group (MD=0.14,95%CI: 0.078 to 0.20) and MED group (MD=0.14,95%CI = 0.074 to 0.20) mice compared to the CON group, and the low-medium dose of Almorexant did not damage the short-term learning and memory performance of APP / PS1 (AD) mice.Compared with the CON, LOW, and MED groups, the HIGH group exhibited a significant decrease in the Aβ plaque-positive area in the cortex (MD= -0.030, 95% CI: -0.035 to -0.025; MD=-0.049, 95% CI: -0.054 to -0.044; and MD=-0.07, 95% CI: -0.076 to -0.066, respectively). CONCLUSION The moderate dose of almorexant (30mg/kg) prolonged the sleep duration of APP/PS1 (AD) mice to a greater extent than the low dose (10mg/kg) without altering learning and memory. The MED mice showed a good sleep response and a small residual effect on the next day. High-dose (60mg / kg) almorexant impaired behavioral learning and memory performance in mice.Compared to the CON group and the LOW group, the MED group exhibited improved working memory. Thus, treatment with almorexant may reduce β-amyloid deposition in AD, slowing neurodegeneration. Additional studies are needed to determine the mechanism of action.
Collapse
Affiliation(s)
- Mengzhen Zhou
- Department of Neurology, Qianfo Mountain Hospital affiliated to Shandong First Medical University ,Jinan, Shandong, China.
| | - Yanran Li
- Department of Neurology, Qianfo Mountain Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
19
|
Yasmin N, Collier AD, Karatayev O, Abdulai AR, Yu B, Fam M, Khalizova N, Leibowitz SF. Subpopulations of hypocretin/orexin neurons differ in measures of their cell proliferation, dynorphin co-expression, projections, and response to embryonic ethanol exposure. Sci Rep 2023; 13:8448. [PMID: 37231149 PMCID: PMC10213024 DOI: 10.1038/s41598-023-35432-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/17/2023] [Indexed: 05/27/2023] Open
Abstract
Numerous studies in animals demonstrate that embryonic exposure to ethanol (EtOH) at low-moderate doses stimulates neurogenesis and increases the number of hypothalamic neurons expressing the peptide, hypocretin/orexin (Hcrt). A recent study in zebrafish showed that this effect on the Hcrt neurons in the anterior hypothalamus (AH) is area specific, evident in the anterior (aAH) but not posterior (pAH) part of this region. To understand specific factors that may determine the differential sensitivity to EtOH of these Hcrt subpopulations, we performed additional measures in zebrafish of their cell proliferation, co-expression of the opioid dynorphin (Dyn), and neuronal projections. In association with the increase in Hcrt neurons in the aAH but not pAH, EtOH significantly increased only in the aAH the proliferation of Hcrt neurons and their number lacking Dyn co-expression. The projections of these subpopulations differed markedly in their directionality, with those from the pAH primarily descending to the locus coeruleus and those from the aAH ascending to the subpallium, and they were both stimulated by EtOH, which induced specifically the most anterior subpallium-projecting Hcrt neurons to become ectopically expressed beyond the aAH. These differences between the Hcrt subpopulations suggest they are functionally distinct in their regulation of behavior.
Collapse
Affiliation(s)
- Nushrat Yasmin
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Adam D Collier
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Olga Karatayev
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Abdul R Abdulai
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Boyi Yu
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Milisia Fam
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Nailya Khalizova
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Sarah F Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
20
|
Yasmin N, Collier AD, Abdulai AR, Karatayev O, Yu B, Fam M, Leibowitz SF. Role of Chemokine Cxcl12a in Mediating the Stimulatory Effects of Ethanol on Embryonic Development of Subpopulations of Hypocretin/Orexin Neurons and Their Projections. Cells 2023; 12:1399. [PMID: 37408233 PMCID: PMC10216682 DOI: 10.3390/cells12101399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 07/07/2023] Open
Abstract
Studies in zebrafish and rats show that embryonic ethanol exposure at low-moderate concentrations stimulates hypothalamic neurons expressing hypocretin/orexin (Hcrt) that promote alcohol consumption, effects possibly involving the chemokine Cxcl12 and its receptor Cxcr4. Our recent studies in zebrafish of Hcrt neurons in the anterior hypothalamus (AH) demonstrate that ethanol exposure has anatomically specific effects on Hcrt subpopulations, increasing their number in the anterior AH (aAH) but not posterior AH (pAH), and causes the most anterior aAH neurons to become ectopically expressed further anterior in the preoptic area (POA). Using tools of genetic overexpression and knockdown, our goal here was to determine whether Cxcl12a has an important function in mediating the specific effects of ethanol on these Hcrt subpopulations and their projections. The results demonstrate that the overexpression of Cxcl12a has stimulatory effects similar to ethanol on the number of aAH and ectopic POA Hcrt neurons and the long anterior projections from ectopic POA neurons and posterior projections from pAH neurons. They also demonstrate that knockdown of Cxcl12a blocks these effects of ethanol on the Hcrt subpopulations and projections, providing evidence supporting a direct role of this specific chemokine in mediating ethanol's stimulatory effects on embryonic development of the Hcrt system.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
21
|
Leino TO, Turku A, Urvas L, Adhikari K, Oksanen J, Steynen Y, Yli-Kauhaluoma J, Xhaard H, Kukkonen JP, Wallén EAA. Azulene as a biphenyl mimetic in orexin/hypocretin receptor agonists. Bioorg Med Chem 2023; 88-89:117325. [PMID: 37209639 DOI: 10.1016/j.bmc.2023.117325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/22/2023]
Abstract
Azulene is a rare ring structure in drugs, and we investigated whether it could be used as a biphenyl mimetic in known orexin receptor agonist Nag 26, which is binding to both orexin receptors OX1 and OX2 with preference towards OX2. The most potent azulene-based compound was identified as an OX1 orexin receptor agonist (pEC50 = 5.79 ± 0.07, maximum response = 81 ± 8% (s.e.m. of five independent experiments) of the maximum response to orexin-A in Ca2+ elevation assay). However, the azulene ring and the biphenyl scaffold are not identical in their spatial shape and electron distribution, and their derivatives may adopt different binding modes in the binding site.
Collapse
Affiliation(s)
- Teppo O Leino
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland; Department of Chemistry and NanoScience Center, University of Jyväskylä, P.O. Box 35, FI-40014 University of Jyväskylä, Finland.
| | - Ainoleena Turku
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland; Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 66, FI-00014 University of Helsinki, Finland
| | - Lauri Urvas
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland; Department of Pharmacology, Faculty of Medicine, University of Helsinki, P.O. Box 63, FI-00014 University of Helsinki, Finland
| | - Karuna Adhikari
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Jouni Oksanen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Yana Steynen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Henri Xhaard
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Jyrki P Kukkonen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 66, FI-00014 University of Helsinki, Finland; Department of Pharmacology, Faculty of Medicine, University of Helsinki, P.O. Box 63, FI-00014 University of Helsinki, Finland
| | - Erik A A Wallén
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| |
Collapse
|
22
|
Malik JA, Agrewala JN. Future perspectives of emerging novel drug targets and immunotherapies to control drug addiction. Int Immunopharmacol 2023; 119:110210. [PMID: 37099943 DOI: 10.1016/j.intimp.2023.110210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/14/2023] [Accepted: 04/15/2023] [Indexed: 04/28/2023]
Abstract
Substance Use Disorder (SUD) is one of the major mental illnesses that is terrifically intensifying worldwide. It is becoming overwhelming due to limited options for treatment. The complexity of addiction disorders is the main impediment to understanding the pathophysiology of the illness. Hence, unveiling the complexity of the brain through basic research, identification of novel signaling pathways, the discovery of new drug targets, and advancement in cutting-edge technologies will help control this disorder. Additionally, there is a great hope of controlling the SUDs through immunotherapeutic measures like therapeutic antibodies and vaccines. Vaccines have played a cardinal role in eliminating many diseases like polio, measles, and smallpox. Further, vaccines have controlled many diseases like cholera, dengue, diphtheria, Haemophilus influenza type b (Hib), human papillomavirus, influenza, Japanese encephalitis, etc. Recently, COVID-19 was controlled in many countries by vaccination. Currently, continuous effort is done to develop vaccines against nicotine, cocaine, morphine, methamphetamine, and heroin. Antibody therapy against SUDs is another important area where serious attention is required. Antibodies have contributed substantially against many serious diseases like diphtheria, rabies, Crohn's disease, asthma, rheumatoid arthritis, and bladder cancer. Antibody therapy is gaining immense momentum due to its success rate in cancer treatment. Furthermore, enormous advancement has been made in antibody therapy due to the generation of high-efficiency humanized antibodies with a long half-life. The advantage of antibody therapy is its instant outcome. This article's main highlight is discussing the drug targets of SUDs and their associated mechanisms. Importantly, we have also discussed the scope of prophylactic measures to eliminate drug dependence.
Collapse
Affiliation(s)
- Jonaid Ahmad Malik
- Immunology laboratory, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Javed N Agrewala
- Immunology laboratory, Indian Institute of Technology Ropar, Rupnagar, Punjab, India.
| |
Collapse
|
23
|
Ye H, Cao T, Shu Q, Chen Y, Lu Y, He Z, Li Z. Blockade of orexin receptor 1 attenuates morphine protracted abstinence-induced anxiety-like behaviors in male mice. Psychoneuroendocrinology 2023; 151:106080. [PMID: 36931057 DOI: 10.1016/j.psyneuen.2023.106080] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/18/2023] [Accepted: 03/07/2023] [Indexed: 03/19/2023]
Abstract
One negative emotional state from morphine protracted abstinence is anxiety which can drive craving and relapse risk in opioid addicts. Although the orexinergic system has been reported to be important in mediating emotion processing and addiction, the role of orexinergic system in anxiety from drug protracted abstinence remains elusive. In this study, by using behavioral test, western blot, electrophysiology and virus-mediated regulation of orexin receptor 1 (OX1R), we found that: (1) Intraperitoneal and intra-VTA administration of a selective OX1R antagonist SB334867 alleviated anxiety-like behaviors in open field test (OFT) but not in elevated plus maze test (EPM) in morphine protracted abstinent male mice. (2) OX1R expression in the VTA was upregulated by morphine withdrawal. (3) Virus-mediated knockdown of OX1R in the VTA prevented morphine abstinence-induced anxiety-like behaviors and virus-mediated overexpression of OX1R in the VTA was sufficient to produce anxiety-like behaviors in male mice. (4) The VTA neuronal activity was increased significantly induced by morphine protracted abstinence, which was mediated by OX1R. (5) OX1R was widely distributed in the neuronal soma and processes of dopaminergic and non-dopaminergic neurons in the VTA. The findings revealed that the OX1R mediates morphine abstinence-induced anxiety-like behaviors and the VTA plays a critical role in this effect.
Collapse
Affiliation(s)
- Hongming Ye
- College of Basic Medical Science, China Three Gorges University, Yichang, China; Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, China
| | - Tong Cao
- College of Basic Medical Science, China Three Gorges University, Yichang, China; Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, China
| | - Qigang Shu
- College of Basic Medical Science, China Three Gorges University, Yichang, China; Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, China
| | - Yue Chen
- College of Basic Medical Science, China Three Gorges University, Yichang, China; Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, China
| | - Yongli Lu
- College of Basic Medical Science, China Three Gorges University, Yichang, China; Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, China
| | - Zhi He
- College of Basic Medical Science, China Three Gorges University, Yichang, China; Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, China.
| | - Zicheng Li
- College of Basic Medical Science, China Three Gorges University, Yichang, China; Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, China.
| |
Collapse
|
24
|
Okuda S, Qureshi ZP, Yanagida Y, Ito C, Homma Y, Tokita S. Factors Associated with Prescriptions for an Orexin Receptor Antagonist Among Japanese Patients with Insomnia: Analysis of a Nationwide Japanese Claims Database. Drugs Real World Outcomes 2023:10.1007/s40801-023-00356-4. [PMID: 36867350 PMCID: PMC9982805 DOI: 10.1007/s40801-023-00356-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Few studies have examined the prescribing patterns of orexin receptor antagonists (ORAs) in the real-world clinical setting in Japan. OBJECTIVE We sought to analyze the factors associated with ORA prescriptions for patients with insomnia in Japan. METHODS Outpatients (aged ≥ 20 to < 75 years old) prescribed one or more hypnotic for insomnia between April 1, 2018 and March 31, 2020 with continuous enrollment for ≥ 12 months were extracted from the JMDC Claims Database. We performed multivariable logistic regression to identify factors (patient demographics and psychiatric comorbidities) associated with ORA prescription in new or non-new users of hypnotics (patients without or with hypnotics prescription history, respectively). RESULTS Of 58,907 new users, 11,589 (19.7%) were prescribed ORA at the index date. Male sex (odds ratio [OR] 1.17, 95% confidence interval [CI] 1.12-1.22) and presence of bipolar disorders (OR 1.36, 95% CI 1.20-1.55) were associated with greater odds of ORA prescription. Among 88,611 non-new users, 15,504 (17.5%) were prescribed ORA at the index date. Younger age and several psychiatric comorbidities, such as neurocognitive disorders (OR 1.64, 95% CI 1.15-2.35), substance use disorders (OR 1.19, 95% CI 1.05-1.35), bipolar disorders (OR 1.14, 95% CI 1.07-1.22), schizophrenia spectrum disorders (OR 1.07, 95% CI 1.01-1.14), and anxiety disorders (OR 1.05, 95% CI 1.00-1.10), were associated with greater odds of ORA prescription. CONCLUSION This is the first study to determine the factors associated with ORA prescriptions in Japan. Our findings could help guide appropriate insomnia treatment using ORAs.
Collapse
Affiliation(s)
| | - Zaina P. Qureshi
- Center for Observational and Real-world Evidence (CORE), Merck & Co., Inc., Rahway, NJ USA
| | | | | | | | | |
Collapse
|
25
|
Maness EB, Blumenthal SA, Burk JA. Dual orexin/hypocretin receptor antagonism attenuates attentional impairments in an NMDA receptor hypofunction model of schizophrenia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.05.527043. [PMID: 36778441 PMCID: PMC9915718 DOI: 10.1101/2023.02.05.527043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a neuropsychiatric condition that is associated with impaired attentional processing and performance. Failure to support increasing attentional load may result, in part, from abnormally overactive basal forebrain projections to the prefrontal cortex, and available antipsychotics often fail to address this issue. Orexin/hypocretin receptors are expressed on corticopetal cholinergic neurons, and their blockade has been shown to decrease the activity of cortical basal forebrain outputs and prefrontal cortical cholinergic neurotransmission. In the present experiment, rats (N = 14) trained in a visual sustained attention task that required discrimination of trials which presented a visual signal from trials during which no signal was presented. Once trained, rats were then co-administered the psychotomimetic N-methyl-D-aspartate (NMDA) receptor antagonist dizocilpine (MK-801: 0 or 0.1 mg/kg, intraperitoneal injections) and the dual orexin receptor antagonist filorexant (MK-6096: 0, 0.1, or 1 mM, intracerebroventricular infusions) prior to task performance across six sessions. Dizocilpine impaired overall accuracy during signal trials, slowed reaction times for correctly-responded trials, and increased the number of omitted trials throughout the task. Dizocilpine-induced increases in signal trial deficits, correct response latencies, and errors of omission were reduced following infusions of the 0.1 mM, but not 1 mM, dose of filorexant. Orexin receptor blockade, perhaps through anticholinergic mechanisms, may improve attentional deficits in a state of NMDA receptor hypofunction. Highlights Schizophrenia is associated with attentional deficits that may stem from abnormally reactive BF projections to the prefrontal cortexOrexin receptor antagonists decrease acetylcholine release and reduce prefrontal cortical activityThe dual orexin receptor antagonist filorexant alleviated impairments of attention following NMDA receptor blockade.
Collapse
Affiliation(s)
- Eden B. Maness
- VA Boston Healthcare System and Department of Psychiatry, Harvard Medical School, West Roxbury, MA, 02132, USA
- Department of Psychological Sciences, College of William and Mary, Williamsburg, VA, 23187, USA
| | - Sarah A. Blumenthal
- Center for Translational Social Neuroscience, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Joshua A. Burk
- Department of Psychological Sciences, College of William and Mary, Williamsburg, VA, 23187, USA
| |
Collapse
|
26
|
Veisi A, Khaleghzadeh-Ahangar H, Fattahi M, Haghparast A. The Role of Orexin-1 Receptors Within the Hippocampal CA1 Area in the Extinction and Reinstatement of Methamphetamine-Seeking Behaviors. Neurochem Res 2023; 48:671-680. [PMID: 36284025 DOI: 10.1007/s11064-022-03793-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 02/04/2023]
Abstract
Psychostimulant addiction is a chronic brain disorder with high relapse rates, requiring new therapeutic strategies. The orexin system is highly implicated in processing reward and addiction through connections with critical areas such as the hippocampus. This study investigated the role of orexin-1 receptors (OX1R) within the CA1 subregion of the hippocampus in the extinction and reinstatement of the methamphetamine-induced conditioned place preference. After cannulae implantation, recovery, and establishing the methamphetamine place preference, 98 male Wistar rats received different doses of bilateral intra-CA1 selective OX1R antagonist, SB334867 (1, 3, 10, and 30 nmol/0.5 μl DMSO per side) during the 10-day extinction period (daily) or after extinction phase, just on the reinstatement day (single dose) in separate experimental and control groups. The findings indicated that bilateral microinjection of SB334867 into the CA1 area during the extinction period could significantly reduce the extinction latency and maintenance of rewarding aspects of methamphetamine dose-dependently (3, 10, and 30 nmol). In another set of experiments, a single dose of bilateral intra-CA1 SB334867 administration on the reinstatement phase prevented the methamphetamine-induced reinstatement of drug-seeking behaviors at the high doses (10, and 30 nmol). The present study provided more evidence for the implication of hippocampal OX1R in the maintenance of rewarding and reinforcing properties of methamphetamine and its role in the relapse of methamphetamine-seeking behavior. Further investigations on the role of the orexin system, including the orexin-2 receptors in treating addiction, are needed to introduce its antagonists as effective therapeutic options for psychostimulant addiction.
Collapse
Affiliation(s)
- Ali Veisi
- Department of Physiology, Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Hossein Khaleghzadeh-Ahangar
- Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
- Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mojdeh Fattahi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O.Box 19615-1178, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O.Box 19615-1178, Tehran, Iran.
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran.
- Department of Basic Sciences, Iranian Academy of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Collier AD, Yasmin N, Karatayev O, Abdulai AR, Yu B, Khalizova N, Fam M, Leibowitz SF. Neuronal chemokine concentration gradients mediate effects of embryonic ethanol exposure on ectopic hypocretin/orexin neurons and behavior in zebrafish. Sci Rep 2023; 13:1447. [PMID: 36702854 PMCID: PMC9880007 DOI: 10.1038/s41598-023-28369-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
Embryonic ethanol exposure in zebrafish and rats, while stimulating hypothalamic hypocretin/orexin (Hcrt) neurons along with alcohol consumption and related behaviors, increases the chemokine receptor Cxcr4 that promotes neuronal migration and may mediate ethanol's effects on neuronal development. Here we performed a more detailed anatomical analysis in zebrafish of ethanol's effects on the Cxcl12a/Cxcr4b system throughout the entire brain as it relates to Hcrt neurons developing within the anterior hypothalamus (AH) where they are normally located. We found that ethanol increased these Hcrt neurons only in the anterior part of the AH and induced ectopic Hcrt neurons further anterior in the preoptic area, and these effects along with ethanol-induced behaviors were completely blocked by a Cxcr4 antagonist. Analysis of cxcl12a transcripts and internalized Cxcr4b receptors throughout the brain showed they both exhibited natural posterior-to-anterior concentration gradients, with levels lowest in the posterior AH and highest in the anterior telencephalon. While stimulating their density in all areas and maintaining these gradients, ethanol increased chemokine expression only in the more anterior and ectopic Hcrt neurons, effects blocked by the Cxcr4 antagonist. These findings demonstrate how increased chemokine expression acting along natural gradients mediates ethanol-induced anterior migration of ectopic Hcrt neurons and behavioral disturbances.
Collapse
Affiliation(s)
- Adam D Collier
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Nushrat Yasmin
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Olga Karatayev
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Abdul R Abdulai
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Boyi Yu
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Nailya Khalizova
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Milisia Fam
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Sarah F Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
28
|
Mogavero MP, Silvani A, Lanza G, DelRosso LM, Ferini-Strambi L, Ferri R. Targeting Orexin Receptors for the Treatment of Insomnia: From Physiological Mechanisms to Current Clinical Evidence and Recommendations. Nat Sci Sleep 2023; 15:17-38. [PMID: 36713640 PMCID: PMC9879039 DOI: 10.2147/nss.s201994] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/08/2023] [Indexed: 01/23/2023] Open
Abstract
After a detailed description of orexins and their roles in sleep and other medical disorders, we discuss here the current clinical evidence on the effects of dual (DORAs) or selective (SORAs) orexin receptor antagonists on insomnia with the aim to provide recommendations for their further assessment in a context of personalized and precision medicine. In the last decade, many trials have been conducted with orexin receptor antagonists, which represent an innovative and valid therapeutic option based on the multiple mechanisms of action of orexins on different biological circuits, both centrally and peripherally, and their role in a wide range of medical conditions which are often associated with insomnia. A very interesting aspect of this new category of drugs is that they have limited abuse liability and their discontinuation does not seem associated with significant rebound effects. Further studies on the efficacy of DORAs are required, especially on children and adolescents and in particular conditions, such as menopause. Which DORA is most suitable for each patient, based on comorbidities and/or concomitant treatments, should be the focus of further careful research. On the contrary, studies on SORAs, some of which seem to be appropriate also in insomnia in patients with psychiatric diseases, are still at an early stage and, therefore, do not allow to draw definite conclusions.
Collapse
Affiliation(s)
- Maria P Mogavero
- Vita-Salute San Raffaele University, Milan, Italy
- Sleep Disorders Center, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Silvani
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe Lanza
- Sleep Research Centre, Oasi Research Institute - IRCCS, Troina, Italy
- Department of Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Lourdes M DelRosso
- Pulmonary and Sleep Medicine, University of California San Francisco-Fresno, Fresno, CA, USA
| | - Luigi Ferini-Strambi
- Vita-Salute San Raffaele University, Milan, Italy
- Sleep Disorders Center, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Raffaele Ferri
- Sleep Research Centre, Oasi Research Institute - IRCCS, Troina, Italy
| |
Collapse
|
29
|
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
|
30
|
Guo R, Vaughan DT, Rojo ALA, Huang YH. Sleep-mediated regulation of reward circuits: implications in substance use disorders. Neuropsychopharmacology 2023; 48:61-78. [PMID: 35710601 PMCID: PMC9700806 DOI: 10.1038/s41386-022-01356-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 12/11/2022]
Abstract
Our modern society suffers from both pervasive sleep loss and substance abuse-what may be the indications for sleep on substance use disorders (SUDs), and could sleep contribute to the individual variations in SUDs? Decades of research in sleep as well as in motivated behaviors have laid the foundation for us to begin to answer these questions. This review is intended to critically summarize the circuit, cellular, and molecular mechanisms by which sleep influences reward function, and to reveal critical challenges for future studies. The review also suggests that improving sleep quality may serve as complementary therapeutics for treating SUDs, and that formulating sleep metrics may be useful for predicting individual susceptibility to SUDs and other reward-associated psychiatric diseases.
Collapse
Affiliation(s)
- Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Allen Institute, Seattle, WA, 98109, USA
| | - Dylan Thomas Vaughan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- The Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Ana Lourdes Almeida Rojo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- The Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Yanhua H Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
- The Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
31
|
Flores-Ramirez FJ, Illenberger JM, Pascasio GE, Matzeu A, Mason BJ, Martin-Fardon R. Alternative use of suvorexant (Belsomra ®) for the prevention of alcohol drinking and seeking in rats with a history of alcohol dependence. Front Behav Neurosci 2022; 16:1085882. [PMID: 36620860 PMCID: PMC9813433 DOI: 10.3389/fnbeh.2022.1085882] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Alcohol use disorder (AUD) is one of the most treatment-resistant medical conditions globally. The orexin (Orx) system regulates diverse physiological processes, including stress, and is a system of interest for the development of pharmaceuticals to treat substance use disorders, particularly AUD. The present study tested the ability of the dual orexin receptor antagonist suvorexant (SUV), marketed by Merck as Belsomra®, for the treatment of insomnia, to decrease alcohol self-administration and the stress-induced reinstatement of alcohol-seeking behavior in male Wistar rats with a history of alcohol dependence. Rats were trained to orally self-administer 10% alcohol (30 min/day for 3 weeks) and were either made dependent via chronic intermittent alcohol vapor exposure (14 h ON, 10 h OFF) for 6 weeks or exposed to air (non-dependent). Starting on week 7, the effect of SUV (0-20 mg/kg, p.o.) was tested on alcohol self-administration at acute abstinence (8 h after vapor was turned OFF) twice weekly. A separate cohort of rats that were prepared in parallel was removed from alcohol vapor exposure and then subjected to extinction training for 14 sessions. Once extinction was achieved, the rats received SUV (0 and 5 mg/kg, p.o.) and were tested for the footshock stress-induced reinstatement of alcohol-seeking behavior. Suvorexant at 5, 10, and 20 mg/kg selectively decreased alcohol intake in dependent rats. Furthermore, 5 mg/kg SUV prevented the stress-induced reinstatement of alcohol-seeking behavior in dependent rats only. These results underscore the significance of targeting the Orx system for the treatment of substance use disorders generally and suggest that repurposing SUV could be an alternative approach for the treatment of AUD.
Collapse
|
32
|
Collier AD, Yasmin N, Chang GQ, Karatayev O, Khalizova N, Fam M, Abdulai AR, Yu B, Leibowitz SF. Embryonic ethanol exposure induces ectopic Hcrt and MCH neurons outside hypothalamus in rats and zebrafish: Role in ethanol-induced behavioural disturbances. Addict Biol 2022; 27:e13238. [PMID: 36301208 PMCID: PMC9625080 DOI: 10.1111/adb.13238] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/02/2022] [Accepted: 09/22/2022] [Indexed: 01/24/2023]
Abstract
Embryonic exposure to ethanol increases the risk for alcohol use disorder in humans and stimulates alcohol-related behaviours in different animal models. Evidence in rats and zebrafish suggests that this phenomenon induced by ethanol at low-moderate concentrations involves a stimulatory effect on neurogenesis and density of hypothalamic neurons expressing the peptides, hypocretin/orexin (Hcrt) and melanin-concentrating hormone (MCH), known to promote alcohol consumption. Building on our report in zebrafish showing that ethanol induces ectopic expression of Hcrt neurons outside the hypothalamus, we investigated here whether embryonic ethanol exposure also induces ectopic peptide neurons in rats similar to zebrafish and affects their morphological characteristics and if these ectopic neurons are functional and have a role in the ethanol-induced disturbances in behaviour. We demonstrate in rats that ethanol at a low-moderate dose, in addition to increasing Hcrt and MCH neurons in the lateral hypothalamus where they are normally concentrated, induces ectopic expression of these peptide neurons further anterior in the nucleus accumbens core and ventromedial caudate putamen where they have not been previously observed and causes morphological changes relative to normally located hypothalamic neurons. Similar to rats, embryonic ethanol exposure at a low-moderate dose in zebrafish induces ectopic Hcrt neurons anterior to the hypothalamus and alters their morphology. Notably, laser ablation of these ectopic Hcrt neurons blocks the behavioural effects induced by ethanol exposure, including increased anxiety and locomotor activity. These findings suggest that the ectopic peptide neurons are functional and contribute to the ethanol-induced behavioural disturbances related to the overconsumption of alcohol.
Collapse
Affiliation(s)
- Adam D. Collier
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065
| | - Nushrat Yasmin
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065
| | - Guo-Qing Chang
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065
| | - Olga Karatayev
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065
| | - Nailya Khalizova
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065
| | - Milisia Fam
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065
| | - Abdul R. Abdulai
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065
| | - Boyi Yu
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065
| | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065
| |
Collapse
|
33
|
Giannotti G, Mottarlini F, Heinsbroek JA, Mandel MR, James MH, Peters J. Oxytocin and orexin systems bidirectionally regulate the ability of opioid cues to bias reward seeking. Transl Psychiatry 2022; 12:432. [PMID: 36195606 PMCID: PMC9532415 DOI: 10.1038/s41398-022-02161-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
As opioid-related fatalities continue to rise, the need for novel opioid use disorder (OUD) treatments could not be more urgent. Two separate hypothalamic neuropeptide systems have shown promise in preclinical OUD models. The oxytocin system, originating in the paraventricular nucleus (PVN), may protect against OUD severity. By contrast, the orexin system, originating in the lateral hypothalamus (LH), may exacerbate OUD severity. Thus, activating the oxytocin system or inhibiting the orexin system are potential therapeutic strategies. The specific role of these systems with regard to specific OUD outcomes, however, is not fully understood. Here, we probed the therapeutic efficacy of pharmacological interventions targeting the orexin or oxytocin system on two distinct metrics of OUD severity in rats-heroin choice (versus choice for natural reward, i.e., food) and cued reward seeking. Using a preclinical model that generates approximately equal choice between heroin and food reward, we examined the impact of exogenously administered oxytocin, an oxytocin receptor antagonist (L-368,899), and a dual orexin receptor antagonist (DORA-12) on opioid choice. Whereas these agents did not alter heroin choice when rewards (heroin and food) were available, oxytocin and DORA-12 each significantly reduced heroin seeking in the presence of competing reward cues when no rewards were available. In addition, the number of LH orexin neurons and PVN oxytocin neurons correlated with specific behavioral economic variables indicative of heroin versus food motivation. These data identify a novel bidirectional role of the oxytocin and orexin systems in the ability of opioid-related cues to bias reward seeking.
Collapse
Affiliation(s)
- Giuseppe Giannotti
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Francesca Mottarlini
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133, Milan, Italy
| | - Jasper A Heinsbroek
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Mitchel R Mandel
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Morgan H James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA
| | - Jamie Peters
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
34
|
Kourosh-Arami M, Gholami M, Alavi-Kakhki SS, Komaki A. Neural correlates and potential targets for the contribution of orexin to addiction in cortical and subcortical areas. Neuropeptides 2022; 95:102259. [PMID: 35714437 DOI: 10.1016/j.npep.2022.102259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 02/01/2023]
Abstract
The orexin (hypocretin) is one of the hypothalamic neuropeptides that plays a critical role in some behaviors including feeding, sleep, arousal, reward processing, and drug addiction. This variety of functions can be described by a united function for orexins in translating states of heightened motivation, for example during physiological requirement states or following exposure to reward opportunities, into planned goal-directed behaviors. An addicted state is characterized by robust activation of orexin neurons from the environment, which triggers downstream circuits to facilitate behavior directed towards obtaining the drug. Two orexin receptors 1 (OX1R) and 2 (OX2R) are widely distributed in the brain. Here, we will introduce and describe the cortical and subcortical brain areas involved in addictive-like behaviors and the impact of orexin on addiction.
Collapse
Affiliation(s)
- Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Gholami
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran.
| | - Seyed Sajjad Alavi-Kakhki
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
35
|
Stoops WW, Strickland JC, Hatton KW, Hays LR, Rayapati AO, Lile JA, Rush CR. Suvorexant maintenance enhances the reinforcing but not subjective and physiological effects of intravenous cocaine in humans. Pharmacol Biochem Behav 2022; 220:173466. [PMID: 36152876 PMCID: PMC9588557 DOI: 10.1016/j.pbb.2022.173466] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Preclinical research has sought to understand the role of the orexin system in cocaine addiction given the connection between orexin producing cells in the lateral hypothalamus and brain limbic areas. Exogenous administration of orexin peptides increased cocaine self-administration whereas selective orexin-1 receptor antagonists reduced cocaine self-administration in non-human animals. The first clinically available orexin antagonist, suvorexant (a dual orexin-1 and orexin-2 receptor antagonist), attenuated motivation for cocaine and cocaine conditioned place preference, as well as cocaine-associated impulsive responding, in rodents. This study aimed to translate those preclinical findings and determine whether suvorexant maintenance altered the pharmacodynamic effects of cocaine in humans. Seven non-treatment seeking subjects with cocaine use disorder completed this within-subject human laboratory study, and a partial data set was obtained from one additional subject. Subjects were maintained for at least three days on 0, 5, 10 and 20 mg oral suvorexant administered at 2230 h daily in random order. Subjects completed experimental sessions in which cocaine self-administration of 0, 10 and 30 mg/70 kg of intravenous cocaine was evaluated on a concurrent progressive ratio drug versus money choice task. Subjective and physiological effects of cocaine were also determined. Cocaine functioned as a reinforcer and produced prototypic dose-related subjective and physiological effects (e.g., increased ratings of "Stimulated" and heart rate). Suvorexant (10, 20 mg) increased self-administration of 10 mg/70 kg cocaine and decreased oral temperature but did not significantly alter any other effects of cocaine. Future research may seek to evaluate the effects of orexin-1 selective antagonists in combination with cocaine.
Collapse
Affiliation(s)
- William W Stoops
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA; Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA; Department of Psychology, University of Kentucky College of Arts and Sciences, 171 Funkhouser Drive, Lexington, KY 40506-0044, USA; Center on Drug and Alcohol Research, University of Kentucky College of Medicine, 845 Angliana Avenue, Lexington, KY 40508, USA.
| | - Justin C Strickland
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD 21224, USA
| | - Kevin W Hatton
- Department of Anesthesiology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536, USA
| | - Lon R Hays
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA
| | - Abner O Rayapati
- Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA
| | - Joshua A Lile
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA; Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA; Department of Psychology, University of Kentucky College of Arts and Sciences, 171 Funkhouser Drive, Lexington, KY 40506-0044, USA
| | - Craig R Rush
- Department of Behavioral Science, University of Kentucky College of Medicine, 1100 Veterans Drive, Medical Behavioral Science Building, Lexington, KY 40536-0086, USA; Department of Psychiatry, University of Kentucky College of Medicine, 245 Fountain Court, Lexington, KY 40509-1810, USA; Department of Psychology, University of Kentucky College of Arts and Sciences, 171 Funkhouser Drive, Lexington, KY 40506-0044, USA
| |
Collapse
|
36
|
Aldridge GM, Zarin TA, Brandner AJ, George O, Gilpin NW, Repunte-Canonigo V, Sanna PP, Koob GF, Vendruscolo LF, Schmeichel BE. Effects of single and dual hypocretin-receptor blockade or knockdown of hypocretin projections to the central amygdala on alcohol drinking in dependent male rats. ADDICTION NEUROSCIENCE 2022; 3:100028. [PMID: 35965958 PMCID: PMC9365098 DOI: 10.1016/j.addicn.2022.100028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Hypocretin/Orexin (HCRT) is a neuropeptide that is associated with both stress and reward systems in humans and rodents. The different contributions of signaling at hypocretin-receptor 1 (HCRT-R1) and hypocretin-receptor 2 (HCRT-R2) to compulsive alcohol drinking are not yet fully understood. Thus, the current studies used pharmacological and viral-mediated targeting of HCRT to determine participation in compulsive alcohol drinking and measured HCRT-receptor mRNA expression in the extended amygdala of both alcohol-dependent and non-dependent male rats. Rats were made dependent through chronic intermittent exposure to alcohol vapor and were tested for the acute effect of HCRT-R1-selective (SB-408124; SB-R1), HCRT-R2-selective (NBI-80713; NB-R2), or dual HCRT-R1/2 (NBI-87571; NB-R1/2) antagonism on alcohol intake. NB-R2 and NB-R1/2 antagonists each dose-dependently decreased overall alcohol drinking in alcohol-dependent rats, whereas, SB-R1 decreased alcohol drinking in both alcohol-dependent and non-dependent rats at the highest dose (30 mg/kg). SB-R1, NB-R2, and NB-R1/2 treatment did not significantly affect water drinking in either alcohol-dependent or non-dependent rats. Additional PCR analyses revealed a significant decrease in Hcrtr1 mRNA expression within the central amygdala (CeA) of dependent rats under acute withdrawal conditions compared to nondependent rats. Lastly, a shRNA-encoding adeno-associated viral vector with retrograde function was used to knockdown HCRT in CeA-projecting neurons from the lateral hypothalamus (LH). LH-CeA HCRT knockdown significantly attenuated alcohol self-administration in alcohol-dependent rats. These observations suggest that HCRT signaling in the CeA is necessary for alcohol-seeking behavior during dependence. Together, these data highlight a role for both HCRT-R1 and -R2 in dependent alcohol-seeking behavior.
Collapse
Affiliation(s)
- Gabriel M. Aldridge
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Tyler A. Zarin
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Adam J. Brandner
- Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Olivier George
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- Department of Psychiatry, School of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Nicholas W. Gilpin
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Vez Repunte-Canonigo
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, United States
| | - Pietro P. Sanna
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, United States
| | - George F. Koob
- Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Leandro F. Vendruscolo
- Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Brooke E. Schmeichel
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
37
|
Villano I, La Marra M, Di Maio G, Monda V, Chieffi S, Guatteo E, Messina G, Moscatelli F, Monda M, Messina A. Physiological Role of Orexinergic System for Health. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:8353. [PMID: 35886210 PMCID: PMC9323672 DOI: 10.3390/ijerph19148353] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 02/06/2023]
Abstract
Orexins, or hypocretins, are excitatory neuropeptides involved in the regulation of feeding behavior and the sleep and wakefulness states. Since their discovery, several lines of evidence have highlighted that orexin neurons regulate a great range of physiological functions, giving it the definition of a multitasking system. In the present review, we firstly describe the mechanisms underlining the orexin system and their interactions with the central nervous system (CNS). Then, the system's involvement in goal-directed behaviors, sleep/wakefulness state regulation, feeding behavior and energy homeostasis, reward system, and aging and neurodegenerative diseases are described. Advanced evidence suggests that the orexin system is crucial for regulating many physiological functions and could represent a promising target for therapeutical approaches to obesity, drug addiction, and emotional stress.
Collapse
Affiliation(s)
- Ines Villano
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Marco La Marra
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Girolamo Di Maio
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Vincenzo Monda
- Department of Movement Sciences and Wellbeing, University of Naples “Parthenope”, 80138 Naples, Italy; (V.M.); (E.G.)
| | - Sergio Chieffi
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Ezia Guatteo
- Department of Movement Sciences and Wellbeing, University of Naples “Parthenope”, 80138 Naples, Italy; (V.M.); (E.G.)
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (G.M.); (F.M.)
| | - Fiorenzo Moscatelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy; (G.M.); (F.M.)
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| | - Antonietta Messina
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.L.M.); (G.D.M.); (S.C.); (M.M.); (A.M.)
| |
Collapse
|
38
|
Morales I. Brain regulation of hunger and motivation: The case for integrating homeostatic and hedonic concepts and its implications for obesity and addiction. Appetite 2022; 177:106146. [PMID: 35753443 DOI: 10.1016/j.appet.2022.106146] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 11/19/2022]
Abstract
Obesity and other eating disorders are marked by dysregulations to brain metabolic, hedonic, motivational, and sensory systems that control food intake. Classic approaches in hunger research have distinguished between hedonic and homeostatic processes, and have mostly treated these systems as independent. Hindbrain structures and a complex network of interconnected hypothalamic nuclei control metabolic processes, energy expenditure, and food intake while mesocorticolimbic structures are though to control hedonic and motivational processes associated with food reward. However, it is becoming increasingly clear that hedonic and homeostatic brain systems do not function in isolation, but rather interact as part of a larger network that regulates food intake. Incentive theories of motivation provide a useful route to explore these interactions. Adapting incentive theories of motivation can enable researchers to better how motivational systems dysfunction during disease. Obesity and addiction are associated with profound alterations to both hedonic and homeostatic brain systems that result in maladaptive patterns of consumption. A subset of individuals with obesity may experience pathological cravings for food due to incentive sensitization of brain systems that generate excessive 'wanting' to eat. Further progress in understanding how the brain regulates hunger and appetite may depend on merging traditional hedonic and homeostatic concepts of food reward and motivation.
Collapse
Affiliation(s)
- Ileana Morales
- Department of Psychology, University of Michigan, 530 Church Street, Ann Arbor, MI, 48109-1043, USA.
| |
Collapse
|
39
|
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
|
40
|
Daiwile AP, Jayanthi S, Cadet JL. Sex differences in methamphetamine use disorder perused from pre-clinical and clinical studies: Potential therapeutic impacts. Neurosci Biobehav Rev 2022; 137:104674. [PMID: 35452744 PMCID: PMC9119944 DOI: 10.1016/j.neubiorev.2022.104674] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 01/31/2023]
Abstract
Methamphetamine (METH) use, and misuse are associated with severe socioeconomic consequences. METH users develop tolerance, lose control over drug taking behaviors, and suffer frequent relapses even during treatment. The clinical course of METH use disorder is influenced by multifactorial METH-induced effects on the central and peripheral nervous systems. Although these METH-induced consequences are observed in humans of all ages, races, and sexes, sexual dimorphism in these outcomes have been observed in both pre-clinical and clinical settings. In this review, we have provided a detailed presentation of the sex differences reported in human and animal studies. We have therefore presented data that identified the influences of sex on METH pharmacokinetics, METH-induced changes in behaviors, cognitive processes, structural changes in the brain, and the effects of the drug on neurotransmitter systems and molecular mechanisms. Finally, we highlighted the potential significance of sex as a critical variable that should be considered when planning the development of new pharmacotherapeutic approaches against MEH use disorder in humans.
Collapse
Affiliation(s)
- Atul P Daiwile
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, USA.
| | - Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, USA.
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, USA.
| |
Collapse
|
41
|
Nazari S, Pourmand SM, Makki SM, Brand S, Vousooghi N. Potential biomarkers of addiction identified by real-time PCR in human peripheral blood lymphocytes: a narrative review. Biomark Med 2022; 16:739-758. [PMID: 35658670 DOI: 10.2217/bmm-2021-0291] [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: 11/21/2022] Open
Abstract
Addiction-related neurobiological factors could be considered as potential biomarkers. The concentration of peripheral biomarkers in tissues like blood lymphocytes may mirror their brain levels. This review is focused on the mRNA expression of potential addiction biomarkers in human peripheral blood lymphocytes (PBLs). PubMed, EMBASE, Web of Science, Scopus and Google Scholar were searched using the keywords 'addiction', 'biomarker', 'peripheral blood lymphocyte', 'gene expression' and 'real-time PCR'. The results showed the alterations in the regulation of genes such as dopamine receptors, opioid receptors, NMDA receptors, cannabinoid receptors, α-synuclein, DYN, MAO-A, FosB and orexin-A as PBLs biomarkers in addiction stages. Such variations could also be found during abstinence and relapse. PBLs biomarkers may help in drug development and have clinical implications.
Collapse
Affiliation(s)
- Shahrzad Nazari
- Department of Neuroscience & Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Seyed Mahmoud Pourmand
- Addiction Department, School of Behavioral Sciences & Mental Health (Tehran Institute of Psychiatry), Iran University of Medical Sciences, Tehran, 1445613111, Iran
| | - Seyed Mohammad Makki
- Department of Psychiatry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Serge Brand
- Center for Affective-, Stress- and Sleep Disorders (ZASS), Psychiatric Clinics (UPK), University of Basel, Basel, 4002, Switzerland.,Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, 6714869914, Iran.,Substance Abuse Prevention Research Center, Kermanshah University of Medical Sciences, Kermanshah, 6714869914, Iran.,Department of Sport, Exercise, and Health, Division of Sport Science and Psychosocial Health, University of Basel, Basel, 4052, Switzerland.,Department of Psychiatry, School of Medicine, Tehran University of Medical Sciences, Tehran, 1417466191, Iran
| | - Nasim Vousooghi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran.,Research Center for Cognitive & Behavioral Sciences, Tehran University of Medical Sciences, Tehran, 13337159140, Iran.,Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, 1336616357, Iran
| |
Collapse
|
42
|
Dunigan AI, Roseberry AG. Actions of feeding-related peptides on the mesolimbic dopamine system in regulation of natural and drug rewards. ADDICTION NEUROSCIENCE 2022; 2:100011. [PMID: 37220637 PMCID: PMC10201992 DOI: 10.1016/j.addicn.2022.100011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The mesolimbic dopamine system is the primary neural circuit mediating motivation, reinforcement, and reward-related behavior. The activity of this system and multiple behaviors controlled by it are affected by changes in feeding and body weight, such as fasting, food restriction, or the development of obesity. Multiple different peptides and hormones that have been implicated in the control of feeding and body weight interact with the mesolimbic dopamine system to regulate many different dopamine-dependent, reward-related behaviors. In this review, we summarize the effects of a selected set of feeding-related peptides and hormones acting within the ventral tegmental area and nucleus accumbens to alter feeding, as well as food, drug, and social reward.
Collapse
Affiliation(s)
- Anna I. Dunigan
- Department of Biology and Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Aaron G. Roseberry
- Department of Biology and Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
43
|
Kaplan GB, Lakis GA, Zhoba H. Sleep-Wake and Arousal Dysfunctions in Post-Traumatic Stress Disorder:Role of Orexin Systems. Brain Res Bull 2022; 186:106-122. [PMID: 35618150 DOI: 10.1016/j.brainresbull.2022.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 04/20/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a trauma-related condition that produces distressing fear memory intrusions, avoidance behaviors, hyperarousal/startle, stress responses and insomnia. This review focuses on the importance of the orexin neural system as a novel mechanism related to the pathophysiology of PTSD. Orexinergic neurons originate in the lateral hypothalamus and project widely to key neurotransmitter system neurons, autonomic neurons, the hypothalamic-pituitaryadrenal (HPA) axis, and fear-related neural circuits. After trauma or stress, the basolateral amygdala (BLA) transmits sensory information to the central nucleus of the amygdala (CeA) and in turn to the hypothalamus and other subcortical and brainstem regions to promote fear and threat. Orexin receptors have a prominent role in this circuit as fear conditioned orexin receptor knockout mice show decreased fear expression while dual orexin receptor antagonists (DORAs) inhibit fear acquisition and expression. Orexin activation of an infralimbic-amygdala circuit impedes fear extinction while DORA treatments enhance it. Increased orexin signaling to the amygdalocortical- hippocampal circuit promotes avoidance behaviors. Orexin has an important role in activating sympathetic nervous system (SNS) activity and the HPA axis stress responses. Blockade of orexin receptors reduces fear-conditioned startle responses. In PTSD models, individuals demonstrate sleep disturbances such as increased sleep latency and more transitions to wakefulness. Increased orexin activity impairs sleep by promoting wakefulness and reducing total sleep time while DORA treatments enhance sleep onset and maintenance. The orexinergic neural system provides important mechanisms for understanding multiple PTSD behaviors and provides new medication targets to treat this often persistent and debilitating illness.
Collapse
Affiliation(s)
- Gary B Kaplan
- Mental Health Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Department of Psychiatry, Boston University School of Medicine, Boston, MA, 02118 USA; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118 USA.
| | - Gabrielle A Lakis
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA; Undergraduate Program in Neuroscience, Boston University, Boston, MA, 02215 USA
| | - Hryhoriy Zhoba
- Research Service, VA Boston Healthcare System, West Roxbury, MA, 02132 USA
| |
Collapse
|
44
|
Kalló I, Omrani A, Meye FJ, de Jong H, Liposits Z, Adan RAH. Characterization of orexin input to dopamine neurons of the ventral tegmental area projecting to the medial prefrontal cortex and shell of nucleus accumbens. Brain Struct Funct 2022; 227:1083-1098. [PMID: 35029758 PMCID: PMC8930802 DOI: 10.1007/s00429-021-02449-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/29/2021] [Indexed: 11/29/2022]
Abstract
Orexin neurons are involved in homeostatic regulatory processes, including arousal and feeding, and provide a major input from the hypothalamus to the ventral tegmental area (VTA) of the midbrain. VTA neurons are a central hub processing reward and motivation and target the medial prefrontal cortex (mPFC) and the shell part of nucleus accumbens (NAcs). We investigated whether subpopulations of dopamine (DA) neurons in the VTA projecting either to the mPFC or the medial division of shell part of nucleus accumbens (mNAcs) receive differential input from orexin neurons and whether orexin exerts differential electrophysiological effects upon these cells. VTA neurons projecting to the mPFC or the mNAcs were traced retrogradely by Cav2-Cre virus and identified by expression of yellow fluorescent protein (YFP). Immunocytochemical analysis showed that a higher proportion of all orexin-innervated DA neurons projected to the mNAcs (34.5%) than to the mPFC (5.2%). Of all sampled VTA neurons projecting either to the mPFC or mNAcs, the dopaminergic (68.3 vs. 79.6%) and orexin-innervated DA neurons (68.9 vs. 64.4%) represented the major phenotype. Whole-cell current clamp recordings were obtained from fluorescently labeled neurons in slices during baseline periods and bath application of orexin A. Orexin similarly increased the firing rate of VTA dopamine neurons projecting to mNAcs (1.99 ± 0.61 Hz to 2.53 ± 0.72 Hz) and mPFC (0.40 ± 0.22 Hz to 1.45 ± 0.56 Hz). Thus, the hypothalamic orexin system targets mNAcs and to a lesser extent mPFC-projecting dopaminergic neurons of the VTA and exerts facilitatory effects on both clusters of dopamine neurons.
Collapse
Affiliation(s)
- Imre Kalló
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Eötvös Loránd Research Center, Budapest, 1083, Hungary
- Department of Neuroscience, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, 1083, Hungary
| | - Azar Omrani
- Department of Translational Neuroscience, UMC Brain Center, University Medical Center Utrecht, Universiteitsweg 100, 3584, Utrecht, The Netherlands
| | - Frank J Meye
- Department of Translational Neuroscience, UMC Brain Center, University Medical Center Utrecht, Universiteitsweg 100, 3584, Utrecht, The Netherlands
| | - Han de Jong
- Department of Translational Neuroscience, UMC Brain Center, University Medical Center Utrecht, Universiteitsweg 100, 3584, Utrecht, The Netherlands
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Eötvös Loránd Research Center, Budapest, 1083, Hungary.
- Department of Neuroscience, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, 1083, Hungary.
| | - Roger A H Adan
- Department of Translational Neuroscience, UMC Brain Center, University Medical Center Utrecht, Universiteitsweg 100, 3584, Utrecht, The Netherlands.
- Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 40530, Goteborg, Sweden.
| |
Collapse
|
45
|
Pantazis CB, James MH, O’Connor S, Shin N, Aston-Jones G. Orexin-1 receptor signaling in ventral tegmental area mediates cue-driven demand for cocaine. Neuropsychopharmacology 2022; 47:741-751. [PMID: 34635803 PMCID: PMC8782853 DOI: 10.1038/s41386-021-01173-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 08/10/2021] [Accepted: 08/27/2021] [Indexed: 02/03/2023]
Abstract
Drug-associated sensory cues increase motivation for drug and the orexin system is importantly involved in this stimulus-enhanced motivation. Ventral tegmental area (VTA) is a major target by which orexin signaling modulates reward behaviors, but it is unknown whether this circuit is necessary for cue-driven motivation for cocaine. Here, we investigated the role of VTA orexin signaling in cue-driven motivation for cocaine using a behavioral economics (BE) paradigm. We found that infusion of the orexin-1 receptor (Ox1R) antagonist SB-334867 (SB) into VTA prior to BE testing reduced motivation when animals were trained to self-administer cocaine with discrete cues and tested on BE with those cues. SB had no effect when animals were trained to self-administer cocaine without cues or tested on BE without cues, indicating that learning to associate cues with drug delivery during self-administration training was necessary for cues to recruit orexin signaling in VTA. These effects were specific to VTA, as injections of SB immediately dorsal had no effect. Moreover, intra-VTA SB did not have an impact on locomotor activity, or low- or high-effort consumption of sucrose. Finally, we microinjected a novel retrograde adeno-associated virus (AAVretro) containing an orexin-specific short hairpin RNA (OxshRNA) into VTA to knock down orexin in the hypothalamus-VTA circuit. These injections significantly reduced orexin expression in lateral hypothalamus (LH) and decreased cue-driven motivation. These studies demonstrate a role for orexin signaling in VTA, specifically when cues predict drug reward.
Collapse
Affiliation(s)
- Caroline B. Pantazis
- grid.430387.b0000 0004 1936 8796Brain Health Institute, Rutgers University/Rutgers Biomedical and Health Sciences, Piscataway, NJ USA ,grid.94365.3d0000 0001 2297 5165Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD USA
| | - Morgan H. James
- grid.430387.b0000 0004 1936 8796Brain Health Institute, Rutgers University/Rutgers Biomedical and Health Sciences, Piscataway, NJ USA ,grid.430387.b0000 0004 1936 8796Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers Unviversity, Piscataway, NJ USA
| | - Shayna O’Connor
- grid.430387.b0000 0004 1936 8796Brain Health Institute, Rutgers University/Rutgers Biomedical and Health Sciences, Piscataway, NJ USA
| | - Noah Shin
- grid.430387.b0000 0004 1936 8796Brain Health Institute, Rutgers University/Rutgers Biomedical and Health Sciences, Piscataway, NJ USA ,grid.430387.b0000 0004 1936 8796Cell Biology and Neuroscience Department, Rutgers University-New Brunswick, Piscataway, NJ USA
| | - Gary Aston-Jones
- Brain Health Institute, Rutgers University/Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA.
| |
Collapse
|
46
|
Neurobiology of the Orexin System and Its Potential Role in the Regulation of Hedonic Tone. Brain Sci 2022; 12:brainsci12020150. [PMID: 35203914 PMCID: PMC8870430 DOI: 10.3390/brainsci12020150] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 01/13/2023] Open
Abstract
Orexin peptides comprise two neuropeptides, orexin A and orexin B, that bind two G-protein coupled receptors (GPCRs), orexin receptor 1 (OXR1) and orexin receptor 2 (OXR2). Although cell bodies that produce orexin peptides are localized in a small area comprising the lateral hypothalamus and adjacent regions, orexin-containing fibres project throughout the neuraxis. Although orexins were initially described as peptides that regulate feeding behaviour, research has shown that orexins are involved in diverse functions that range from the modulation of autonomic functions to higher cognitive functions, including reward-seeking, behaviour, attention, cognition, and mood. Furthermore, disruption in orexin signalling has been shown in mood disorders that are associated with low hedonic tone or anhedonia, including depression, anxiety, attention deficit hyperactivity disorder, and addiction. Notably, projections of orexin neurons overlap circuits involved in the modulation of hedonic tone. Evidence shows that orexins may potentiate hedonic behaviours by increasing the feeling of pleasure or reward to various signalling, whereas dysregulation of orexin signalling may underlie low hedonic tone or anhedonia. Further, orexin appears to play a key role in regulating behaviours in motivationally charged situations, such as food-seeking during hunger, or drug-seeking during withdrawal. Therefore, it would be expected that dysregulation of orexin expression or signalling is associated with changes in hedonic tone. Further studies investigating this association are warranted.
Collapse
|
47
|
McGregor R, Wu MF, Holmes B, Lam HA, Maidment NT, Gera J, Yamanaka A, Siegel JM. Hypocretin/Orexin Interactions with Norepinephrine Contribute to the Opiate Withdrawal Syndrome. J Neurosci 2022; 42:255-263. [PMID: 34853083 PMCID: PMC8802943 DOI: 10.1523/jneurosci.1557-21.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/21/2021] [Accepted: 10/09/2021] [Indexed: 11/21/2022] Open
Abstract
We previously found that human heroin addicts and mice chronically exposed to morphine exhibit a significant increase in the number of detected hypocretin/orexin (Hcrt)-producing neurons. However, it remains unknown how this increase affects target areas of the hypocretin system involved in opioid withdrawal, including norepinephrine containing structures locus coeruleus (LC) and A1/A2 medullary regions. Using a combination of immunohistochemical, biochemical, imaging, and behavioral techniques, we now show that the increase in detected hypocretin cell number translates into a significant increase in hypocretin innervation and tyrosine hydroxylase (TH) levels in the LC without affecting norepinephrine-containing neuronal cell number. We show that the increase in TH is completely dependent on Hcrt innervation. The A1/A2 regions were unaffected by morphine treatment. Manipulation of the Hcrt system may affect opioid addiction and withdrawal.SIGNIFICANCE STATEMENT Previously, we have shown that the hypothalamic hypocretin system undergoes profound anatomic changes in human heroin addicts and in mice exposed to morphine, suggesting a role of this system in the development of addictive behaviors. The locus coeruleus plays a key role in opioid addiction. Here we report that the hypothalamic hypocretin innervation of the locus coeruleus increases dramatically with morphine administration to mice. This increase is correlated with a massive increase in tyrosine hydroxylase expression in locus coeruleus. Elimination of hypocretin neurons prevents the tyrosine hydroxylase increase in locus coeruleus and dampens the somatic and affective components of opioid withdrawal.
Collapse
Affiliation(s)
- Ronald McGregor
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California 90095
- Veterans Administration Greater Los Angeles Healthcare System, North Hills, Los Angeles, California 91343
| | - Ming-Fung Wu
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California 90095
- Veterans Administration Greater Los Angeles Healthcare System, North Hills, Los Angeles, California 91343
| | - Brent Holmes
- Veterans Administration Greater Los Angeles Healthcare System, North Hills, Los Angeles, California 91343
- Department of Medicine, University of California, Los Angeles, Los Angeles, 90095
| | - Hoa Anh Lam
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California 90095
- Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, California 90095
| | - Nigel T Maidment
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California 90095
- Brain Research Institute, University of California, Los Angeles, Los Angeles, California 90095
- Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, California 90095
| | - Joseph Gera
- Veterans Administration Greater Los Angeles Healthcare System, North Hills, Los Angeles, California 91343
- Department of Medicine, University of California, Los Angeles, Los Angeles, 90095
- Jonnson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, 90095
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, 90095
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Jerome M Siegel
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California 90095
- Veterans Administration Greater Los Angeles Healthcare System, North Hills, Los Angeles, California 91343
- Brain Research Institute, University of California, Los Angeles, Los Angeles, California 90095
| |
Collapse
|
48
|
Brown RM, Dayas CV, James MH, Smith RJ. New directions in modelling dysregulated reward seeking for food and drugs. Neurosci Biobehav Rev 2022; 132:1037-1048. [PMID: 34736883 PMCID: PMC8816817 DOI: 10.1016/j.neubiorev.2021.10.043] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 10/08/2021] [Accepted: 10/19/2021] [Indexed: 01/03/2023]
Abstract
Behavioral models are central to behavioral neuroscience. To study the neural mechanisms of maladaptive behaviors (including binge eating and drug addiction), it is essential to develop and utilize appropriate animal models that specifically focus on dysregulated reward seeking. Both food and cocaine are typically consumed in a regulated manner by rodents, motivated by reward and homeostatic mechanisms. However, both food and cocaine seeking can become dysregulated, resulting in binge-like consumption and compulsive patterns of intake. The speakers in this symposium for the 2021 International Behavioral Neuroscience Meeting utilize behavioral models of dysregulated reward-seeking to investigate the neural mechanisms of binge-like consumption, enhanced cue-driven reward seeking, excessive motivation, and continued use despite negative consequences. In this review, we outline examples of maladaptive patterns of intake and explore recent animal models that drive behavior to become dysregulated, including stress exposure and intermittent access to rewards. Lastly, we explore select behavioral and neural mechanisms underlying dysregulated reward-seeking for both food and drugs.
Collapse
Affiliation(s)
- Robyn M Brown
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC, 3052, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia.
| | - Christopher V Dayas
- School of Biomedical Sciences & Pharmacy, Faculty of Health, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Morgan H James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA; Brain Health Institute, Rutgers University, Piscataway, NJ, 08854, USA.
| | - Rachel J Smith
- Department of Psychological & Brain Sciences, Institute for Neuroscience, Texas A&M University, College Station, TX, 77843, USA
| |
Collapse
|
49
|
Guo L, Hu A, Zhao X, Xiang X. Reduction of Orexin-A Is Associated With Anxiety and the Level of Depression of Male Methamphetamine Users During the Initial Withdrawal Period. Front Psychiatry 2022; 13:900135. [PMID: 35859609 PMCID: PMC9289462 DOI: 10.3389/fpsyt.2022.900135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 06/10/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Orexin has been linked to the regulation of reward and motivation in animals, but there have been few human studies to validate its regulatory effects. We aimed to determine how orexin-A levels changed during different stages of withdrawal, as well as the relationship between orexin-A levels and withdrawal symptoms in male METH users. METHODS This study included 76 METH users and 35 control participants. The METH users were divided into three groups: group 1 (abstinence within 1 week, n = 23), group 2 (abstinence between 1 week and 3 months, n = 38), and group 3 (abstinence over 3 months, n = 15). At baseline and 1 month of enrollment, the plasma orexin-A level was examined. To track the withdrawal symptoms, self-report questionnaires (anxiety, depression, craving, and sleep quality) were collected at two points. RESULTS The orexin-A levels of groups 1 (p < 0.001) and 2 (p < 0.001) were lower than that of the controls at baseline but not group 3. One month later, the orexin-A levels of group 2 (p < 0.05) significantly increased, while no significant changes in those of groups 1 and 3 were observed. Furthermore, the orexin-A levels of group 1 were positively linked with depression (p < 0.01) and anxiety (p < 0.01) at baseline. CONCLUSIONS The decrease in orexin-A levels was only transitory during the initial abstinence phase, and it was eventually restored near to normal with continued abstinence among the male METH users. Furthermore, a lower concentration of orexin-A may serve as a risk factor for negative emotions during METH withdrawal.
Collapse
Affiliation(s)
- Lei Guo
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Aqian Hu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoxi Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaojun Xiang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
50
|
Orexin receptor blockers: A tool for lowering alcohol intake and alcohol addictive behavior in the light of preclinical studies. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Alcohol use disorder (AUD) is a severe and globally widespread neurological and psychiatric problem. The treatment with currently used drugs often does not bring the expected effect. New optimization methods or directions in pharmacotherapy are still being sought. The group of bioactive ligands, targeted at neuropeptides called orexins (OXs) and their receptors (OXRs), affects a number of functions including ingestion, sleep-wake regulation, as well as the brain reward system which is the basis of addiction.
The purpose of this paper is to systematize the knowledge in the field of preclinical behavioral studies on rodents (rats and mice) in several models of alcohol consumption using the OXRs antagonists.
The results of the experiments indicated a potential efficacy of particular OXRs antagonists in the AUD treatment, especially those selectively blocking the OX1R. Among them, SB-334867 in the lowest effective dose of 3 mg/kg i.p. was most studied, as shown in the model of two-bottle choice using C57BL/6 mice. Moreover, this compound did not affect the reduction of cognitive functions. GSK1059865 was also involved in the selective reduction of ethanol intake, and simultaneously did not alter the consumption of sugar solution. The other group of selective OX2R antagonists, such as TCS-OX2-29 and LSN2424100, was less efficient.
In summary, the OX1R antagonists proved to have the potential in AUD therapy, not only through the reduction of ethanol consumption but also in the treatment of coexisting behavioral and physiological disorders, such as insomnia and anxiety.
Collapse
|