1
|
Somach RT, Lim MM, Cohen AS. Effects of Traumatic Brain Injury on the Orexin/Hypocretin System. Neurotrauma Rep 2025; 6:322-335. [PMID: 40309161 PMCID: PMC12040569 DOI: 10.1089/neur.2024.0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Traumatic Brain Injuries (TBIs) are known to cause a myriad of symptoms in patients. One common symptom after injury is sleep disruptions. One neuropeptide system has been studied repeatedly as a potential cause of sleep disruptions after TBI- the orexin/hypocretin system. Orexin promotes wakefulness and arousal while disrupting the orexin system causes increased sleepiness and narcolepsy. Studies of TBI in human and animal subjects have shown that TBI affects the orexin system. This review serves as an overview of how TBI affects the orexin/hypocretin system, including structural and functional changes to the neurons after injury. This review is the first to include studies that examine how TBI affects orexin/hypocretin receptors. This review also examines how sex is accounted for in the studies of the orexin system after TBI.
Collapse
Affiliation(s)
| | - Miranda M. Lim
- Oregon Health and Science University, Portland, Oregon, USA
- VA Portland Health Care System, Portland, Oregon, USA
| | - Akiva S. Cohen
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
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 PMCID: PMC12078786 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] [Download PDF] [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
|
3
|
Equihua-Benítez AC, Espinoza-Abad R, García-García F. Sleep Loss and Substance Use Disorders: An Issue from Adolescents to Adults. Behav Sci (Basel) 2025; 15:220. [PMID: 40001851 PMCID: PMC11852296 DOI: 10.3390/bs15020220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Unsatisfactory sleep is a worldwide concern, as evidenced by the high prevalence of insomnia symptoms and diagnosis in the general population, and an issue that has also risen among adolescents. These circumstances are a cause of worry due to, among other factors, the observed bidirectional association of sleep disturbances and the risk of substance use disorder development. In this regard, across the globe, several reports indicate that substance consumption is at an all-time high, with alcohol, nicotine, and cannabis leading the charts. Additionally, the age of onset has dropped, with reports suggesting that first contact is usually during adolescence. Although the nature of the link between poor sleep and substance use disorder development is still not fully understood, it is possible that an overactive orexinergic system could play a role, as it has been observed that treatment with orexinergic antagonists improves insomnia symptoms and that postmortem studies show an increase in orexin immunoreactive neurons in sections obtained from habitual opioid consumers. We further argue that it is during adolescence that this maladaptive loop can be established, priming for the development of substance use disorders.
Collapse
Affiliation(s)
- Ana Clementina Equihua-Benítez
- Biology Sleep Laboratory, Biomedicine Department, Health Sciences Institute, Veracruzana University, Xalapa 91190, Ver, Mexico;
| | - Rodolfo Espinoza-Abad
- Graduate Program in Health Sciences, Health Sciences Institute, Veracruzana University, Xalapa 91190, Ver, Mexico;
| | - Fabio García-García
- Biology Sleep Laboratory, Biomedicine Department, Health Sciences Institute, Veracruzana University, Xalapa 91190, Ver, Mexico;
| |
Collapse
|
4
|
Martin T, Pasquier F, Denise P, Davenne D, Quarck G. The relationship between the vestibular system and the circadian timing system: A review. Sleep Med 2025; 126:148-158. [PMID: 39672094 DOI: 10.1016/j.sleep.2024.12.002] [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: 05/16/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
This review attempts to analyze the relationship between the vestibular system and the circadian timing system. The activity of the biological clock allows an organism to optimally perform its tasks throughout the nychtemeron. To achieve this, the biological clock is subjected to exogenous factors that entrain it to a 24h period. While the most powerful synchronizer is the light-dark cycle produced by the Earth's rotation, research has led to the hypothesis of the vestibular system as a possible non-photic time cue used to entrain circadian rhythms. Demonstrated neuroanatomical pathways between vestibular nuclei and suprachiasmatic nuclei could transmit this message. Moreover, functional evidence in both humans and animals has shown that vestibular disruption or stimulation may lead to changes in circadian rhythms characteristics. Vestibular stimulations could be considered to act synergistically with other synchronizers, such as light, to ensure the entrainment of biological rhythms over the 24-h reference period.
Collapse
Affiliation(s)
- Tristan Martin
- Le Mans Université, Movement - Interactions - Performance, MIP, UR 4334, Avenue Olivier Messiaen, 72085 Le Mans CEDEX 9, France; Université de Caen Normandie, INSERM, Normandie Université, COMETE UMR-S 1075, GIP Cyceron, Caen, F-14000, France.
| | - Florane Pasquier
- Université de Caen Normandie, INSERM, Normandie Université, COMETE UMR-S 1075, GIP Cyceron, Caen, F-14000, France
| | - Pierre Denise
- Université de Caen Normandie, INSERM, Normandie Université, COMETE UMR-S 1075, GIP Cyceron, Caen, F-14000, France
| | - Damien Davenne
- Université de Caen Normandie, INSERM, Normandie Université, COMETE UMR-S 1075, GIP Cyceron, Caen, F-14000, France
| | - Gaëlle Quarck
- Université de Caen Normandie, INSERM, Normandie Université, COMETE UMR-S 1075, GIP Cyceron, Caen, F-14000, France
| |
Collapse
|
5
|
Dyachuk V. The Role and Mechanisms of the Hypocretin System in Zebrafish ( Danio rerio). Int J Mol Sci 2024; 26:256. [PMID: 39796111 PMCID: PMC11719587 DOI: 10.3390/ijms26010256] [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: 11/06/2024] [Revised: 12/28/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Sleep is the most important physiological function of all animals studied to date. Sleep disorders include narcolepsy, which is characterized by excessive daytime sleepiness, disruption of night sleep, and muscle weakness-cataplexy. Narcolepsy is known to be caused by the degeneration of orexin-synthesizing neurons (hypocretin (HCRT) neurons or orexin neurons) in the hypothalamus. In mammals, HCRT neurons primarily regulate the sleep/wake cycle, nutrition, reward seeking, and addiction development. The hypocretin system of the brain is involved in a number of neurological disorders. The distinctive pathologies associated with the disruption of HCRT neurons are narcolepsy and cataplexy, which are caused by the loss of hypocretin neurons that produce HCRT. In Danio, the hypocretin system is also involved in the regulation of sleep and wakefulness. It is represented by a single hcrt gene that encodes the peptides HCRT1 and HCRT2, as well as one HCRT receptor (HCRTR), which is structurally closest to the mammalian HCRTR2. The overexpression of the hcrt gene in Danio rerio larvae causes wakefulness, whereas the physical destruction of HCRT cells or a pharmacological blockade of the type 2 hypocretin receptor leads to fragmentation of sleep in fish larvae, which is also observed in patients with narcolepsy. These data confirm the evolutionary conservatism of the hypocretin system. Thus, Danio rerio is an ideal model for studying the functions of HCRT neural networks and their functions.
Collapse
Affiliation(s)
- Vyacheslav Dyachuk
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690041 Vladivostok, Russia
| |
Collapse
|
6
|
Bjorness TE, Greene RW. Orexin-mediated motivated arousal and reward seeking. Peptides 2024; 180:171280. [PMID: 39159833 DOI: 10.1016/j.peptides.2024.171280] [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: 05/19/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024]
Abstract
The neuromodulator orexin has been identified as a key factor for motivated arousal including recent evidence that sleep deprivation-induced enhancement of reward behavior is modulated by orexin. While orexin is not necessary for either reward or arousal behavior, orexin neurons' broad projections, ability to sense the internal state of the animal, and high plasticity of signaling in response to natural rewards and drugs of abuse may underlie heightened drug seeking, particularly in a subset of highly motivated reward seekers. As such, orexin receptor antagonists have gained deserved attention for putative use in addiction treatments. Ongoing and future clinical trials are expected to identify individuals most likely to benefit from orexin receptor antagonist treatment to promote abstinence, such as those with concurrent sleep disorders or high craving, while attention to methodological considerations will aid interpretation of the numerous preclinical studies investigating disparate aspects of the role of orexin in reward and arousal.
Collapse
Affiliation(s)
- Theresa E Bjorness
- Research Service, VA North Texas Health Care System, Dallas, TX 75126, USA; Departments of Psychiatry University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Robert W Greene
- Departments of Psychiatry University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba 305-8577, Japan
| |
Collapse
|
7
|
Sagi D, Tibi M, Admati I, Lerer-Goldshtein T, Hochgerner H, Zeisel A, Appelbaum L. Single-Cell Profiling Uncovers Evolutionary Divergence of Hypocretin/Orexin Neuronal Subpopulations. J Neurosci 2024; 44:e0095242024. [PMID: 39122556 PMCID: PMC11376333 DOI: 10.1523/jneurosci.0095-24.2024] [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: 01/15/2024] [Revised: 06/17/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024] Open
Abstract
Brain nuclei are traditionally defined by their anatomy, activity, and expression of specific markers. The hypothalamus contains discrete neuronal populations that coordinate fundamental behavioral functions, including sleep and wakefulness, in all vertebrates. Particularly, the diverse roles of hypocretin/orexin (Hcrt)-releasing neurons suggest functional heterogeneity among Hcrt neurons. Using single-cell RNA sequencing (scRNA-seq) and high-resolution imaging of the adult male and female zebrafish hypothalamic periventricular zone, we identified 21 glutamatergic and 28 GABAergic cell types. Integration of zebrafish and mouse scRNA-seq revealed evolutionary conserved and divergent hypothalamic cell types. The expression of specific genes, including npvf, which encodes a sleep-regulating neuropeptide, was enriched in subsets of glutamatergic Hcrt neurons in both larval and adult zebrafish. The genetic profile, activity, and neurite processing of the neuronal subpopulation that coexpresses both Hcrt and Npvf (Hcrt+Npvf+) differ from other Hcrt neurons. These interspecies findings provide a unified annotation of hypothalamic cell types and suggest that the heterogeneity of Hcrt neurons enables multifunctionality, such as consolidation of both wake and sleep by the Hcrt- and Npvf-releasing neuronal subpopulation.
Collapse
Affiliation(s)
- Dana Sagi
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 590002, Israel
| | - Muhammad Tibi
- The Faculty of Biotechnology and Food Engineering, Technion 3200, Israel
| | - Inbal Admati
- The Faculty of Biotechnology and Food Engineering, Technion 3200, Israel
| | - Tali Lerer-Goldshtein
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 590002, Israel
| | - Hannah Hochgerner
- The Faculty of Biotechnology and Food Engineering, Technion 3200, Israel
| | - Amit Zeisel
- The Faculty of Biotechnology and Food Engineering, Technion 3200, Israel
| | - Lior Appelbaum
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 590002, Israel
| |
Collapse
|
8
|
Illenberger JM, Flores-Ramirez FJ, Pascasio G, Franco M, Mendonsa B, Martin-Fardon R. Pivotal role of orexin signaling in the posterior paraventricular nucleus of the thalamus during the stress-induced reinstatement of oxycodone-seeking behavior. J Psychopharmacol 2024; 38:647-660. [PMID: 38888086 PMCID: PMC11407285 DOI: 10.1177/02698811241260989] [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] [Indexed: 06/20/2024]
Abstract
BACKGROUND The orexin (OX) system has received increasing interest as a potential target for treating substance use disorder. OX transmission in the posterior paraventricular nucleus of the thalamus (pPVT), an area activated by highly salient stimuli that are both reinforcing and aversive, mediates cue- and stress-induced reinstatement of reward-seeking behavior. Oral administration of suvorexant (SUV), a dual OX receptor (OXR) antagonist (DORA), selectively reduced conditioned reinstatement of oxycodone-seeking behavior and stress-induced reinstatement of alcohol-seeking behavior in dependent rats. AIMS This study tested whether OXR blockade in the pPVT with SUV reduces oxycodone or sweetened condensed milk (SCM) seeking elicited by conditioned cues or stress. METHODS Male Wistar rats were trained to self-administer oxycodone (0.15 mg/kg, i.v., 8 h/day) or SCM (0.1 ml, 2:1 dilution [v/v], 30 min/day). After extinction, we tested the ability of intra-pPVT SUV (15 µg/0.5 µl) to prevent reinstatement of oxycodone or SCM seeking elicited by conditioned cues or footshock stress. RESULTS The rats acquired oxycodone and SCM self-administration, and oxycodone intake correlated with signs of physical opioid withdrawal, confirming dependence. Following extinction, the presentation of conditioned cues or footshock elicited reinstatement of oxycodone- and SCM-seeking behavior. Intra-pPVT SUV blocked stress-induced reinstatement of oxycodone seeking but not conditioned reinstatement of oxycodone or SCM seeking or stress-induced reinstatement of SCM seeking. CONCLUSIONS The results indicate that OXR signaling in the pPVT is critical for stress-induced reinstatement of oxycodone seeking, further corroborating OXRs as treatment targets for opioid use disorder.
Collapse
|
9
|
Harel BT, Gattuso JJ, Latzman RD, Maruff P, Scammell TE, Plazzi G. The nature and magnitude of cognitive impairment in narcolepsy type 1, narcolepsy type 2, and idiopathic hypersomnia: a meta-analysis. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2024; 5:zpae043. [PMID: 39036743 PMCID: PMC11258808 DOI: 10.1093/sleepadvances/zpae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Indexed: 07/23/2024]
Abstract
People with narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), and idiopathic hypersomnia (IH) often report cognitive impairment which can be quite burdensome but is rarely evaluated in routine clinical practice. In this systematic review and meta-analysis, we assessed the nature and magnitude of cognitive impairment in NT1, NT2, and IH in studies conducted from January 2000 to October 2022. We classified cognitive tests assessing memory, executive function, and attention by cognitive domain. Between-group differences were analyzed as standardized mean differences (Cohen's d), and Cohen's d for individual tests were integrated according to cognitive domain and clinical disease group. Eighty-seven studies were screened for inclusion; 39 satisfied inclusion criteria, yielding 73 comparisons (k): NT1, k = 60; NT2, k = 8; IH, k = 5. Attention showed large impairment in people with NT1 (d = -0.90) and IH (d = -0.97), and moderate impairment in NT2 (d = -0.60). Executive function was moderately impaired in NT1 (d = -0.30) and NT2 (d = -0.38), and memory showed small impairments in NT1 (d = -0.33). A secondary meta-analysis identified sustained attention as the most impaired domain in NT1, NT2, and IH (d ≈ -0.5 to -1). These meta-analyses confirm that cognitive impairments are present in NT1, NT2, and IH, and provide quantitative confirmation of reports of cognitive difficulties made by patients and clinicians. These findings provide a basis for the future design of studies to determine whether cognitive impairments can improve with pharmacologic and nonpharmacologic treatments for narcolepsy and IH.
Collapse
Affiliation(s)
- Brian T Harel
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - James J Gattuso
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Robert D Latzman
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | | | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Giuseppe Plazzi
- IRCCS-Institute of Neurological Sciences, Bologna, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
10
|
Borgatti DA, Rowlett JK, Berro LF. Effects of methamphetamine on actigraphy-based sleep parameters in female rhesus monkeys: Orexin receptor mechanisms. Drug Alcohol Depend 2024; 259:111285. [PMID: 38636173 PMCID: PMC11111337 DOI: 10.1016/j.drugalcdep.2024.111285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/05/2024] [Accepted: 04/01/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND The orexin system has been implicated as a mechanism underlying insomnia and methamphetamine-induced sleep disruptions, with a potential role for OX2 receptors in the sleep-modulating effects of orexin. The aim of the present study was to investigate the extent to which orexin receptors mediate the effects of acute methamphetamine administration on actigraphy-based sleep in female rhesus monkeys. METHODS Actigraphy-based sleep measures were obtained in female rhesus monkeys (n=5) under baseline and acute test conditions. First, morning (10h) i.m. injections of methamphetamine (0.03 - 0.56mg/kg) were administered to determine the effects of methamphetamine alone. Then, saline or methamphetamine (0.3mg/kg) were administered at 10h, and evening (17h30) oral treatments with vehicle, the non-selective orexin receptor antagonist suvorexant (1 - 10mg/kg, p.o.), or the OX2-selective orexin receptor antagonist MK-1064 (1 - 10mg/kg, p.o.) were given. The ability of suvorexant and MK-1064 (10mg/kg, p.o.) to improve actigraphy-based sleep was also assessed in a group of female monkeys quantitatively identified with "short-duration sleep" (n=4). RESULTS Methamphetamine dose-dependently disrupted actigraphy-based sleep parameters. Treatment with either suvorexant or MK-1064 dose-dependently improved actigraphy-based sleep in monkeys treated with methamphetamine. Additionally, both suvorexant and MK-1064 promoted actigraphy-based sleep in a group of monkeys with baseline short actigraphy-based sleep. CONCLUSIONS These findings suggest that orexin-mediated mechanisms play a role in the effects of methamphetamine on actigraphy-based sleep in female monkeys. Targeting the orexin system, in particular OX2 receptors, could be an effective option for treating sleep disruptions observed in individuals with methamphetamine use disorder.
Collapse
Affiliation(s)
- Daniel A Borgatti
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA; Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - James K Rowlett
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA; Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA; 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, University of Mississippi Medical Center, Jackson, MS 39216, USA; Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA; Center for Innovation and Discovery in Addictions, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
11
|
Gan HW, Cerbone M, Dattani MT. Appetite- and Weight-Regulating Neuroendocrine Circuitry in Hypothalamic Obesity. Endocr Rev 2024; 45:309-342. [PMID: 38019584 PMCID: PMC11074800 DOI: 10.1210/endrev/bnad033] [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: 02/04/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Since hypothalamic obesity (HyOb) was first described over 120 years ago by Joseph Babinski and Alfred Fröhlich, advances in molecular genetic laboratory techniques have allowed us to elucidate various components of the intricate neurocircuitry governing appetite and weight regulation connecting the hypothalamus, pituitary gland, brainstem, adipose tissue, pancreas, and gastrointestinal tract. On a background of an increasing prevalence of population-level common obesity, the number of survivors of congenital (eg, septo-optic dysplasia, Prader-Willi syndrome) and acquired (eg, central nervous system tumors) hypothalamic disorders is increasing, thanks to earlier diagnosis and management as well as better oncological therapies. Although to date the discovery of several appetite-regulating peptides has led to the development of a range of targeted molecular therapies for monogenic obesity syndromes, outside of these disorders these discoveries have not translated into the development of efficacious treatments for other forms of HyOb. This review aims to summarize our current understanding of the neuroendocrine physiology of appetite and weight regulation, and explore our current understanding of the pathophysiology of HyOb.
Collapse
Affiliation(s)
- Hoong-Wei Gan
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Manuela Cerbone
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Mehul Tulsidas Dattani
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
12
|
Stanyer EC, Hoffmann J, Holland PR. Orexins and primary headaches: an overview of the neurobiology and clinical impact. Expert Rev Neurother 2024; 24:487-496. [PMID: 38517280 PMCID: PMC11034548 DOI: 10.1080/14737175.2024.2328728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/19/2024] [Indexed: 03/23/2024]
Abstract
INTRODUCTION Primary headaches, including migraines and cluster headaches, are highly prevalent disorders that significantly impact quality of life. Several factors suggest a key role for the hypothalamus, including neuroimaging studies, attack periodicity, and the presence of altered homeostatic regulation. The orexins are two neuropeptides synthesized almost exclusively in the lateral hypothalamus with widespread projections across the central nervous system. They are involved in an array of functions including homeostatic regulation and nociception, suggesting a potential role in primary headaches. AREAS COVERED This review summarizes current knowledge of the neurobiology of orexins, their involvement in sleep-wake regulation, nociception, and functions relevant to the associated symptomology of headache disorders. Preclinical reports of the antinociceptive effects of orexin-A in preclinical models are discussed, as well as clinical evidence for the potential involvement of the orexinergic system in headache. EXPERT OPINION Several lines of evidence support the targeted modulation of orexinergic signaling in primary headaches. Critically, orexins A and B, acting differentially via the orexin 1 and 2 receptors, respectively, demonstrate differential effects on trigeminal pain processing, indicating why dual-receptor antagonists failed to show clinical efficacy. The authors propose that orexin 1 receptor agonists or positive allosteric modulators should be the focus of future research.
Collapse
Affiliation(s)
- Emily C. Stanyer
- Headache Group, Wolfson Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- Sir Jules Thorne Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jan Hoffmann
- Headache Group, Wolfson Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Philip R. Holland
- Headache Group, Wolfson Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| |
Collapse
|
13
|
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
|
14
|
Tagawa N, Mori K, Koebis M, Aiba A, Iino Y, Tsuneoka Y, Funato H. Activation of lateral preoptic neurons is associated with nest-building in male mice. Sci Rep 2024; 14:8346. [PMID: 38594484 PMCID: PMC11004109 DOI: 10.1038/s41598-024-59061-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/06/2024] [Indexed: 04/11/2024] Open
Abstract
Nest-building behavior is a widely observed innate behavior. A nest provides animals with a secure environment for parenting, sleep, feeding, reproduction, and temperature maintenance. Since animal infants spend their time in a nest, nest-building behavior has been generally studied as parental behaviors, and the medial preoptic area (MPOA) neurons are known to be involved in parental nest-building. However, nest-building of singly housed male mice has been less examined. Here we show that male mice spent longer time in nest-building at the early to middle dark phase and at the end of the dark phase. These two periods are followed by sleep-rich periods. When a nest was removed and fresh nest material was introduced, both male and female mice built nests at Zeitgeber time (ZT) 6, but not at ZT12. Using Fos-immunostaining combined with double in situ hybridization of Vgat and Vglut2, we found that Vgat- and Vglut2-positive cells of the lateral preoptic area (LPOA) were the only hypothalamic neuron population that exhibited a greater number of activated cells in response to fresh nest material at ZT6, compared to being naturally awake at ZT12. Fos-positive LPOA neurons were negative for estrogen receptor 1 (Esr1). Both Vgat-positive and Vglut2-positive neurons in both the LPOA and MPOA were activated at pup retrieval by male mice. Our findings suggest the possibility that GABAergic and glutamatergic neurons in the LPOA are associated with nest-building behavior in male mice.
Collapse
Affiliation(s)
- Natsuki Tagawa
- Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, 143-8540, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Keita Mori
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Michinori Koebis
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Atsu Aiba
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-Ku, Tokyo, 113-0033, Japan
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Yuichi Iino
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Yousuke Tsuneoka
- Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, 143-8540, Japan.
| | - Hiromasa Funato
- Department of Anatomy, Graduate School of Medicine, Toho University, Tokyo, 143-8540, Japan.
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
15
|
Choi PP, Wang Q, Brenner LA, Li AJ, Ritter RC, Appleyard SM. Lesion of NPY Receptor-expressing Neurons in Perifornical Lateral Hypothalamus Attenuates Glucoprivic Feeding. Endocrinology 2024; 165:bqae021. [PMID: 38368624 PMCID: PMC11043786 DOI: 10.1210/endocr/bqae021] [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: 06/07/2023] [Revised: 01/19/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Glucoprivic feeding is one of several counterregulatory responses (CRRs) that facilitates restoration of euglycemia following acute glucose deficit (glucoprivation). Our previous work established that glucoprivic feeding requires ventrolateral medullary (VLM) catecholamine (CA) neurons that coexpress neuropeptide Y (NPY). However, the connections by which VLM CA/NPY neurons trigger increased feeding are uncertain. We have previously shown that glucoprivation, induced by an anti-glycolygic agent 2-deoxy-D-glucose (2DG), activates perifornical lateral hypothalamus (PeFLH) neurons and that expression of NPY in the VLM CA/NPY neurons is required for glucoprivic feeding. We therefore hypothesized that glucoprivic feeding and possibly other CRRs require NPY-sensitive PeFLH neurons. To test this, we used the ribosomal toxin conjugate NPY-saporin (NPY-SAP) to selectively lesion NPY receptor-expressing neurons in the PeFLH of male rats. We found that NPY-SAP destroyed a significant number of PeFLH neurons, including those expressing orexin, but not those expressing melanin-concentrating hormone. The PeFLH NPY-SAP lesions attenuated 2DG-induced feeding but did not affect 2DG-induced increase in locomotor activity, sympathoadrenal hyperglycemia, or corticosterone release. The 2DG-induced feeding response was also significantly attenuated in NPY-SAP-treated female rats. Interestingly, PeFLH NPY-SAP lesioned male rats had reduced body weights and decreased dark cycle feeding, but this effect was not seen in female rats. We conclude that a NPY projection to the PeFLH is necessary for glucoprivic feeding, but not locomotor activity, hyperglycemia, or corticosterone release, in both male and female rats.
Collapse
Affiliation(s)
- Pique P Choi
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Qing Wang
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Lynne A Brenner
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Ai-Jun Li
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Robert C Ritter
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Suzanne M Appleyard
- Neuroscience Program, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
16
|
Narai E, Watanabe T, Koba S. Hypothalamic Orexinergic Neurons Projecting to the Mesencephalic Locomotor Region Are Activated by Voluntary Wheel Running Exercise in Rats. Yonago Acta Med 2024; 67:52-60. [PMID: 38371276 PMCID: PMC10867236 DOI: 10.33160/yam.2024.02.006] [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: 11/21/2023] [Accepted: 12/18/2023] [Indexed: 02/20/2024]
Abstract
Background Cardiovascular changes during exercise are regulated by a motor volitional signal, called central command, which originates in the rostral portions of the brain and simultaneously regulates somatomotor and autonomic nervous systems. Whereas we recently elucidated mesencephalic locomotor region (MLR) neurons projecting to the rostral ventrolateral medulla as a crucial component of the central circuit responsible for transmitting central command signals, upstream circuits that regulate the MLR neurons remain unknown. Orexinergic neurons, which primarily originate from the perifornical area (PeFA) of the hypothalamus and reportedly play roles in eliciting locomotion and elevating sympathetic activity, send axonal projection to the MLR. The knowledge led us to investigate whether central command signals are relayed through orexinergic neurons projecting to the MLR. Methods We performed anterograde transsynaptic tagging with AAV1 encoding Cre to confirm the presence of MLR neurons postsynaptic to the PeFA in rats. We also conducted retrograde neural tracing with retrograde AAV, combined with immunohistochemical staining, to examine the excitability of MLR-projecting orexinergic neurons in rats that were allowed to freely run on the wheel for 90 min. Results A significant number of MLR neurons were labeled with Cre, indicating that PeFA neurons make synaptic contacts with MLR neurons. Moreover, immunoreactivities of Fos, a marker of neuronal excitation, were found in many MLR-projecting orexinergic neurons by voluntary wheel running exercise, compared to non-exercising control rats, especially in the intermediate-posterior, rather than anterior, and medial, rather than lateral, portions within the orexinergic neuron-distributing domain. Conclusion The findings suggest that specifically located orexinergic neurons transmit central command signals onto the MLR for running exercise. Elucidating the role of these MLR-projecting orexinergic neurons in somatomotor control and autonomic cardiovascular control deserves further study to unveil central circuit mechanisms responsible for central command function.
Collapse
Affiliation(s)
- Emi Narai
- Division of Integrative Physiology, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Tatsuo Watanabe
- Division of Integrative Physiology, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
| | - Satoshi Koba
- Division of Integrative Physiology, School of Medicine, Tottori University Faculty of Medicine, Yonago 683-8503, Japan
- Division of Veterinary Physiology, Tottori University Faculty of Agriculture, Tottori 680-8550, Japan
| |
Collapse
|
17
|
Chen KT, Huang MC, Lin C, Chang HM, Kao CF. GxE interaction effects of HCRTR2 single nucleotide polymorphism and adverse childhood experiences on methamphetamine use disorder. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2024; 50:84-94. [PMID: 38295363 DOI: 10.1080/00952990.2023.2297661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/18/2023] [Indexed: 02/02/2024]
Abstract
Background: Methamphetamine use disorder (MUD) is a worldwide health concern. The hypothalamic orexin system regulates stress response and addictive behaviors. The genetic variation in the hypocretin receptor 2 (HCRTR2), rs2653349, is associated with substance use disorder.Objectives: We explored the gene-environment (GxE) interaction of rs2653349 and adverse childhood experiences (ACEs) associated with MUD susceptibility.Methods: Four hundred and one individuals (336 males, 65 females) with MUD and 348 healthy controls (288 males, 60 females) completed a self-report questionnaire evaluating ACEs, encompassing childhood abuse and household dysfunction categories, and were genotyped for SNP rs2653349. Methamphetamine use variables were collected using the Diagnostic Interview for Genetic Studies. We used regression analyses to assess the GxE effect on MUD risk.Results: The MUD group had a comparable genotypic distribution for rs2653349 to the control group, albeit with a higher prevalence and number of types of ACEs, correlating with an increased MUD risk (p < .05). No significant genetic impact of rs2653349 on MUD risk was found. However, we observed a GxE interaction effect between the minor allele of rs2653349 and the number of childhood abuse or household dysfunction types, correlating with a reduced MUD risk (OR = -0.71, p = .04, Benjamini-Hochberg adjusted p = .08 and OR = -0.59, p = .045, Benjamini-Hochberg adjusted p = .09, respectively).Conclusion: HCRTR2 SNP rs2653349 has no significant impact on MUD risk, but ACEs may increase this risk. GxE results suggest that rs2653349 could offer protection against developing MUD in individuals experiencing multiple types of ACEs.
Collapse
Affiliation(s)
- Kai-Ting Chen
- Department of General Medicine, Taipei City Hospital, Taipei, Taiwan
| | - Ming-Chyi Huang
- Department of Psychiatry, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Psychiatric Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chun Lin
- Department of Psychiatry, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan
| | - Hu-Ming Chang
- Department of Psychiatry, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan
| | - Chung-Feng Kao
- Department of Agronomy, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung, Taiwan
- Advanced Plant and Food Crop Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
18
|
Chen Y, Li H, Kong T, Shan L, Hao L, Wang F. The low ratio of ghrelin in plasma and cerebrospinal fluid might be beneficial to sleep. Pharmacol Biochem Behav 2023; 233:173672. [PMID: 37944671 DOI: 10.1016/j.pbb.2023.173672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVE Ghrelin is physiologically important for maintaining sleep rhythm. Cigarette smoking has been demonstrated to significantly increase the risk of insufficient sleep by regulating ghrelin at the central and peripheral levels. No research has been published to study the relationship between active smoking and sleep via ghrelin level in cerebrospinal fluid (CSF). METHODS A total of 139 Chinese males were recruited and divided into active smokers (n = 77) and non-smokers (n = 62). The levels of CSF and plasma ghrelin were measured. The Pittsburgh Sleep Quality Index (PSQI) was used to evaluate sleep. RESULTS Non-smokers had lower PSQI scores (1.71 ± 1.93) than active smokers (3.70 ± 1.78). Non-smokers have significantly lower plasma ghrelin levels and lower plasma/CSF ghrelin ratio but higher CSF ghrelin than active smokers. Among non-smokers, plasma ghrelin levels were not correlated with PSQI scores (all p > 0.05), CSF ghrelin levels were positively correlated with PSQI scores (r = 0.309, p = 0.019), and the plasma/CSF ghrelin ratio was negatively correlated with PSQI scores (r = -0.346, p = 0.008). CONCLUSIONS This study is the first to reveal the relationship between cigarette smoking, high CSF ghrelin levels and insufficient sleep, suggesting that maintaining a normal plasma/CSF ghrelin ratio may be the physiological mechanism of healthy sleep, and the insufficient sleep population must quit smoking.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot 010110, China
| | - Hui Li
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Tiantian Kong
- Xinjiang Key Laboratory of Neurological Disorder Research, the Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, China
| | - Ligang Shan
- Department of Anesthesiology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen 361021, China
| | - Lei Hao
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot 010110, China.
| | - Fan Wang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing 100096, China.
| |
Collapse
|
19
|
Evans R, Kimura H, Nakashima M, Ishikawa T, Yukitake H, Suzuki M, Hazel J, Faessel H, Wu J, Hang Y, Alexander R, Rosen L, Hartman DS, Ratti E. Orexin 2 receptor-selective agonist danavorexton (TAK-925) promotes wakefulness in non-human primates and healthy individuals. J Sleep Res 2023; 32:e13878. [PMID: 36934366 DOI: 10.1111/jsr.13878] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/20/2023]
Abstract
The orexin 2 receptor-selective agonist danavorexton (TAK-925) has been shown to produce wake-promoting effects in wild-type mice, narcolepsy-model mice, and individuals with narcolepsy type 1 and type 2. Here, we report wake-promoting effects of danavorexton in non-human primates and healthy men during their sleep phase. Electroencephalogram analyses revealed that subcutaneous administration of danavorexton significantly increased wakefulness in common marmosets (p < 0.05 at 0.1 mg kg-1 , and p < 0.001 at 1 mg kg-1 and 10 mg kg-1 ) and cynomolgus monkeys (p ≤ 0.05 at 1 mg kg-1 and 3 mg kg-1 ). In a phase 1b crossover, randomized, double-blind, placebo-controlled and active-controlled study in sleep-deprived healthy participants (ClinicalTrials.gov identifier: NCT03522506), modafinil 300 mg (used to demonstrate assay sensitivity) and continuous infusion of danavorexton 44 mg and danavorexton 112 mg showed statistically superior wake-promoting effects to placebo (n = 18). Measured using the Maintenance of Wakefulness Test, mean (standard deviation) sleep latencies during infusion of danavorexton 44 mg, danavorexton 112 mg and placebo were 21.4 (8.9), 31.8 (3.2) and 9.2 (6.4) min, respectively. Least-squares mean difference from placebo in average sleep latency was 16.8 min with danavorexton 44 mg and 30.2 min with danavorexton 112 mg (both p < 0.001). Karolinska Sleepiness Scale scores were statistically significantly lower (indicating decreased sleepiness) for participants receiving danavorexton than for those receiving placebo during infusion (danavorexton 44 mg, p = 0.010; danavorexton 112 mg, p < 0.001). Together, these results indicate that an orexin 2 receptor agonist increases wakefulness in non-human primates and healthy individuals during their sleep phase.
Collapse
Affiliation(s)
- Rebecca Evans
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Haruhide Kimura
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Masato Nakashima
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Takashi Ishikawa
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Hiroshi Yukitake
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Motohisa Suzuki
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - James Hazel
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Hélène Faessel
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Jingtao Wu
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Yaming Hang
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Robert Alexander
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Laura Rosen
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Deborah S Hartman
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Emiliangelo Ratti
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
20
|
Hung C, Yamanaka A. The role of orexin neuron activity in sleep/wakefulness regulation. Peptides 2023; 165:171007. [PMID: 37030519 DOI: 10.1016/j.peptides.2023.171007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 04/10/2023]
Abstract
Orexin (also known as hypocretin) is a neuropeptide exclusively synthesized in the neurons of the lateral hypothalamus (LH). Initially orexin was thought to be involved in the regulation of feeding behavior. However, it is now known to also be a critical regulator of sleep/wakefulness, especially the maintenance of wakefulness. Although the somas of orexin neurons are exclusively located in the LH, these neurons send axons throughout the brain and spinal cord. Orexin neurons integrate inputs from various brain regions and project to neurons that are involved in the regulation of sleep/wakefulness. Orexin knockout mice have a fragmentation of sleep/wakefulness and cataplexy-like behavior arrest, which is similar to the sleep disorder narcolepsy. Recent progress with manipulation of neural activity of targeted neurons, using experimental tools such as optogenetics and chemogenetics, has emphasized the role of orexin neuron activity on the regulation of sleep/wakefulness. Recording of orexin neuron activity in vivo using electrophysiological and gene-encoded calcium indicator proteins revealed that these cells have specific activity patterns across sleep/wakefulness state changes. Here, we also discuss not only the role of the orexin peptide, but also the role of other co-transmitters that are synthesized and released from orexin neurons and involved in sleep/wakefulness regulation.
Collapse
Affiliation(s)
- Chijung Hung
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Akihiro Yamanaka
- Chinese Institute for Brain Research, Beijing (CIBR), Beijing, 102206, China; National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8585 Japan; Division of Brain Sciences Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| |
Collapse
|
21
|
Ten-Blanco M, Flores Á, Cristino L, Pereda-Pérez I, Berrendero F. Targeting the orexin/hypocretin system for the treatment of neuropsychiatric and neurodegenerative diseases: from animal to clinical studies. Front Neuroendocrinol 2023; 69:101066. [PMID: 37015302 DOI: 10.1016/j.yfrne.2023.101066] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/15/2023] [Accepted: 03/30/2023] [Indexed: 04/06/2023]
Abstract
Orexins (also known as hypocretins) are neuropeptides located exclusively in hypothalamic neurons that have extensive projections throughout the central nervous system and bind two different G protein-coupled receptors (OX1R and OX2R). Since its discovery in 1998, the orexin system has gained the interest of the scientific community as a potential therapeutic target for the treatment of different pathological conditions. Considering previous basic science research, a dual orexin receptor antagonist, suvorexant, was the first orexin agent to be approved by the US Food and Drug Administration to treat insomnia. In this review, we discuss and update the main preclinical and human studies involving the orexin system with several psychiatric and neurodegenerative diseases. This system constitutes a nice example of how basic scientific research driven by curiosity can be the best route to the generation of new and powerful pharmacological treatments.
Collapse
Affiliation(s)
- Marc Ten-Blanco
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - África Flores
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Neurosciences Institute, University of Barcelona and Bellvitge University Hospital-IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Luigia Cristino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Inmaculada Pereda-Pérez
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Fernando Berrendero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain.
| |
Collapse
|
22
|
Core body temperature varies according to the time of exercise without affecting orexin-A production in the dorsolateral hypothalamus in rat. J Therm Biol 2023. [DOI: 10.1016/j.jtherbio.2023.103522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
|
23
|
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
|
24
|
James MH, Aston-Jones G. Orexin Reserve: A Mechanistic Framework for the Role of Orexins (Hypocretins) in Addiction. Biol Psychiatry 2022; 92:836-844. [PMID: 36328706 PMCID: PMC10184826 DOI: 10.1016/j.biopsych.2022.06.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 11/22/2022]
Abstract
In 2014, we proposed that orexin signaling transformed motivationally relevant states into adaptive behavior directed toward exploiting an opportunity or managing a threat, a process we referred to as motivational activation. Advancements in animal models since then have permitted higher-resolution measurements of motivational states; in particular, the behavioral economics approach for studying drug demand characterizes conditions that lead to the enhanced motivation that underlies addiction. This motivational plasticity is paralleled by persistently increased orexin expression in a topographically specific manner-a finding confirmed across species, including in humans. Normalization of orexin levels also reduces drug motivation in addiction models. These new advancements lead us to update our proposed framework for the orexin function. We now propose that the capacity of orexin neurons to exhibit dynamic shifts in peptide production contributes to their role in adaptive motivational regulation and that this is achieved via a pool of reserve orexin neurons. This reserve is normally bidirectionally recruited to permit motivational plasticity that promotes flexible, adaptive behavior. In pathological states such as addiction, however, we propose that the orexin system loses capacity to adaptively adjust peptide production, resulting in focused hypermotivation for drug, driven by aberrantly and persistently high expression in the orexin reserve pool. This mechanistic framework has implications for the understanding and treatment of several psychiatric disorders beyond addiction, particularly those characterized by motivational dysfunction.
Collapse
Affiliation(s)
- Morgan H James
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, New Jersey; Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, New Jersey.
| | - Gary Aston-Jones
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, New Jersey; Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, New Jersey.
| |
Collapse
|
25
|
López JM, Carballeira P, Pozo J, León-Espinosa G, Muñoz A. Hypothalamic orexinergic neuron changes during the hibernation of the Syrian hamster. Front Neuroanat 2022; 16:993421. [PMID: 36157325 PMCID: PMC9501701 DOI: 10.3389/fnana.2022.993421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
Hibernation in small mammals is a highly regulated process with periods of torpor involving drops in body temperature and metabolic rate, as well as a general decrease in neural activity, all of which proceed alongside complex brain adaptive changes that appear to protect the brain from extreme hypoxia and low temperatures. All these changes are rapidly reversed, with no apparent brain damage occurring, during the short periods of arousal, interspersed during torpor—characterized by transitory and partial rewarming and activity, including sleep activation, and feeding in some species. The orexins are neuropeptides synthesized in hypothalamic neurons that project to multiple brain regions and are known to participate in the regulation of a variety of processes including feeding behavior, the sleep-wake cycle, and autonomic functions such as brown adipose tissue thermogenesis. Using multiple immunohistochemical techniques and quantitative analysis, we have characterized the orexinergic system in the brain of the Syrian hamster—a facultative hibernator. Our results revealed that orexinergic neurons in this species consisted of a neuronal population restricted to the lateral hypothalamic area, whereas orexinergic fibers distribute throughout the rostrocaudal extent of the brain, particularly innervating catecholaminergic and serotonergic neuronal populations. We characterized the changes of orexinergic cells in the different phases of hibernation based on the intensity of immunostaining for the neuronal activity marker C-Fos and orexin A (OXA). During torpor, we found an increase in C-Fos immunostaining intensity in orexinergic neurons, accompanied by a decrease in OXA immunostaining. These changes were accompanied by a volume reduction and a fragmentation of the Golgi apparatus (GA) as well as a decrease in the colocalization of OXA and the GA marker GM-130. Importantly, during arousal, C-Fos and OXA expression in orexinergic neurons was highest and the structural appearance and the volume of the GA along with the colocalization of OXA/GM-130 reverted to euthermic levels. We discuss the involvement of orexinergic cells in the regulation of mammalian hibernation and, in particular, the possibility that the high activation of orexinergic cells during the arousal stage guides the rewarming as well as the feeding and sleep behaviors characteristic of this phase.
Collapse
Affiliation(s)
- Jesús M. López
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
| | - Paula Carballeira
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
| | - Javier Pozo
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
| | - Gonzalo León-Espinosa
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-Centro de Estudios Universitarios (CEU), Madrid, Spain
| | - Alberto Muñoz
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica (CTB), Universidad Politécnica de Madrid, Madrid, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- *Correspondence: Alberto Muñoz,
| |
Collapse
|
26
|
Gao XB, Horvath TL. From Molecule to Behavior: Hypocretin/orexin Revisited From a Sex-dependent Perspective. Endocr Rev 2022; 43:743-760. [PMID: 34792130 PMCID: PMC9277634 DOI: 10.1210/endrev/bnab042] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Indexed: 11/19/2022]
Abstract
The hypocretin/orexin (Hcrt/Orx) system in the perifornical lateral hypothalamus has been recognized as a critical node in a complex network of neuronal systems controlling both physiology and behavior in vertebrates. Our understanding of the Hcrt/Orx system and its array of functions and actions has grown exponentially in merely 2 decades. This review will examine the latest progress in discerning the roles played by the Hcrt/Orx system in regulating homeostatic functions and in executing instinctive and learned behaviors. Furthermore, the gaps that currently exist in our knowledge of sex-related differences in this field of study are discussed.
Collapse
Affiliation(s)
- Xiao-Bing Gao
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
27
|
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: 22] [Impact Index Per Article: 7.3] [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
|
28
|
Xiang X, Chen Y, Li KX, Fang J, Bickler PE, Guan Z, Zhou W. Neuroanatomical Basis for the Orexinergic Modulation of Anesthesia Arousal and Pain Control. Front Cell Neurosci 2022; 16:891631. [PMID: 35558876 PMCID: PMC9090436 DOI: 10.3389/fncel.2022.891631] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/04/2022] [Indexed: 12/02/2022] Open
Abstract
Hypothalamic orexin (hypocretin) neurons play crucial roles in arousal control. Their involvement in anesthesia and analgesia remains to be better understood. In order to enhance our view on the neuroanatomy, we systematically mapped the projections of orexin neurons with confocal microscope and light sheet microscope. We specifically expressed optogenetic opsins tagged with fluorescence markers in orexin neurons through adeno-associated viral infection in the mouse brain. The imaging results revealed fine details and novel features of the orexin projections throughout the brain, particularly related to the nuclei regulating arousal and pain. We then optogenetically activated orexin neurons in the lateral hypothalamus to study the effects on anesthesia-related behaviors. cFos staining showed that optogenetic stimulation can activate orexin neurons in the ChR2-mCherry group, but not the control mCherry group (62.86 ± 3.923% vs. 7.9 ± 2.072%; P < 0.0001). In behavior assays, optogenetic stimulation in the ChR2-mCherry group consistently elicited robust arousal from light isoflurane anesthesia (9.429 ± 3.804 s vs. 238.2 ± 17.42 s; P < 0.0001), shortened the emergence time after deep isoflurane anesthesia (109.5 ± 13.59 s vs. 213.8 ± 21.77 s; P = 0.0023), and increased the paw withdrawal latency in a hotplate test (11.45 ± 1.185 s vs. 8.767 ± 0.7775; P = 0.0317). The structural details of orexin fibers established the neuroanatomic basis for studying the role of orexin in anesthesia and analgesia.
Collapse
Affiliation(s)
- Xuaner Xiang
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Yuzhang Chen
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Ke-Xin Li
- Department of Physiology, University of California, San Francisco, San Francisco, CA, United States
| | - Jianqiao Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Philip E. Bickler
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Wei Zhou
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
- *Correspondence: Wei Zhou,
| |
Collapse
|
29
|
Dale NC, Hoyer D, Jacobson LH, Pfleger KDG, Johnstone EKM. Orexin Signaling: A Complex, Multifaceted Process. Front Cell Neurosci 2022; 16:812359. [PMID: 35496914 PMCID: PMC9044999 DOI: 10.3389/fncel.2022.812359] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/07/2022] [Indexed: 11/15/2022] Open
Abstract
The orexin system comprises two G protein-coupled receptors, OX1 and OX2 receptors (OX1R and OX2R, respectively), along with two endogenous agonists cleaved from a common precursor (prepro-orexin), orexin-A (OX-A) and orexin-B (OX-B). For the receptors, a complex array of signaling behaviors has been reported. In particular, it becomes obvious that orexin receptor coupling is very diverse and can be tissue-, cell- and context-dependent. Here, the early signal transduction interactions of the orexin receptors will be discussed in depth, with particular emphasis on the direct G protein interactions of each receptor. In doing so, it is evident that ligands, additional receptor-protein interactions and cellular environment all play important roles in the G protein coupling profiles of the orexin receptors. This has potential implications for our understanding of the orexin system's function in vivo in both central and peripheral environments, as well as the development of novel agonists, antagonists and possibly allosteric modulators targeting the orexin system.
Collapse
Affiliation(s)
- Natasha C. Dale
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, WA, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, WA, Australia
| | - Daniel Hoyer
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Laura H. Jacobson
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Kevin D. G. Pfleger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, WA, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, WA, Australia
- Dimerix Limited, Nedlands, WA, Australia
| | - Elizabeth K. M. Johnstone
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, WA, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
30
|
Missig G, Mehta N, Robbins JO, Good CH, Iliopoulos-Tsoutsouvas C, Makriyannis A, Nikas SP, Bergman J, Carlezon WA, Paronis CA. Altered sleep during spontaneous cannabinoid withdrawal in male mice. Behav Pharmacol 2022; 33:195-205. [PMID: 35288510 PMCID: PMC8928162 DOI: 10.1097/fbp.0000000000000674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cessation of cannabinoid use in humans often leads to a withdrawal state that includes sleep disruption. Despite important health implications, little is known about how cannabinoid abstention affects sleep architecture, in part because spontaneous cannabinoid withdrawal is difficult to model in animals. In concurrent work we report that repeated administration of the high-efficacy cannabinoid 1 (CB1) receptor agonist AM2389 to mice for 5 days led to heightened locomotor activity and paw tremor following treatment discontinuation, potentially indicative of spontaneous cannabinoid withdrawal. Here, we performed parallel studies to examine effects on sleep. Using implantable electroencephalography (EEG) and electromyography (EMG) telemetry we examined sleep and neurophysiological measures before, during, and after 5 days of twice-daily AM2389 injections. We report that AM2389 produces decreases in locomotor activity that wane with repeated treatment, whereas discontinuation produces rebound increases in activity that persist for several days. Likewise, AM2389 initially produces profound increases in slow-wave sleep (SWS) and decreases in rapid eye movement (REM) sleep, as well as consolidation of sleep. By the third AM2389 treatment, this pattern transitions to decreases in SWS and total time sleeping. This pattern persists following AM2389 discontinuation and is accompanied by emergence of sleep fragmentation. Double-labeling immunohistochemistry for hypocretin/orexin (a sleep-regulating peptide) and c-Fos (a neuronal activity marker) in lateral hypothalamus revealed decreases in c-Fos/orexin+ cells following acute AM2389 and increases following discontinuation, aligning with the sleep changes. These findings indicate that AM2389 profoundly alters sleep in mice and suggest that sleep disruption following treatment cessation reflects spontaneous cannabinoid withdrawal.
Collapse
Affiliation(s)
- Galen Missig
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Niyati Mehta
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - James O. Robbins
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Cameron H. Good
- Neurolux Inc, Northfield, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
| | | | | | | | - Jack Bergman
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - William A. Carlezon
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Carol A. Paronis
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| |
Collapse
|
31
|
Gugula A, Trenk A, Celary A, Cizio K, Tylko G, Blasiak A, Hess G. Early-life stress modifies the reactivity of neurons in the ventral tegmental area and lateral hypothalamus to acute stress in female rats. Neuroscience 2022; 490:49-65. [DOI: 10.1016/j.neuroscience.2022.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/29/2022] [Accepted: 02/14/2022] [Indexed: 10/19/2022]
|
32
|
Agee LA, Nemchek V, Malone CA, Lee HJ, Monfils MH. Appetitive Behavior in the Social Transmission of Food Preference Paradigm Predicts Activation of Orexin-A producing Neurons in a Sex-Dependent Manner. Neuroscience 2022; 481:30-46. [PMID: 34843892 PMCID: PMC9246717 DOI: 10.1016/j.neuroscience.2021.11.032] [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: 06/12/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 11/22/2022]
Abstract
Orexin-producing cells in the lateral hypothalamic area have been shown to be involved in a wide variety of behavioral and cognitive functions, including the recall of appetitive associations and a variety of social behaviors. Here, we investigated the role of orexin in the acquisition and recall of socially transmitted food preferences in the rat. Rats were euthanized following either acquisition, short-term recall, or long-term recall of a socially transmitted food preference and their brains were processed for orexin-A and c-Fos expression. We found that while there were no significant differences in c-Fos expression between control and experimental subjects at any of the tested timepoints, females displayed significantly more activity in both orexinergic and non-orexinergic cells in the lateral hypothalamus. In the infralimbic cortex, we found that social behavior was significantly predictive of c-Fos expression, with social behaviors related to olfactory exploration appearing to be particularly influential. We additionally found that appetitive behavior was significantly predictive of orexin-A activity in a sex-dependent matter, with the total amount eaten correlating negatively with orexin-A/c-Fos colocalization in male rats but not female rats. These findings suggest a potential sex-specific role for the orexin system in balancing the stimulation of feeding behavior with the sleep/wake cycle.
Collapse
Affiliation(s)
- Laura A Agee
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA
| | - Victoria Nemchek
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA
| | - Cassidy A Malone
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA
| | - Hongjoo J Lee
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA; Institute for Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - Marie-H Monfils
- The University of Texas at Austin, Department of Psychology, Austin, TX, USA; Institute for Neuroscience, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
33
|
Miyata K, Ikoma Y, Murata K, Kusumoto-Yoshida I, Kobayashi K, Kuwaki T, Ootsuka Y. Multifaceted roles of orexin neurons in mediating methamphetamine-induced changes in body temperature and heart rate. IBRO Neurosci Rep 2022; 12:108-120. [PMID: 35128515 PMCID: PMC8804267 DOI: 10.1016/j.ibneur.2022.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 11/26/2022] Open
|
34
|
Parameswaran G, Ray DW. Sleep, circadian rhythms, and type 2 diabetes mellitus. Clin Endocrinol (Oxf) 2022; 96:12-20. [PMID: 34637144 PMCID: PMC8939263 DOI: 10.1111/cen.14607] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 01/01/2023]
Abstract
Over the last 60 years we have seen a significant rise in metabolic disease, especially type 2 diabetes. In the same period, the emergence of electricity and artificial lighting has allowed our behavioural cycles to be independent of external patterns of sunlight. This has led to a corresponding increase in sleep deprivation, estimated to be about 1 hour per night, as well as circadian misalignment (living against the clock). Evidence from experimental animals as well as controlled human subjects have shown that sleep deprivation and circadian misalignment can both directly drive metabolic dysfunction, causing diabetes. However, the precise mechanism by which these processes contribute to insulin resistance remains poorly understood. In this article, we will review the new literature in the field and propose a model attempting to reconcile the experimental observations made. We believe our model will serve as a useful point of reference to understand how metabolic dysfunction can emerge from sleep or circadian rhythm disruptions, providing new directions for research and therapy.
Collapse
Affiliation(s)
- Gokul Parameswaran
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordOxfordUK
- NIHR Oxford Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
| | - David W. Ray
- Oxford Centre for Diabetes, Endocrinology and MetabolismUniversity of OxfordOxfordUK
- NIHR Oxford Biomedical Research CentreJohn Radcliffe HospitalOxfordUK
| |
Collapse
|
35
|
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
|
36
|
Sleep dysregulation in binge eating disorder and "food addiction": the orexin (hypocretin) system as a potential neurobiological link. Neuropsychopharmacology 2021; 46:2051-2061. [PMID: 34145404 PMCID: PMC8505614 DOI: 10.1038/s41386-021-01052-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022]
Abstract
It has been proposed that binge eating reflects a pathological compulsion driven by the "addictive" properties of foods. Proponents of this argument highlight the large degree of phenomenological and diagnostic overlap between binge eating disorder (BED) and substance use disorders (SUDs), including loss of control over how much is consumed and repeated unsuccessful attempts to abstain from consumption, as well as commonalities in brain structures involved in food and drug craving. To date, very little attention has been given to an additional behavioral symptom that BED shares with SUDs-sleep dysregulation-and the extent to which this may contribute to the pathophysiology of BED. Here, we review studies examining sleep outcomes in patients with BED, which collectively point to a heightened incidence of sleep abnormalities in BED. We identify the orexin (hypocretin) system as a potential neurobiological link between compulsive eating and sleep dysregulation in BED, and provide a comprehensive update on the evidence linking this system to these processes. Finally, drawing on evidence from the SUD literature indicating that the orexin system exhibits significant plasticity in response to drugs of abuse, we hypothesize that chronic palatable food consumption likewise increases orexin system activity, resulting in dysregulated sleep/wake patterns. Poor sleep, in turn, is predicted to exacerbate binge eating, contributing to a cycle of uncontrolled food consumption. By extension, we suggest that pharmacotherapies normalizing orexin signaling, which are currently being trialed for the treatment of SUDs, might also have utility in the clinical management of BED.
Collapse
|
37
|
Bumgarner JR, Walker WH, Nelson RJ. Circadian rhythms and pain. Neurosci Biobehav Rev 2021; 129:296-306. [PMID: 34375675 PMCID: PMC8429267 DOI: 10.1016/j.neubiorev.2021.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 06/30/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
The goal of this review is to provide a perspective on the nature and importance of the relationship between the circadian and pain systems. We provide: 1) An overview of the circadian and pain systems, 2) a review of direct and correlative evidence that demonstrates diurnal and circadian rhythms within the pain system; 3) a perspective highlighting the need to consider the role of a proposed feedback loop of circadian rhythm disruption and maladaptive pain; 4) a perspective on the nature of the relationship between circadian rhythms and pain. In summary, we propose that there is no single locus responsible for producing the circadian rhythms of the pain system. Instead, circadian rhythms of pain are a complex result of the distributed rhythms present throughout the pain system, especially those of the descending pain modulatory system, and the rhythms of the systems with which it interacts, including the opioid, endocrine, and immune systems.
Collapse
Affiliation(s)
- Jacob R Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA.
| | - William H Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
38
|
Azeez IA, Igado OO, Olopade JO. An overview of the orexinergic system in different animal species. Metab Brain Dis 2021; 36:1419-1444. [PMID: 34224065 DOI: 10.1007/s11011-021-00761-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 06/06/2021] [Indexed: 01/13/2023]
Abstract
Orexin (hypocretin), is a neuropeptide produced by a subset of neurons in the lateral hypothalamus. From the lateral hypothalamus, the orexin-containing neurons project their fibres extensively to other brain structures, and the spinal cord constituting the central orexinergic system. Generally, the term ''orexinergic system'' usually refers to the orexin peptides and their receptors, as well as to the orexin neurons and their projections to different parts of the central nervous system. The extensive networks of orexin axonal fibres and their terminals allow these neuropeptidergic neurons to exert great influence on their target regions. The hypothalamic neurons containing the orexin neuropeptides have been implicated in diverse functions, especially related to the control of a variety of homeostatic functions including feeding behaviour, arousal, wakefulness stability and energy expenditure. The broad range of functions regulated by the orexinergic system has led to its description as ''physiological integrator''. In the last two decades, the orexinergic system has been a topic of great interest to the scientific community with many reports in the public domain. From the documentations, variations exist in the neuroanatomical profile of the orexinergic neuron soma, fibres and their receptors from animal to animal. Hence, this review highlights the distinct variabilities in the morphophysiological aspects of the orexinergic system in the vertebrate animals, mammals and non-mammals, its presence in other brain-related structures, including its involvement in ageing and neurodegenerative diseases. The presence of the neuropeptide in the cerebrospinal fluid and peripheral tissues, as well as its alteration in different animal models and conditions are also reviewed.
Collapse
Affiliation(s)
- Idris A Azeez
- Department of Veterinary Anatomy, University of Jos, Jos, Nigeria
| | - Olumayowa O Igado
- Department of Veterinary Anatomy, University of Ibadan, Ibadan, Nigeria
| | - James O Olopade
- Department of Veterinary Anatomy, University of Ibadan, Ibadan, Nigeria.
| |
Collapse
|
39
|
Koekkoek LL, Masís-Vargas A, Kool T, Eggels L, van der Gun LL, Lamuadni K, Slomp M, Diepenbroek C, Kalsbeek A, la Fleur SE. Sucrose drinking mimics effects of nucleus accumbens µ-opioid receptor stimulation on fat intake and brain c-Fos-expression. Nutr Neurosci 2021; 25:2408-2420. [PMID: 34490827 DOI: 10.1080/1028415x.2021.1975365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Objectives: We have previously shown that the combined consumption of fat and a sucrose solution induces overeating, and there is evidence indicating that sucrose drinking directly stimulates fat intake. One neurochemical pathway by which sucrose may enhance fat intake is through the release of endogenous opioids in the nucleus accumbens (NAC).Methods: To test this hypothesis, we provided rats with a free-choice high-fat diet for two weeks. During the second week, rats had access to an additional bottle of water or a 30% sucrose solution for five minutes per day. After these two weeks, we infused vehicle or the μ-opioid receptor agonist [D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) into the NAC 30 min after their daily access to the additional bottle of water or the sucrose solution.Results: Sucrose drinking had two effects, (1) it stimulated fat intake in the absence of DAMGO infusion, (2) it diminished sensitivity to DAMGO, as it prevented the rapid increase in fat intake typically seen upon DAMGO infusion in the nucleus accumbens. In a second experiment, we confirmed that these results are not due to the ingested calories of the sucrose solution. Lastly, we investigated which brain areas are involved in the observed effects on fat intake by assessing c-Fos-expression in brain areas previously linked to DAMGO's effects on food intake. Both intra-NAC DAMGO infusion and sucrose consumption in the absence of DAMGO infusion had no effect on c-Fos-expression in orexin neurons and the central amygdala but increased c-Fos-expression in the NAC as well as the basolateral amygdala.Discussion: In conclusion, we confirm that sucrose drinking stimulates fat intake, likely through the release of endogenous opioids.
Collapse
Affiliation(s)
- L L Koekkoek
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Metabolism and Reward Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - A Masís-Vargas
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Metabolism and Reward Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - T Kool
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Metabolism and Reward Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - L Eggels
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Metabolism and Reward Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - L L van der Gun
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Metabolism and Reward Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - K Lamuadni
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Metabolism and Reward Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - M Slomp
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Metabolism and Reward Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - C Diepenbroek
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Metabolism and Reward Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - A Kalsbeek
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - S E la Fleur
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Neuroscience Amsterdam, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam, The Netherlands.,Metabolism and Reward Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Hopkins K, Mukherjee S, Ponce D, Mangum J, Jacobson LH, Hoyer D. Development of a LC-ESI-MRM method for the absolute quantification of orexin A in the CSF of individual mice. MEDICINE IN DRUG DISCOVERY 2021. [DOI: 10.1016/j.medidd.2021.100102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
41
|
Dong Z, Huang B, Jiang C, Chen J, Lin H, Lian Q, Wu B. The Adenosine A2A Receptor Activation in Nucleus Accumbens Suppress Cue-Induced Reinstatement of Propofol Self-administration in Rats. Neurochem Res 2021; 46:1081-1091. [PMID: 33616808 PMCID: PMC8053194 DOI: 10.1007/s11064-021-03238-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/30/2020] [Accepted: 01/06/2021] [Indexed: 01/04/2023]
Abstract
Propofol has shown strong addictive properties in rats and humans. Adenosine A2A receptors (A2AR) in the nucleus accumbens (NAc) modulate dopamine signal and addictive behaviors such as cocaine- and amphetamine-induced self-administration. However, whether A2AR can modulate propofol addiction remains unknown. AAV-shA2AR was intra-NAc injected 3 weeks before the propofol self-administration training to test the impacts of NAc A2AR on establishing the self-administration model with fixed ratio 1 (FR1) schedule. Thereafter, the rats were withdrawal from propofol for 14 days and tested cue-induced reinstatement of propofol seeking behavior on day 15. The propofol withdrawal rats received one of the doses of CGS21680 (A2AR agonist, 2.5-10.0 ng/site), MSX-3 (A2AR antagonist, 5.0-20.0 μg/site) or eticlopride (D2 receptor (D2R) antagonist, 0.75-3.0 μg/site) or vehicle via intra-NAc injection before relapse behavior test. The numbers of active and inactive nose-poke response were recorded. Focal knockdown A2AR by shA2AR did not affect the acquisition of propofol self-administration behavior, but enhance cue-induced reinstatement of propofol self-administration compared with the AAV-shCTRLgroup. Pharmacological activation of the A2AR by CGS21680 (≥ 5.0 ng/site) attenuated cue-induced reinstatement of propofol self-administration behavior. Similarly, pharmacological blockade of D2R by eticlopride (0.75-3.0 μg/site) attenuated propofol seeking behavior. These effects were reversed by the administration of MSX-3 (5.0-20.0 μg/site). The A2AR- and D2R-mediated effects on propofol relapse were not confounded by the learning process, and motor activity as the sucrose self-administration and locomotor activity were not affected by all the treatments. This study provides genetic and pharmacological evidence that NAc A2AR activation suppresses cue-induced propofol relapse in rats, possibly by interacting with D2R.
Collapse
Affiliation(s)
- Zhanglei Dong
- Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, 325027, Zhejiang, China
| | - Bingwu Huang
- Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, 325027, Zhejiang, China
| | - Chenchen Jiang
- Clinical Research Unit, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiangfan Chen
- Molecular Neuropharmacology Laboratory, Wenzhou Medical University, Wenzhou, China
| | - Han Lin
- Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, 325027, Zhejiang, China
| | - Qingquan Lian
- Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, 325027, Zhejiang, China.
| | - Binbin Wu
- Department of Anesthesiology, Perioperative and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, 325027, Zhejiang, China.
| |
Collapse
|
42
|
Bjorness TE, Greene RW. Interaction between cocaine use and sleep behavior: A comprehensive review of cocaine's disrupting influence on sleep behavior and sleep disruptions influence on reward seeking. Pharmacol Biochem Behav 2021; 206:173194. [PMID: 33940055 DOI: 10.1016/j.pbb.2021.173194] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 04/20/2021] [Accepted: 04/27/2021] [Indexed: 12/21/2022]
Abstract
Dopamine, orexin (hypocretin), and adenosine systems have dual roles in reward and sleep/arousal suggesting possible mechanisms whereby drugs of abuse may influence both reward and sleep/arousal. While considerable variability exists across studies, drugs of abuse such as cocaine induce an acute sleep loss followed by an immediate recovery pattern that is consistent with a normal response to loss of sleep. Under more chronic cocaine exposure conditions, an abnormal recovery pattern is expressed that includes a retention of sleep disturbance under withdrawal and into abstinence conditions. Conversely, experimentally induced sleep disturbance can increase cocaine seeking. Thus, complementary, sleep-related therapeutic approaches may deserve further consideration along with development of non-human models to better characterize sleep disturbance-reward seeking interactions across drug experience.
Collapse
Affiliation(s)
- Theresa E Bjorness
- Research Service, VA North Texas Health Care System, Dallas, TX 75126, USA; Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Robert W Greene
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, 305-8577, Japan
| |
Collapse
|
43
|
Lonstein JS, Linning-Duffy K, Tang Y, Moody A, Yan L. Impact of daytime light intensity on the central orexin (hypocretin) system of a diurnal rodent (Arvicanthis niloticus). Eur J Neurosci 2021; 54:4167-4181. [PMID: 33899987 DOI: 10.1111/ejn.15248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 11/29/2022]
Abstract
The neuropeptide orexin/hypocretin is implicated in sleep and arousal, energy expenditure, reward, affective state and cognition. Our previous work using diurnal Nile grass rats (Arvicanthis niloticus) found that orexin mediates the effects of environmental light, particularly daytime light intensity, on affective and cognitive behaviours. The present study further investigated how daytime light intensity affects the central orexin system in male and female grass rats. Subjects were housed for 4 weeks in 12:12 hr dim light:dark (50 lux, dimLD) or in 12:12 hr bright light:dark cycle (1000 lux, brightLD). Day/night fluctuations in some orexin measures were also assessed. Despite similar hypothalamic prepro-orexin mRNA expression across all conditions, there were significantly more orexin-immunoreactive neurons, larger somata, greater optical density or higher orexin A content at night (ZT14) than during the day (ZT2), and/or in animals housed in brightLD compared to dimLD. Grass rats in brightLD also had higher cisternal CSF levels of orexin A. Furthermore, orexin receptor OX1R and OX2R proteins in the medial prefrontal cortex were higher in brightLD than dimLD males, but lower in brightLD than dimLD females. In the CA1 and dorsal raphe nucleus, females had higher OX1R than males without any significant effects of light condition, and OX2R levels were unaffected by sex or light. These results reveal that daytime light intensity alters the central orexin system of both male and female diurnal grass rats, sometimes sex-specifically, and provides insight into the mechanisms underlying how daytime light intensity impacts orexin-regulated functions.
Collapse
Affiliation(s)
- Joseph S Lonstein
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Katrina Linning-Duffy
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Yuping Tang
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Anna Moody
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Lily Yan
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
44
|
Ahmadi-Soleimani SM, Azizi H, Abbasi-Mazar A. Intermittent REM sleep deprivation attenuates the development of morphine tolerance and dependence in male rats. Neurosci Lett 2021; 748:135735. [PMID: 33592307 DOI: 10.1016/j.neulet.2021.135735] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 02/04/2023]
Abstract
Opioid agonists are used in clinic for pain management, however this application is challenged by development of tolerance and dependence following prolonged exposure. Various approaches have been suggested to address this concern, however, there is still no consensus among the researchers. Neural processing of sleep and nociception are co-regulated through shared brain regions having bidirectional interplays. Thus, we aimed to investigate whether application of REM sleep deprivation (REM-SD) could affect morphine analgesic tolerance and dependence. To this end, adult male rats underwent sleep deprivation during light and dark phases (LSD and DSD, respectively) using the inverted flower pot method and then tolerance and dependence was induced by repeated injection of morphine for 7 days (10 mg/kg, daily, i.p.). Results indicated that REM-SD delays the development of tolerance to morphine during both phases; however this effect was more potent following LSD. Moreover, LSD decreased the baseline thermal threshold and total withdrawal score. One possible hypothesis for our observations is REM-SD-induced attenuation of orexin system which is still controversial among the researchers. Other stronger possibilities might be down-regulation of opioid receptors in response to sleep loss experience. Finally, it seems that modification of sleep periods may assist to decrease the severity of opioid tolerance and dependence.
Collapse
Affiliation(s)
- S Mohammad Ahmadi-Soleimani
- Deparment of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Alireza Abbasi-Mazar
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| |
Collapse
|
45
|
Tan Y, Hang F, Liu ZW, Stoiljkovic M, Wu M, Tu Y, Han W, Lee AM, Kelley C, Hajós M, Lu L, de Lecea L, De Araujo I, Picciotto MR, Horvath TL, Gao XB. Impaired hypocretin/orexin system alters responses to salient stimuli in obese male mice. J Clin Invest 2021; 130:4985-4998. [PMID: 32516139 DOI: 10.1172/jci130889] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 06/03/2020] [Indexed: 12/27/2022] Open
Abstract
The brain has evolved in an environment where food sources are scarce, and foraging for food is one of the major challenges for survival of the individual and species. Basic and clinical studies show that obesity or overnutrition leads to overwhelming changes in the brain in animals and humans. However, the exact mechanisms underlying the consequences of excessive energy intake are not well understood. Neurons expressing the neuropeptide hypocretin/orexin (Hcrt) in the lateral/perifonical hypothalamus (LH) are critical for homeostatic regulation, reward seeking, stress response, and cognitive functions. In this study, we examined adaptations in Hcrt cells regulating behavioral responses to salient stimuli in diet-induced obese mice. Our results demonstrated changes in primary cilia, synaptic transmission and plasticity, cellular responses to neurotransmitters necessary for reward seeking, and stress responses in Hcrt neurons from obese mice. Activities of neuronal networks in the LH and hippocampus were impaired as a result of decreased hypocretinergic function. The weakened Hcrt system decreased reward seeking while altering responses to acute stress (stress-coping strategy), which were reversed by selectively activating Hcrt cells with chemogenetics. Taken together, our data suggest that a deficiency in Hcrt signaling may be a common cause of behavioral changes (such as lowered arousal, weakened reward seeking, and altered stress response) in obese animals.
Collapse
Affiliation(s)
- Ying Tan
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Fu Hang
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Guangxi Reproductive Medical Research Center, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhong-Wu Liu
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Milan Stoiljkovic
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Mingxing Wu
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Ophthalmology, Second Affiliate Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Tu
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Traditional Chinese Medicine Health Preservation, Second Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wenfei Han
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Angela M Lee
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Craig Kelley
- Joint Biomedical Engineering Program, SUNY Downstate and NYU Tandon, Brooklyn, New York, USA
| | - Mihály Hajós
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Ivan De Araujo
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Xiao-Bing Gao
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
46
|
Shankar A, Williams CT. The darkness and the light: diurnal rodent models for seasonal affective disorder. Dis Model Mech 2021; 14:dmm047217. [PMID: 33735098 PMCID: PMC7859703 DOI: 10.1242/dmm.047217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The development of animal models is a critical step for exploring the underlying pathophysiological mechanisms of major affective disorders and for evaluating potential therapeutic approaches. Although most neuropsychiatric research is performed on nocturnal rodents, differences in how diurnal and nocturnal animals respond to changing photoperiods, combined with a possible link between circadian rhythm disruption and affective disorders, has led to a call for the development of diurnal animal models. The need for diurnal models is most clear for seasonal affective disorder (SAD), a widespread recurrent depressive disorder that is linked to exposure to short photoperiods. Here, we briefly review what is known regarding the etiology of SAD and then examine progress in developing appropriate diurnal rodent models. Although circadian disruption is often invoked as a key contributor to SAD, a mechanistic understanding of how misalignment between endogenous circadian physiology and daily environmental rhythms affects mood is lacking. Diurnal rodents show promise as models of SAD, as changes in affective-like behaviors are induced in response to short photoperiods or dim-light conditions, and symptoms can be ameliorated by brief exposure to intervals of bright light coincident with activity onset. One exciting avenue of research involves the orexinergic system, which regulates functions that are disturbed in SAD, including sleep cycles, the reward system, feeding behavior, monoaminergic neurotransmission and hippocampal neurogenesis. However, although diurnal models make intuitive sense for the study of SAD and are more likely to mimic circadian disruption, their utility is currently hampered by a lack of genomic resources needed for the molecular interrogation of potential mechanisms.
Collapse
Affiliation(s)
- Anusha Shankar
- Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK 99775, USA
| | - Cory T Williams
- Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK 99775, USA
| |
Collapse
|
47
|
Sagi D, de Lecea L, Appelbaum L. Heterogeneity of Hypocretin/Orexin Neurons. FRONTIERS OF NEUROLOGY AND NEUROSCIENCE 2021; 45:61-74. [PMID: 34052814 PMCID: PMC8961008 DOI: 10.1159/000514964] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 02/02/2021] [Indexed: 01/21/2023]
Abstract
The multifunctional, hypothalamic hypocretin/orexin (HCRT)-producing neurons regulate an array of physiological and behavioral states including arousal, sleep, feeding, emotions, stress, and reward. How a presumably uniform HCRT neuron population regulates such a diverse set of functions is not clear. The role of the HCRT neuropeptides may vary depending on the timing and localization of secretion and neuronal activity. Moreover, HCRT neuropeptides may not mediate all functions ascribed to HCRT neurons. Some could be orchestrated by additional neurotransmitters and neuropeptides that are expressed in HCRT neurons. We hypothesize that HCRT neurons are segregated into genetically, anatomically and functionally distinct subpopulations. We discuss accumulating data that suggest the existence of such HCRT neuron subpopulations that may effectuate the diverse functions of these neurons in mammals and fish.
Collapse
Affiliation(s)
- Dana Sagi
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Luis de Lecea
- Dept of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Lior Appelbaum
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel.,Corresponding author: Lior Appelbaum, Bar-Ilan University, Ramat-Gan 5290002, Israel. Telephone: +972-3-7384536,
| |
Collapse
|
48
|
Morphine-Conditioned Placebo Analgesia in Female and Male Rats with Chronic Neuropathic Pain: c-Fos Expression in the Rostral Ventromedial Medulla. Neuroscience 2020; 457:51-73. [PMID: 33285237 DOI: 10.1016/j.neuroscience.2020.11.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/17/2022]
Abstract
Placebo analgesia has great potential to overcome the inadequacies of current drug therapies to treat conditions of chronic pain. The rostral ventromedial medulla (RVM) has been implicated as a critical relay in the antinociceptive pathway underpinning placebo analgesia in humans. We developed a model of opiate-conditioned placebo analgesia in rats with neuropathic injury to identify medullary nuclei active during placebo analgesia. Using female and male rats the degree of thermal allodynia was first determined following nerve injury, and a pharmacological conditioning procedure, pairing contextual cues with the experience of morphine-induced analgesia, was used to elicit placebo analgesic reactions. This protocol revealed clear subpopulations of placebo reactors (36% of males, 25% of females) and non-reactors in proportions similar to those reported in human studies. We detected injury-specific c-Fos expression in the gracile nucleus and morphine-specific c-Fos expression in the serotonergic midline raphe nuclei and the caudal nuclei of the solitary tract. However, c-Fos expression did not differ between placebo reactors and non-reactors in either serotonergic or non-serotonergic neurons of the RVM. Despite a subpopulation of rats demonstrating placebo reactions, we found no evidence for enhanced activity in the nuclei from which the classical RVM → spinal cord descending analgesic pathways emerge.
Collapse
|
49
|
Morales-Mulia S, Magdaleno-Madrigal VM, Nicolini H, Genis-Mendoza A, Morales-Mulia M. Orexin-A up-regulates dopamine D2 receptor and mRNA in the nucleus accumbens Shell. Mol Biol Rep 2020; 47:9689-9697. [PMID: 33170427 DOI: 10.1007/s11033-020-05979-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 11/03/2020] [Indexed: 01/11/2023]
Abstract
Orexins-A (OrxA) and -B (OrxB) neuropeptides are synthesized by a group of neurons located in the lateral hypothalamus and adjacent perifornical area, which send their projections to the mesolimbic dopaminergic (DAergic) system including ventral tegmental area and nucleus accumbens (NAc), where orexin receptors are expressed. NAc plays a central role in reward-seeking behavior and drug abuse. NAc-neurons express dopamine-1 (D1R) and dopamine-2 (D2R) receptors. Orexins bind to their two cognate G-protein-coupled receptors, orexin-receptor type-1 (Orx1R) and type-2 (Orx2R). Orexin receptor signaling is involved in behaviors such as motivation and addiction. Orexin-containing neurons modulate DAergic activity that is key in synaptic plasticity induced by addictive drugs. However, the effect of OrxA on expression and content of DAergic receptors in NAc is unknown. The purpose of this study was to investigate whether OrxA can alter gene expression and protein levels of D1R/D2R in NAc. Gene expression was evaluated by real-time PCR analysis and protein levels by western blot in rats. The results show that intracerebroventricular (i.c.v.) injection of OrxA increases both gene transcription and protein content of D2R but fails to modify D1R. This effect was also confirmed with OrxA infusion in NAc/Shell. Our results demonstrate for the first time that OrxA induces up-regulation of gene and protein of D2R in NAc. These findings support the hypothesis that OrxA modulates the DAergic transmission and this may serve to understand how orexin signaling enhances DA responses at baseline conditions and in response to psychostimulants.
Collapse
Affiliation(s)
- Sandra Morales-Mulia
- Departamento de Biología Celular, Facultad de Ciencias, UNAM, Mexico, DF, Mexico
| | | | - Humberto Nicolini
- Laboratory of Genomics of Psychiatric Diseases, Neurodegenerative and Addictions, National Institute of Genomic Medicine, Mexico, Mexico
| | - Alma Genis-Mendoza
- Laboratory of Genomics of Psychiatric Diseases, Neurodegenerative and Addictions, National Institute of Genomic Medicine, Mexico, Mexico.,Hospital Psiquiátrico Infantil "Dr. Juan N. Navarro" Psychiatric Attention Services, Mexico, Mexico
| | - Marcela Morales-Mulia
- Bases Moleculares de las Adicciones, Subdirección de Investigaciones Clínicas, INPRFM, Calzada México-Xochimilco 101, San Lorenzo Huipulco, Tlalpan, 14370, Mexico, DF, Mexico.
| |
Collapse
|
50
|
Tiseo C, Vacca A, Felbush A, Filimonova T, Gai A, Glazyrina T, Hubalek IA, Marchenko Y, Overeem LH, Piroso S, Tkachev A, Martelletti P, Sacco S. Migraine and sleep disorders: a systematic review. J Headache Pain 2020; 21:126. [PMID: 33109076 PMCID: PMC7590682 DOI: 10.1186/s10194-020-01192-5] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
Migraine and sleep disorders are common and often burdensome chronic conditions with a high prevalence in the general population, and with considerable socio-economic impact and costs.The existence of a relationship between migraine and sleep disorders has been recognized from centuries by clinicians and epidemiological studies. Nevertheless, the exact nature of this association, the underlying mechanisms and interactions are complex and not completely understood. Recent biochemical and functional imaging studies identified central nervous system structures and neurotransmitters involved in the pathophysiology of migraine and also important for the regulation of normal sleep architecture, suggesting a possible causative role, in the pathogenesis of both disorders, of a dysregulation in these common nervous system pathways.This systematic review summarizes the existing data on migraine and sleep disorders with the aim to evaluate the existence of a causal relationship and to assess the presence of influencing factors. The identification of specific sleep disorders associated with migraine should induce clinicians to systematically assess their presence in migraine patients and to adopt combined treatment strategies.
Collapse
Affiliation(s)
- Cindy Tiseo
- Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, L'Aquila, Italy
- Regional Referral Headache Centre, S.S. Filippo e Nicola Hospital, Avezzano, L'Aquila, Italy
| | - Alessandro Vacca
- Headache Center, Department of Neuroscience "Rita Levi Montalcini", University of Torino, Torino, Italy
| | - Anton Felbush
- Pain Treatment Center, OOO "Vertebra", Samara City, Russia
| | - Tamara Filimonova
- Federal State Budget Educational Institution of Higher Education "Academician Ye. A. Vagner Perm State Medical University" of the Ministry of Healthcare of the Russian Federation, Perm, Russia
| | - Annalisa Gai
- Headache Center, Department of Neuroscience "Rita Levi Montalcini", University of Torino, Torino, Italy
| | | | - Irina Anna Hubalek
- Department of Neurology, Headache Center, Charité University Medicine Berlin, Berlin, Germany
| | - Yelena Marchenko
- V. A. Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - Lucas Hendrik Overeem
- Charité - Universitätsmedizin Berlin Charité Centrum Neurologie, Neurochirurgie und Psychiatrie CC, Berlin, Germany
| | - Serena Piroso
- Department of Human Neurosciences, Sapienza University of Rome, Roma, Italy
| | - Alexander Tkachev
- Department of Neurology, Neurosurgery, medical genetics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Paolo Martelletti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Roma, Italy
- Regional Referral Headache Centre, Sant'Andrea Hospital, Rome, Italy
| | - Simona Sacco
- Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, L'Aquila, Italy.
- Regional Referral Headache Centre, S.S. Filippo e Nicola Hospital, Avezzano, L'Aquila, Italy.
| |
Collapse
|