1
|
Sarfi M, Elahdadi Salmani M, Lashkarbolouki T, Goudarzi I. Divergent effects of noradrenergic activation and orexin receptor 1 blockade on hippocampal structure, anxiety-like behavior, and social interaction following chronic stress. Pharmacol Biochem Behav 2025; 250:173997. [PMID: 40073949 DOI: 10.1016/j.pbb.2025.173997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Chronic stress (Ch.S) has detrimental effects on the brain's structure and function, particularly in the hippocampus. The noradrenergic and orexinergic systems play crucial roles in the stress response and regulation of stress-related behaviors. This study aimed to investigate the interaction between noradrenergic activation and orexin receptor 1 inhibition on chronic stress-induced hippocampal alterations. The study conducted experiments on male Wistar rats, subjected to Ch.S, OXr1 blocking, noradrenergic activation, or a combination of these treatments. Plasma corticosterone level was measured using a fluorometric method. Behavioral assessment of social maze, elevated plus maze (EPM) and novel object recognition (NOR) test were performed. Then, the expression of prepro-orexin, OXr1, and glucocorticoid receptor (GR) was analyzed using semiquantitative RT-PCR. Neuronal populations were quantified through Nissl staining. The data revealed that all stress and yohimbine groups had elevated plasma corticosterone levels. Ch.S significantly altered behavior, impairing social interaction, disrupting object recognition memory and increasing anxiety-like responses in the EPM. OXr1 blocking reversed these stress-induced behavioral deficits, while yohimbine did not improve these behavioral outcomes. Chronic stress led to a significant increase in prepro-orexin, OXr1, and GR expression. While blocking OXr1 helped counteract these stress-induced changes, yohimbine failed to restore the expression levels. Ch.S reduced hippocampal neuronal populations, while OXr1 blocking partially reversed this effect, and yohimbine further recovered the reversal. These findings indicate that blocking hippocampal OXr1 can mitigate the adverse effects of chronic stress on both hippocampal structure and anxiety-like behaviors, while noradrenergic signaling appears to have differential effects on behavioral and cellular measures.
Collapse
Affiliation(s)
| | | | | | - Iran Goudarzi
- School of Biology, Damghan University, Damghan, Iran.
| |
Collapse
|
2
|
Mariani S, Honisch C, Ruzza P, Tartaggia S. Unexpected Conversion of Tyrosine into a Coumaric Acid Residue at the N-Terminal Side of an Orexin Peptide Fragment Induced by UV Irradiation. Chemistry 2025; 31:e202500383. [PMID: 40019305 DOI: 10.1002/chem.202500383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 03/01/2025]
Abstract
The presence of reactive species exceeding the physiological self-defense mechanisms in cells and tissues, known as oxidative stress, usually leads to damage of DNA and proteins. In this work, we analyzed the impact of UV irradiation and radical species from different sources to the Tyr17-Leu33 segment of orexin A peptide, which is the minimal fragment with affinity for orexin 1 receptor. As a clear oxidation process, we detected the conversion of terminal Tyrosine residue into a mixture of E/Z coumaric acid derivatives. In fact, UV irradiation and nitrosyl radicals were found to selectively induce the deamination of tyrosine into the corresponding olefin derivative, which was confirmed by HPLC-MS and NMR investigations.
Collapse
Affiliation(s)
- Simone Mariani
- Institute of Biomolecular Chemistry, CNR National Research Council of Italy, Via Marzolo 1, 35131, Padova, Italy
| | - Claudia Honisch
- Institute of Biomolecular Chemistry, CNR National Research Council of Italy, Via Marzolo 1, 35131, Padova, Italy
| | - Paolo Ruzza
- Institute of Biomolecular Chemistry, CNR National Research Council of Italy, Via Marzolo 1, 35131, Padova, Italy
| | - Stefano Tartaggia
- Institute of Biomolecular Chemistry, CNR National Research Council of Italy, Via Marzolo 1, 35131, Padova, Italy
| |
Collapse
|
3
|
Kukkonen JP, Turunen PM, Rinne MK. Detection of reduced orexin-A/hypocretin-1 and its fragments by orexin-A "gold-standard" radioimmunoassay. Sleep Med 2025; 131:106505. [PMID: 40250157 DOI: 10.1016/j.sleep.2025.106505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Orexin-A/hypocretin-1 level is determined in the cerebrospinal fluid samples as a part of clinical narcolepsy diagnostics utilizing a specific commercial radioimmunoassay (RIA); this assay is also widely used in research of many other conditions. The specificity of RIAs is in general variable, and little has been firmly disclosed about the specificity of this RIA assay. Thus, the validity of many research results obtained using the kit is unclear. At least metabolites of orexin-A have been proposed as potential interfering substances. METHODS Since this issue has not been systematically assessed, we decided to investigate it using synthetic variants of orexin-A and -B (intact peptides and peptide fragments as well as reduced orexin-A). RESULTS Our synthetic orexin-A bound correspondingly to the orexin-A standard included in the kit while orexin-B did not bind even at 10000-fold higher concentrations. Reduction of the disulfide bridges in orexin-A (giving orexin-A-SS) decreased its binding 25-fold. C-terminal truncation of orexin-A-SS was well tolerated - some of the fragments actually bound better than orexin-A-SS - while N-terminal truncation was not allowed. CONCLUSIONS The results demonstrate that the RIA kit is fairly selective for intact orexin-A among the peptides tested. However, this does not as such prove that it measures intact orexin-A in the physiological samples, and further studies including identification of physiological orexin-A metabolites are thus required. We also suggest that the redox milieu of cerebrospinal fluid - that has been suggested to vary in different diseases - may have an impact on what is measured with the kit.
Collapse
Affiliation(s)
- Jyrki P Kukkonen
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland.
| | - Pauli M Turunen
- Department of Physiology, Medicum, University of Helsinki, Helsinki, Finland
| | - Maiju K Rinne
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland
| |
Collapse
|
4
|
Ishikawa T, Kurimoto E, Joyal AA, Koike T, Kimura H, Scammell TE. An orexin agonist promotes wakefulness and inhibits cataplexy through distinct brain regions. Curr Biol 2025:S0960-9822(25)00356-2. [PMID: 40233754 DOI: 10.1016/j.cub.2025.03.040] [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: 11/15/2024] [Revised: 02/12/2025] [Accepted: 03/20/2025] [Indexed: 04/17/2025]
Abstract
Narcolepsy type 1, caused by selective loss of the orexin-producing neurons, is characterized by poor maintenance of wakefulness and cataplexy. Clinical trials show that orexin receptor 2 (OX2R) agonists substantially improve narcolepsy symptoms, but the key brain regions through which OX2R signaling produces these benefits are only partially understood. To address this question, we produced recombinant mice expressing the human diphtheria toxin receptor driven by the endogenous orexin promoter (orexinDTR mice). After injection with diphtheria toxin, orexinDTR mice had severe and selective loss of the orexin neurons, leading to narcolepsy symptoms, including poor maintenance of wakefulness and cataplexy; these symptoms were substantially improved by an OX2R-selective agonist OX-201. We then crossed orexinDTR mice with OX2R transcription-disrupted (TD) mice to produce a new model lacking orexin neurons and OX2R. We focally restored OX2R expression in specific brain regions of OX2R TD::orexinDTR mice and assessed whether OX-201 improves specific aspects of narcolepsy. In mice expressing OX2R only in the tuberomammillary nucleus (TMN) or basal forebrain (BF) regions, OX-201 improved maintenance of wakefulness but did not suppress cataplexy. In contrast, in mice expressing OX2R in the ventrolateral periaqueductal gray and lateral pontine tegmentum (vlPAG/LPT), OX-201 suppressed cataplexy without improving maintenance of wakefulness. These results suggest that OX2R signaling in the TMN and BF regions can stabilize wakefulness and OX2R signaling in the vlPAG/LPT region can suppress cataplexy, providing key insights into how orexins regulate wakefulness and muscle tone and how OX2R agonists improve the symptoms of narcolepsy.
Collapse
Affiliation(s)
- Takashi Ishikawa
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Emi Kurimoto
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan; Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Adam A Joyal
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Tatsuki Koike
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Haruhide Kimura
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
5
|
Moshirpour M, Horsley KG, Puche Saud S, McCance C, Scotland M, Antle MC. Agonists for glutamate, acetylcholine, and orexin cause non-photic phase shifts when applied to the intergeniculate leaflet. Neuroscience 2025; 574:114-123. [PMID: 40194656 DOI: 10.1016/j.neuroscience.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
The intergeniculate leaflet (IGL) and its neurotransmitter, Neuropeptide Y, are both necessary and sufficient inputs to the SCN to mediate non-photic phase shifting of circadian rhythms. In this study we examined what arousal inputs might participate in activation of the IGL during a non-photic manipulation. The ACh agonist carbachol caused non-photic phase shifts when applied to the IGL at CT6, but blocking ACh muscarinic receptors in the IGL with atropine did not attenuate phase shifts to a 3 h sleep deprivation (SD) procedure during the midday. Orexin, an important arousal neuropeptide, densely innervates the IGL. Pretreatment with the dual OX1/OX2 receptor antagonist MK-6096 did not attenuate phase shifts to 3 h midday SD. When injected into the IGL alone, orexin produced small and inconsistent phase shifts that overall did not differ significantly from vehicle control. The glutamate agonist NMDA caused non-photic-like phase shifts when applied to the IGL. While a cocktail of both carbachol and NMDA inhibited each other's phase shifting effects, a cocktail that included orexin, carbachol and NMDA reversed this inhibition and yielded the largest phase shifts of all. This suggests that the IGL is likely activated by numerous convergent arousal inputs during a phase-shifting non-photic manipulation.
Collapse
Affiliation(s)
- Mahtab Moshirpour
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Katelyn G Horsley
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Susana Puche Saud
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Chantelle McCance
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
| | - Maeve Scotland
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael C Antle
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
6
|
Duske J, D'Souza N, Mayer D, Dieterich DC, Fendt M. Orexinergic modulation of chronic jet lag-induced deficits in mouse cognitive flexibility. Neuropsychopharmacology 2025; 50:762-771. [PMID: 39478089 PMCID: PMC11914050 DOI: 10.1038/s41386-024-02017-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 03/19/2025]
Abstract
Cognitive flexibility and working memory are important executive functions mediated by the prefrontal cortex and can be impaired by circadian rhythm disturbances such as chronic jet lag (CJL) or shift work. In the present study, we used mice to investigate whether (1) simulated CJL impairs cognitive flexibility, (2) the orexin system is involved in such impairment, and (3) nasal administration of orexin A is able to reverse CJL-induced deficits in cognitive flexibility and working memory. Mice were exposed to either standard light-dark conditions or simulated CJL consisting of series of advance time shifts. Experiment (1) investigated the effects of a mild CJL protocol on cognitive flexibility using the attentional set shifting task. Experiment (2) used a stronger CJL protocol and examined CJL effects on the orexin system utilizing c-Fos and orexin immunohistochemistry. Experiment (3) tested whether nasal orexin application can rescue CJL-induced deficits in cognitive flexibility and working memory, the latter by measuring spontaneous alternation in the Y-maze. The present data show that CJL (1) impairs cognitive flexibility and (2) reduces the activity of orexin neurons in the lateral hypothalamus. (3) Nasal administration of orexin A rescued CJL-induced deficits in working memory and cognitive flexibility. These findings suggest that executive function impairments by circadian rhythm disturbances such as CJL are caused by dysregulation of orexinergic input to the prefrontal cortex. Compensation of decreased orexinergic input by nasal administration of orexin A could be a potential therapy for CJL- or shift work-induced human deficits in executive functions.
Collapse
Affiliation(s)
- Julius Duske
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Nicole D'Souza
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Institute of Neurophysiology, Goethe University, Frankfurt, Germany
| | - Dana Mayer
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniela C Dieterich
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center of Behavioural Brain Sciences, Otto-von-Guericke University, Magdeburg, Germany
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
- Center of Behavioural Brain Sciences, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
7
|
van Lemmen M, Dahan A, Hang Y, Jansen SC, Lu H, Naylor M, Olsson T, Sheikh S, Sullivan D, Tolkoff M, van der Schrier R, van Velzen M, von Rosenstiel P, Wu RL, Meyer S. TAK-925 (Danavorexton), an Orexin Receptor 2 Agonist, Reduces Opioid-induced Respiratory Depression and Sedation without Affecting Analgesia in Healthy Men. Anesthesiology 2025; 142:628-638. [PMID: 39804333 DOI: 10.1097/aln.0000000000005375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
BACKGROUND Orexin neuropeptides help regulate sleep/wake states, respiration, and pain. However, their potential role in regulating breathing, particularly in perioperative settings, is not well understood. TAK-925 (danavorexton), a novel orexin receptor 2-selective agonist, directly activates neurons associated with respiratory control in the brain and improves respiratory parameters in rodents undergoing fentanyl-induced sedation. This study assessed the safety and effect of danavorexton on ventilation in healthy men in an established remifentanil-induced respiratory depression model. METHODS This single-center, double-blind, placebo-controlled, two-way crossover, phase 1 trial randomized (1:1) 13 healthy men to danavorexton (11 mg [low-dose], then 19 mg [high-dose]) or placebo, under remifentanil infusion, on two occasions separated by a 36-h or longer washout period. Remifentanil infusion was titrated under isohypercapnic conditions to achieve an approximately 30 to 40% decrease in minute ventilation (from approximately 20 to approximately 14 l/min) before danavorexton/placebo administration. Assessments included safety, ventilation measurements, sedation, and pain tolerance. RESULTS A total of four (30.8%) danavorexton-treated participants and one (8.3%) placebo-treated participant experienced treatment-emergent adverse events (all mild in severity). Insomnia, lasting 1 day, occurred in one participant, and was considered related to danavorexton. Compared with placebo, low- and high-dose danavorexton significantly increased ventilation variables (observed mean [95% CI] change, sensitivity analysis model-based P values) including minute volume (8.2 [95% CI, 5.0 to 11.4] and 13.0 [95% CI, 9.4 to 16.5] l/min), tidal volume (312 [95% CI, 180 to 443] and 483 [95% CI, 309 to 657] ml), and respiratory rate (3.8 [95% CI, 1.9 to 5.7] and 5.2 [95% CI, 2.7 to 7.7] breaths/min; all P < 0.001). High-dose danavorexton significantly decreased sedation on a visual analog scale (-29.7 [95% CI, -54.1 to -5.3] mm; P < 0.001) and the Richmond Agitation Sedation Scale (0.4 [95% CI, 0.0 to 0.7]; P < 0.001) compared with placebo. Improvements in respiratory variables continued beyond completion of danavorexton infusion. No significant differences in pain tolerance were observed between danavorexton doses or between danavorexton and placebo (approximately 13% increase from baseline; low dose, P = 0.491; high dose, P = 0.140). CONCLUSIONS Danavorexton has effects on respiration and wakefulness in an opioid-induced respiratory depression setting without reversing opioid analgesia.
Collapse
Affiliation(s)
- Maarten van Lemmen
- Department of Anesthesiology, Anesthesia & Pain Research Unit, Leiden University Medical Center, Leiden, The Netherlands
| | - Albert Dahan
- Department of Anesthesiology, Anesthesia & Pain Research Unit, Leiden University Medical Center, Leiden, The Netherlands
| | - Yaming Hang
- Qualitative Clinical Pharmacology, Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | - Simone C Jansen
- Department of Anesthesiology, Anesthesia & Pain Research Unit, Leiden University Medical Center, Leiden, The Netherlands
| | - Hong Lu
- Qualitative Clinical Pharmacology, Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | - Melissa Naylor
- Global Portfolio Statistics, Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | - Tina Olsson
- Clinical Science, Neuroscience, Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | - Sarah Sheikh
- Neuroscience, Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | - Danielle Sullivan
- Statistics, Neuroscience, Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | - Max Tolkoff
- Statistics, Neuroscience, Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | - Rutger van der Schrier
- Department of Anesthesiology, Anesthesia & Pain Research Unit, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique van Velzen
- Department of Anesthesiology, Anesthesia & Pain Research Unit, Leiden University Medical Center, Leiden, The Netherlands
| | - Philipp von Rosenstiel
- Clinical Science, Neuroscience, Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | - Rebecca L Wu
- Clinical Science, Neuroscience, Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | - Seetha Meyer
- Clinical Science, Neuroscience, Takeda Development Center Americas, Inc., Lexington, Massachusetts
| |
Collapse
|
8
|
Ramou I, Janvier S, Druwé S, Sys C, Dekeyzer L, Claes P, Pardon E, Menet C, Steyaert J. Expression and purification of an activated orexin receptor 1- G-protein complex. Protein Expr Purif 2025; 228:106660. [PMID: 39761735 DOI: 10.1016/j.pep.2025.106660] [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: 11/20/2024] [Revised: 12/28/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
Orexin receptors constitute a family of class A G-protein coupled receptors. There are two subtypes of orexin receptors, namely OX1R and OX2R. OX1R and OX2R are widely distributed in the central nervous system and are the targets for the peptide neurotransmitters orexin-A and orexin-B. Orexins are involved in a plethora of key physiological functions such as regulation of the sleep/wake cycle, feeding behavior, energy homeostasis, and cognition. Dysfunction of the orexin system has been linked to various pathological conditions, such as narcolepsy, insomnia, obesity, addiction, cognitive impairment, and depression. The active state structure of OX2R has been elucidated, while the active state structure of OX1R remains unresolved. Here, we describe a method for the expression and purification of an activated OX1R bound to its native peptide ligand, orexin-A, in complex with a Dominant Negative Gsq protein and Nb35. The proteins were expressed in Hi5 insect cells and subsequently purified via two consecutive affinity chromatography steps, followed by a final polishing Size Exclusion Chromatography step. This study could stimulate further research into the activation mechanisms of OX1R and the structural determination of its active state structure.
Collapse
Affiliation(s)
- Ioanna Ramou
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, Brussels, Belgium
| | - Steven Janvier
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, Brussels, Belgium
| | | | | | | | | | - Els Pardon
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, Brussels, Belgium
| | | | - Jan Steyaert
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, Brussels, Belgium.
| |
Collapse
|
9
|
Chaki S. Orexin receptors: possible therapeutic targets for psychiatric disorders. Psychopharmacology (Berl) 2025:10.1007/s00213-025-06767-1. [PMID: 40153060 DOI: 10.1007/s00213-025-06767-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/30/2025]
Abstract
RATIONALE Orexins, comprising orexin-A and orexin-B, are neuropeptides with extensive projections throughout the central nervous system. They are implicated in a variety of physiological processes through their receptors, orexin type 1 (OX1) and orexin type 2 (OX2) receptors. Among the physiological functions of orexins, their role in sleep/wake regulation has garnered significant attention. Consequently, three orexin receptor antagonists that block both OX1 and OX2 receptors (dual orexin receptor antagonist; DORA) are available on the market for the treatment of insomnia. Additionally, another DORA, vornorexant, has been submitted for approval. OBJECTIVE Beyond sleep disorders, the orexin system is deeply implicated in the pathophysiology of several psychiatric disorders, including depression, anxiety, and substance use disorders. RESULTS Accumulating evidence indicates that orexin receptor antagonists improve behavioral abnormalities that mimic certain psychiatric disorders in animal models and are effective in treating these disorders or their symptoms in humans. Moreover, orexin receptor antagonists are expected not only to alleviate core symptoms of psychiatric disorders but also to improve sleep disturbances, which are often comorbid with these conditions. CONCLUSION Drug discovery and development targeting orexin receptors should provide novel therapeutic options for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Shigeyuki Chaki
- Taisho Pharmaceutical Co., Ltd, Toshima-Ku, Tokyo, 170-8633, Japan.
- Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| |
Collapse
|
10
|
Zhuang C, Yan H, Lu J, Zhou Y, Liu Y, Shi G, Li Y. Compensatory enhancement of orexinergic system functionality induced by amyloid-β protein: a neuroprotective response in Alzheimer's disease. Front Physiol 2025; 16:1529981. [PMID: 40196718 PMCID: PMC11973307 DOI: 10.3389/fphys.2025.1529981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
Background Amyloid-β protein (Aβ) accumulation is a defining characteristic of Alzheimer's disease (AD), resulting in neurodegeneration and a decline in cognitive function. Given orexin's well-documented role in enhancing memory and cognition, this study investigates its potential to regulate Aβ-induced neurotoxicity, offering new perspectives into AD management. Methods This paper simulated Aβ accumulation in the hippocampus of AD patients by administering Aβ1-42 oligomers into the bilateral hippocampal dentate gyrus of ICR mice. Inflammatory cytokines (IL-6, TNF-α) and orexin-A levels were measured by ELISA. Additionally, the excitability of orexinergic neurons was assessed by IHC targeting c-Fos expression. These methodologies evaluated the Aβ-induced neuroinflammation, orexinergic system functionality, and dexamethasone's (Dex) effects on these processes. Results Injection of Aβ1-42 oligomer resulted in elevated levels of IL-6, TNF-α, and orexin-A in the hippocampus, as well as increased excitability of orexinergic neurons in the lateral hypothalamus (LH). Dex treatment reduced neuroinflammation, causing a reduction in orexin-A levels and the excitability of orexinergic neurons. Conclusion Aβ-induced neuroinflammation is accompanied by enhanced levels of orexin-A and orexinergic neuron excitability. These findings suggest that the enhanced functionality of the orexinergic system may become a compensatory neuroprotective mechanism to counteract neuroinflammation and enhance cognitive function.
Collapse
Affiliation(s)
- Chenyu Zhuang
- Medical College, Yangzhou University, Yangzhou, China
| | - Hengyu Yan
- Medical College, Yangzhou University, Yangzhou, China
| | - Jiayu Lu
- Medical College, Yangzhou University, Yangzhou, China
| | - Yifan Zhou
- Medical College, Yangzhou University, Yangzhou, China
| | - Yanqing Liu
- Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Guoshan Shi
- Department of Basic Medical Sciences, Guizhou University of Chinese Medicine, Guiyang, China
| | - Yan Li
- Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China
- Department of Traditional Chinese Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, China
| |
Collapse
|
11
|
Berry EA, Huhulea EN, Ishibashi M, McGregor R, Siegel JM, Leonard CS. Chronic but not acute morphine exposure reversibly impairs spike generation and repetitive firing in a functionally distinct subpopulation of orexin neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644444. [PMID: 40196653 PMCID: PMC11974729 DOI: 10.1101/2025.03.20.644444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Orexin (hypocretin) neuropeptides regulate numerous essential functions including sleep/wake state stability and reward processing. Orexin synthesizing neurons respond to drug cues and undergo structural changes following persistent drug exposure. Post-mortem brains from opioid users, and opioid-treated rodents have orexin somata that become ~20 % smaller and ~50% more numerous and are postulated to promote hyper-motivation for drug-seeking though increased orexin release. Biophysical considerations suggest that decreased soma size should increase cellular excitability, however the impact of chronic opioids on firing ability, which drives peptide release, has not been explored. To test this, we assessed the intrinsic electrophysiological properties of orexin neurons by whole-cell recordings in slices from male orexin-EGFP mice treated by daily morphine or saline injections for two weeks. Paradoxically, we found that while daily morphine decreased average soma size, it impaired excitability in a subpopulation of orexin neurons identified by electrophysiological criteria as "H-type", while entirely sparing "D-type" neurons. This impairment was manifest by smaller, broader action potentials, variable firing and a downscaling of firing gain. These adaptations required more than a single morphine dose and recovered, along with soma size, after four weeks of passive withdrawal. Taken together, these observations indicate that daily opioid exposure differentially impacts H-type orexin neurons and predicts that the ability of these neurons to encode synaptic inputs into spike trains and to release neuropeptides becomes impaired in conjunction with opioid dependence.
Collapse
Affiliation(s)
| | - Ellen N. Huhulea
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Masaru Ishibashi
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ronald McGregor
- Neuropsychiatric Institute, University of California, Los Angeles, CA and Veterans Administration, Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Jerome M. Siegel
- Neuropsychiatric Institute, University of California, Los Angeles, CA and Veterans Administration, Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | | |
Collapse
|
12
|
Raggio M, Conte I, de Girolamo P, D'Angelo L. Modelling orexinergic system in ageing in the African turquoise killifish. Biogerontology 2025; 26:72. [PMID: 40085285 PMCID: PMC11909093 DOI: 10.1007/s10522-025-10214-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/01/2025] [Indexed: 03/16/2025]
Abstract
The orexinergic system is anatomically and functionally conserved in almost all vertebrates, and the role in healthy ageing and age-associated diseases has been studied in mammals. Here, we review the main findings on the age-related regulation of orexinergic system in mammals, including human patients and highlights how the fish Nothobranchius furzeri serves as an exceptional model to spearhead research and unravel the intricate mechanisms underlying orexinergic regulation during ageing. The ageing brain of this teleost is characterized by the presence of neurodegenerative processes similar to those associated with human pathologies rather than those of healthy ageing. We present an in-depth summary and discussion on the groundbreaking advances in understanding the neuroanatomical organization of the orexinergic system, its pivotal role in mammalian and fish models, and its profound involvement in healthy ageing and age-associated diseases.
Collapse
Affiliation(s)
- Maria Raggio
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Ivan Conte
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Paolo de Girolamo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Livia D'Angelo
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
13
|
Xie LQ, Hu B, Lu RB, Cheng YL, Chen X, Wen J, Xiao Y, An YZ, Peng N, Dai Y, Xie G, Guo Q, Peng H, Luo XH. Raptin, a sleep-induced hypothalamic hormone, suppresses appetite and obesity. Cell Res 2025; 35:165-185. [PMID: 39875551 PMCID: PMC11909135 DOI: 10.1038/s41422-025-01078-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 12/31/2024] [Indexed: 01/30/2025] Open
Abstract
Sleep deficiency is associated with obesity, but the mechanisms underlying this connection remain unclear. Here, we identify a sleep-inducible hypothalamic protein hormone in humans and mice that suppresses obesity. This hormone is cleaved from reticulocalbin-2 (RCN2), and we name it Raptin. Raptin release is timed by the circuit from vasopressin-expressing neurons in the suprachiasmatic nucleus to RCN2-positive neurons in the paraventricular nucleus. Raptin levels peak during sleep, which is blunted by sleep deficiency. Raptin binds to glutamate metabotropic receptor 3 (GRM3) in neurons of the hypothalamus and stomach to inhibit appetite and gastric emptying, respectively. Raptin-GRM3 signaling mediates anorexigenic effects via PI3K-AKT signaling. Of note, we verify the connections between deficiencies in the sleeping state, impaired Raptin release, and obesity in patients with sleep deficiency. Moreover, humans carrying an RCN2 nonsense variant present with night eating syndrome and obesity. These data define a unique hormone that suppresses food intake and prevents obesity.
Collapse
Affiliation(s)
- Ling-Qi Xie
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Biao Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ren-Bin Lu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ya-Lun Cheng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xin Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jie Wen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yao Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yu-Ze An
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ning Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yu Dai
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Genqing Xie
- Department of Endocrinology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hui Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Xiang-Hang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China.
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- FuRong Laboratory, Changsha, Hunan, China.
| |
Collapse
|
14
|
Mondino A, Vandewege MW, Artigas R, Delucchi L, Hermida KM, Yanez CE, Cullen JN, Friedenberg SG, Meurs KM, Stern JA, Olby NJ. Familial Narcolepsy in Dogo Argentino Dogs Is Caused by a Tandem Duplication Mutation in HCRTR2. J Vet Intern Med 2025; 39:e70056. [PMID: 40095233 PMCID: PMC11912018 DOI: 10.1111/jvim.70056] [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: 10/24/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Familial narcolepsy in dogs has been associated with mutations in the HCRTR2 gene in Labrador retrievers, dachshunds, and Doberman pinschers, with the causal mutation differing between breeds. OBJECTIVE To characterize the genetic mutation responsible for familial narcolepsy in Dogo Argentino dogs. ANIMALS Ten Dogo Argentino dogs, three narcoleptic and seven clinically normal, of which four were related and three were unrelated to the narcoleptic dogs. METHODS Case control prospective study. DNA was extracted from blood samples of all dogs. Whole-genome sequencing was performed on two affected dogs, and variants were identified using bioinformatic pipelines, with comparisons made to a database of 2766 dogs. Structural variants were validated through PCR and Sanger sequencing. RESULTS A novel tandem duplication in the HCRTR2 gene was identified. All three affected dogs and the clinically normal parents of one affected dog had this duplication, suggesting an autosomal recessive pattern of inheritance. This duplication was absent in the 2766 dogs in the database, emphasizing its potential relevance in the Dogo Argentino breed. CONCLUSIONS AND CLINICAL IMPORTANCE This discovery emphasizes the critical role of the HCRTR2 gene in narcolepsy in dogs, and the diversity of mutations that can lead to this condition. Further genetic testing in this breed is warranted to identify carriers and prevent the further spread of this condition.
Collapse
Affiliation(s)
- Alejandra Mondino
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Michael W Vandewege
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Rody Artigas
- Department of Animal Production and Health of Productive Systems, Facultad de Veterinaria, Academic Unit of Genetics and Animal Breeding, Montevideo, Uruguay
| | - Luis Delucchi
- Department of Pathology and Small Animal Clinic, Facultad de Veterinaria, Neurology Unit, Montevideo, Uruguay
| | - Karen M Hermida
- Department of Pathology and Small Animal Clinic, Facultad de Veterinaria, Neurology Unit, Montevideo, Uruguay
| | - Camila E Yanez
- Department of Pathology and Small Animal Clinic, Facultad de Veterinaria, Neurology Unit, Montevideo, Uruguay
| | - Jonah N Cullen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, Minnesota, USA
| | - Steven G Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, Minnesota, USA
| | - Kathryn M Meurs
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Joshua A Stern
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Natasha J Olby
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
15
|
Raymond JS, Athanasopoulos AG, Badolato CJ, Doolan TJ, Scicluna RL, Everett NA, Bowen MT, James MH. Emerging medications and pharmacological treatment approaches for substance use disorders. Pharmacol Biochem Behav 2025; 248:173952. [PMID: 39719161 DOI: 10.1016/j.pbb.2024.173952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Medications to treat substance use disorders (SUDs) remain suboptimal or, in the case of stimulants and cannabis, non-existent. Many factors have contributed to this paucity, including the biological complexity of addiction, regulatory challenges, and a historical lack of enthusiasm among pharmaceutical companies to commit resources to this disease space. Despite these headwinds, the recent opioid crisis has highlighted the devastating consequences of SUDs for both individuals and society, stimulating urgent efforts to identify novel treatment approaches. In addition, several neurobiological systems have been recently implicated in unique aspects of drug reward, opening the door to candidate medications with novel mechanisms of action. Here, we provide an overview of efforts to target several of these new systems, with a focus on those that are the subject of ongoing clinical trials as well as being areas of interest among the authors' research groups (MHJ, MTB, NAE). Specifically, we discuss new classes of medications targeting the serotonin 2A receptor (i.e., psychedelics), glucagon-like peptide 1 receptor, cannabidiol, dynorphin/kappa opioid receptor, orexin/hypocretin, and oxytocin receptor systems, as well as emergent approaches for modulating the more canonical dopaminergic system via agonist therapies for stimulant use disorders. Collectively, innovations in this space give reason for optimism for an improved therapeutic landscape for substance use disorders in the near future.
Collapse
Affiliation(s)
- Joel S Raymond
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA; Rutgers Addiction Research Center, Brain Health Institute, Rutgers Health, Piscataway, NJ, USA
| | - Alexander G Athanasopoulos
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Connie J Badolato
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Tylah J Doolan
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Rhianne L Scicluna
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Nicholas A Everett
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Michael T Bowen
- School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Morgan H James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA; Rutgers Addiction Research Center, Brain Health Institute, Rutgers Health, Piscataway, NJ, USA; School of Psychology, Faculty of Science, University of Sydney, Sydney, NSW, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
16
|
Li N, Huang L, Zhang B, Zhu W, Dai W, Li S, Xu H. The mechanism of different orexin/hypocretin neuronal projections in wakefulness and sleep. Brain Res 2025; 1850:149408. [PMID: 39706239 DOI: 10.1016/j.brainres.2024.149408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Since the discovery of orexin/hypocretin, numerous studies have accumulated evidence demonstrating its key role in various aspects of neuromodulation, including addiction, motivation, and arousal. This paper focuses on the projection of orexin neurons to specific target brain regions through distinct neural pathways to regulate sleep and arousal. We provide a detailed discussion of the projection mechanisms of orexin neurons to downstream neurons, particularly emphasizing their activation of monoaminergic and cholinergic neurons associated with arousal. Additionally, we briefly explore the immune response and inflammatory factors linked to the loss of orexin neurons. Our findings underscore the significance of understanding specific neural projections in the generation and maintenance of arousal, which could guide advancements in neuroscience and lead to new therapeutic opportunities for treating insomnia or narcolepsy.
Collapse
Affiliation(s)
- Nanxi Li
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Lishan Huang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Bin Zhang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Wenwen Zhu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Wenbin Dai
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Sen Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University.
| | - Houping Xu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
17
|
Carro-Domínguez M, Huwiler S, Oberlin S, Oesch TL, Badii G, Lüthi A, Wenderoth N, Meissner SN, Lustenberger C. Pupil size reveals arousal level fluctuations in human sleep. Nat Commun 2025; 16:2070. [PMID: 40021662 PMCID: PMC11871316 DOI: 10.1038/s41467-025-57289-5] [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: 10/20/2023] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
Recent animal research has revealed the intricate dynamics of arousal levels that are important for maintaining proper sleep resilience and memory consolidation. In humans, changes in arousal level are believed to be a determining characteristic of healthy and pathological sleep but tracking arousal level fluctuations has been methodologically challenging. Here we measured pupil size, an established indicator of arousal levels, by safely taping the right eye open during overnight sleep and tested whether pupil size affects cortical response to auditory stimulation. We show that pupil size dynamics change as a function of important sleep events across different temporal scales. In particular, our results show pupil size to be inversely related to the occurrence of sleep spindle clusters, a marker of sleep resilience. Additionally, we found pupil size prior to auditory stimulation to influence the evoked response, most notably in delta power, a marker of several restorative and regenerative functions of sleep. Recording pupil size dynamics provides insights into the interplay between arousal levels and sleep oscillations.
Collapse
Affiliation(s)
- Manuel Carro-Domínguez
- Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, 8092, Zurich, Switzerland
| | - Stephanie Huwiler
- Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, 8092, Zurich, Switzerland
| | - Stella Oberlin
- Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, 8092, Zurich, Switzerland
| | - Timona Leandra Oesch
- Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, 8092, Zurich, Switzerland
| | | | - Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Nicole Wenderoth
- Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, 8092, Zurich, Switzerland
- Future Health Technologies, Singapore-ETH Center, Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
- Neuroscience Center Zurich (ZNZ), University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Sarah Nadine Meissner
- Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, 8092, Zurich, Switzerland
| | - Caroline Lustenberger
- Neural Control of Movement Lab, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, 8092, Zurich, Switzerland.
- Neuroscience Center Zurich (ZNZ), University of Zurich, ETH Zurich, Zurich, Switzerland.
- Center of Competence Sleep & Health Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
18
|
Ruhrländer J, Syntila S, Schieffer E, Schieffer B. The Orexin System and Its Impact on the Autonomic Nervous and Cardiometabolic System in Post-Acute Sequelae of COVID-19. Biomedicines 2025; 13:545. [PMID: 40149526 PMCID: PMC11940130 DOI: 10.3390/biomedicines13030545] [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: 01/14/2025] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 03/29/2025] Open
Abstract
Orexins (OXs) are critical for regulating circadian rhythms, arousal, appetite, energy metabolism, and electrolyte balance, affecting both the autonomic nervous system (ANS) and the cardiovascular system (CVS). Disruption of the OX system can result in symptoms similar to those observed in post-acute sequelae of COVID-19 (PASC). This review emphasizes the adverse effects of OX dysregulation on autonomic and cardiometabolic functions in patients with PASC. Additionally, we highlight the potential of anti-OX therapies to provide neuroprotective, anti-inflammatory, and immunoregulatory benefits, offering hope for alleviating some of the debilitating symptoms associated with PASC.
Collapse
Affiliation(s)
- Jana Ruhrländer
- Department of Cardiology, Angiology and Critical Care Medicine, Philipps University Marburg, 35043 Marburg, Germany; (J.R.); (S.S.); (E.S.)
- State of Hessen Post-COVID Coordination Center, 35043 Marburg, Germany
| | - Styliani Syntila
- Department of Cardiology, Angiology and Critical Care Medicine, Philipps University Marburg, 35043 Marburg, Germany; (J.R.); (S.S.); (E.S.)
| | - Elisabeth Schieffer
- Department of Cardiology, Angiology and Critical Care Medicine, Philipps University Marburg, 35043 Marburg, Germany; (J.R.); (S.S.); (E.S.)
| | - Bernhard Schieffer
- Department of Cardiology, Angiology and Critical Care Medicine, Philipps University Marburg, 35043 Marburg, Germany; (J.R.); (S.S.); (E.S.)
- State of Hessen Post-COVID Coordination Center, 35043 Marburg, Germany
| |
Collapse
|
19
|
Li S, Mei Y, Jiang L, Yang X, Zeng W, Du Y. Oxazole and isoxazole-containing pharmaceuticals: targets, pharmacological activities, and their SAR studies. RSC Med Chem 2025:d4md00777h. [PMID: 40008190 PMCID: PMC11848632 DOI: 10.1039/d4md00777h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Oxazole, a five-membered aromatic heterocycle featuring a nitrogen and an oxygen atom separated by a carbon atom, and its isomer isoxazole, with directly attached oxygen and nitrogen atoms, have been pivotal in medicinal chemistry. Over the past few decades, the U.S. Food and Drug Administration (FDA) has approved more than 20 drugs containing these nuclei for various clinical conditions, including Tafamidis and Oxaprozin. Due to their unique physicochemical properties, these drugs often exhibit superior pharmacokinetic profiles and pharmacological effects compared to those with similar heterocycles. This review provides a comprehensive overview of all FDA-approved drugs containing oxazole and isoxazole nuclei, focusing on their pharmacological activities and structure-activity relationships.
Collapse
Affiliation(s)
- Shanshan Li
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University Tianjin 300072 China +86 22 27406121
| | - Yiou Mei
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University Tianjin 300072 China +86 22 27406121
| | - Luchen Jiang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University Tianjin 300072 China +86 22 27406121
| | - Xueyan Yang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University Tianjin 300072 China +86 22 27406121
| | - Wei Zeng
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University Tianjin 300072 China +86 22 27406121
| | - Yunfei Du
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University Tianjin 300072 China +86 22 27406121
| |
Collapse
|
20
|
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
|
21
|
Stapel B, Alvarenga ME, Kahl KG. Pharmacological and psychological approaches to insomnia treatment in cardiac patients: a narrative literature review. Front Psychiatry 2025; 16:1490585. [PMID: 40018681 PMCID: PMC11865029 DOI: 10.3389/fpsyt.2025.1490585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/24/2025] [Indexed: 03/01/2025] Open
Abstract
Sleep disorders are highly prevalent in the general population and are considered a major public health issue. Insomnia constitutes the most frequent sleep disorder in healthy individuals and has been shown to be even more frequent in patients with physical illnesses including cardiovascular diseases. Inadequate sleep quality and short sleep duration, independent of underlying causes, have been linked to the development and progression of cardiometabolic disorders. Additionally, insomnia has been found to be associated with adverse outcome measures, including daytime sleepiness, fatigue, decreased self-reported physical functioning, lower exercise capacity, poor health related quality of life, depressive symptoms, higher rates of hospitalization and increased mortality in patients with cardiovascular diseases. Against this background, comparatively little information is available in the literature regarding the treatment of chronic insomnia in cardiac patient populations. While guidelines for the general population suggest cognitive behavioral therapy for insomnia as a first-line treatment option and preliminary evidence suggests this treatment to be beneficial in cardiac patients with insomnia symptoms, it is often limited by availability and possibly the clinician's poor understanding of sleep issues in cardiac patients. Therefore, pharmacologic treatment remains an important option indicated by the high number of hypnotic drug prescriptions in the general population and in patients with cardiovascular disorders. In this narrative review of the literature, we summarize treatment options for chronic insomnia based on clinical guidelines for the general population and highlight necessary considerations for the treatment of patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Britta Stapel
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Marlies E. Alvarenga
- Institute of Health and Wellbeing, Federation University Australia and Victorian Heart Institute, Melbourne, VIC, Australia
| | - Kai G. Kahl
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| |
Collapse
|
22
|
Maski K, Heckler G, Worhach J, Mylonas D, Wang G, Szilagyi K, Zhang B, Diniz Behn C, Scammell TE, Stickgold R. Impaired sleep-dependent memory consolidation in pediatric narcolepsy type 1. Sleep 2025; 48:zsae238. [PMID: 39420719 PMCID: PMC11807881 DOI: 10.1093/sleep/zsae238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/15/2024] [Indexed: 10/19/2024] Open
Abstract
STUDY OBJECTIVES Disrupted nighttime sleep is common in pediatric narcolepsy type 1, yet its cognitive impact is unknown. As N2 sleep spindles are necessary for sleep-dependent memory consolidation, we hypothesized that narcolepsy type 1 impairs memory consolidation via N2 sleep fragmentation and N2 sleep spindle alterations. METHODS We trained 28 pediatric narcolepsy type 1 participants and 27 healthy controls (HCs) on a spatial declarative memory task before a nocturnal in-lab polysomnogram and then gave them a cued recall test upon awakening in the morning. We extracted wake and sleep stage bout numbers and N2 spindle characteristics from the polysomnogram and conducted mixed model analysis of sleep-dependent memory consolidation to identify group differences. RESULTS Narcolepsy type 1 participants had shorter N2 bout durations and associated shorter N2 spindles versus HC, but other N2 spindle features were similar. Narcolepsy type 1 participants had worse memory performance postsleep than HCs after adjusting for age and gender (mean memory consolidation HC: -3.1% ± 18.7, NT1: -15.6 ± 24.8, main effect group × time of testing F = 5.3, p = .03). We did not find significant relationships between sleep-dependent memory consolidation and N2 spindle characteristics. Notably, increased N1% was associated with worse sleep-dependent memory consolidation with results driven by the narcolepsy type 1 group. CONCLUSIONS Sleep-dependent memory consolidation is mildly impaired in youth with narcolepsy type 1 and findings may be attributed to increases in N1 sleep. Further studies are needed to determine if these findings are generalizable and reversible with sleep-based therapies.
Collapse
Affiliation(s)
- Kiran Maski
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Gillian Heckler
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Jennifer Worhach
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Dimitrios Mylonas
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MAUSA
| | - Grace Wang
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Klara Szilagyi
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Bo Zhang
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Cecilia Diniz Behn
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, CO, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Robert Stickgold
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
23
|
Dieguez C, López M, Casanueva F. Hypothalamic GHRH. Rev Endocr Metab Disord 2025:10.1007/s11154-025-09951-y. [PMID: 39913072 DOI: 10.1007/s11154-025-09951-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/28/2025] [Indexed: 02/07/2025]
Abstract
Despite initial discovery in pancreatic tumors, GHRH is a 44-amino acid peptide primarily expressed in the hypothalamus. Recent RNA sequencing clarifies GHRH expression: predominantly hypothalamic in humans, with some basal ganglia presence, while extending to additional central nervous system (CNS) regions in other species. GHRH binds to its G-protein coupled receptor (GHRHR) in the arcuate (ARC), ventromedial (VMH), and periventricular (PeN) nuclei of the hypothalamus to exert its effects. Notably, the highest non-brain expression is found in somatotroph cells of the pituitary, directly targeting growth hormone (GH) production. GHRH is the primary regulator of pulsatile GH secretion, counteracted by somatostatin. While early models proposed alternating GHRH/somatostatin bursts, others implicate somatostatin as the primary regulator of GH pulse timing. These models fail to fully explain species and gender differences, particularly regarding nutritional status. The discovery of ghrelin, acting via GHS-R1a on GHRH neurons, significantly advanced understanding of GH regulation. Ghrelin interacts intricately with GHRH, modulating its expression and neuronal activity. Ghrelin also exerts GHRH-independent GH stimulation and synergizes with GHRH. The crucial role of GHRH in GH regulation is demonstrated by its key involvement in the action of other GH regulators, such as leptin, neuropeptide Y (NPY), and orexins. However, these interactions have also revealed that the physiological effects of GHRH extend far beyond its canonical role as a GH secretagogue. In this context, GHRH is thought to be a key regulator of the sleep-wake cycle and may be involved in whole-body energy homeostasis. The objective of this review is to summarize the current knowledge on GHRH and to discuss the potential pleiotropic effect of this hypothalamic neuropeptide, far beyond its classical action as regulator of the somatotroph axis.
Collapse
Affiliation(s)
- Carlos Dieguez
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, 15782,, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain.
| | - Miguel López
- Department of Physiology, CiMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, 15782,, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
| | - Felipe Casanueva
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, 15706, Spain
- Department of Medicine, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, 15782, Santiago de Compotela, Spain
- Complejo Universitario de Santiago de Compostela, Santiago de Compostela, 15706, Spain
| |
Collapse
|
24
|
Lin H, Xu Y, Xiong H, Wang L, Shi Y, Wang D, Wang Z, Ren J, Wang S. Mechanism of action of Panax ginseng alcohol extract based on orexin-mediated autophagy in the treatment of sleep and cognition in aged sleep-deprived rats. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118907. [PMID: 39389397 DOI: 10.1016/j.jep.2024.118907] [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: 08/13/2024] [Revised: 09/29/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax ginseng (P. ginseng) C. A. Meyer. has been used extensively globally as a medicine. It has a therapeutic effect on sleep and is an attractive alternative for patients with insomnia. The United States Patent of Invention has approved the use of P. ginseng alcohol extract (GAE) in nutraceuticals or food to improve sleep. It has shown promise as an effective therapeutic agent for improving sleep and cognition. However, its mechanism of action is not yet fully understood. AIM OF THE STUDY To investigate the therapeutic benefits of GAE on sleep and cognition and its underlying mechanism in aged sleep-deprived rats, with a focus on orexin-mediated autophagy function. MATERIALS AND METHODS We conducted in vivo tests in an aged sleep-deprivation rat model produced using p-chlorophenylalanine (PCPA) coupled with modified multi-platform method to examine the therapeutic effects and mechanisms of GAE. A pentobarbital sodium-induced sleep test and water maze were used to assess sleep and cognitive performance, respectively. An enzyme-linked immunosorbent assay was used to determine orexin levels and aging and sleep markers in serum and hypothalamic tissues. Hematoxylin-eosin staining and Nissl staining were used to assess histopathological changes, and autophagy levels were assessed using transmission electron microscopy, immunofluorescence. Western blot and immunohistochemical staining were performed to detect the levels of orexin, orexin-receptor proteins, and autophagy-associated proteins to study the effects of GAE on hippocampal neurons, and the underlying mechanisms. RESULTS In aged sleep-deprived rats, GAE treatment prolonged sleep duration, improved cognitive function, prevented hippocampal neuronal damage, increased the number of Nissl bodies, improved aging and sleep markers, and enhanced the LC3A/B expression in autophagosomes and neurons. The amount of orexin in serum and hypothalamic tissue and OX1R, OX2R, and phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) proteins also reduced, which resulted in the inhibition of the PI3K/Akt/mTOR pathway and activation of the autophagy process. CONCLUSIONS GAE may reduce hypothalamic orexin secretion and interact with orexin receptors to inhibit the PI3K/Akt/mTOR signalling network and activate autophagy. This may be a potential mechanism of action of GAE in regulating sleep-related cognitive function.
Collapse
Affiliation(s)
- Haining Lin
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yunlong Xu
- Prevention and Treatment Center, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Huazhong Xiong
- Prevention and Treatment Center, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Lichao Wang
- Prevention and Treatment Center, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Yuqing Shi
- College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Dongyi Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Zixu Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Jixiang Ren
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China; Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China.
| | - Siming Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China; Key Laboratory of Ginseng Efficacy Substance Base and Biological Mechanism Research, Ministry of Education, Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
25
|
Aliev F, De Sa Nogueira D, Aston-Jones G, Dick DM. Genetic associations between orexin genes and phenotypes related to behavioral regulation in humans, including substance use. Mol Psychiatry 2025:10.1038/s41380-025-02895-4. [PMID: 39880903 DOI: 10.1038/s41380-025-02895-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/23/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025]
Abstract
The hypothalamic neuropeptide system of orexin (hypocretin) neurons provides projections throughout the neuraxis and has been linked to sleep regulation, feeding and motivation for salient rewards including drugs of abuse. However, relatively little has been done to examine genes associated with orexin signaling and specific behavioral phenotypes in humans. Here, we tested for association of twenty-seven genes involved in orexin signaling with behavioral phenotypes in humans. We tested the full gene set, functional subsets, and individual genes involved in orexin signaling. Our primary phenotype of interest was Externalizing, a composite factor comprised of behaviors and disorders associated with reward-seeking, motivation, and behavioral regulation. We also tested for association with additional phenotypes that have been related to orexin regulation in model organism studies, including alcohol consumption, problematic alcohol use, daytime sleepiness, insomnia, cigarettes per day, smoking initiation, and body mass index. The composite set of 27 genes corresponding to orexin function was highly associated with Externalizing, as well as with alcohol consumption, insomnia, cigarettes per day, smoking initiation and BMI. In addition, all gene subsets (except the OXR2/HCRTR2 subset) were associated with Externalizing. BMI was significantly associated with all gene subsets. The "validated factors for PPOX/HCRT" and "PPOX/HCRT upregulation" gene subsets also were associated with alcohol consumption. Individually, 8 genes showed a strong association with Externalizing, 12 with BMI, 7 with smoking initiation, 3 with alcohol consumption, and 2 with problematic alcohol use, after correction for multiple testing. This study indicates that orexin genes are associated with multiple behaviors and disorders related to self-regulation in humans. This is consistent with prior work in animals that implicated orexin signaling in motivational activation induced by salient stimuli, and supports the hypothesis that orexin signaling is an important potential therapeutic target for numerous behavioral disorders.
Collapse
Affiliation(s)
- Fazil Aliev
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - David De Sa Nogueira
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - Gary Aston-Jones
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - Danielle M Dick
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA.
| |
Collapse
|
26
|
Krauth N. Tug-of-War: Orexin and Dynorphin Effects on Reward Processing Circuits. J Neurosci 2025; 45:e1979242024. [PMID: 39843226 PMCID: PMC11756618 DOI: 10.1523/jneurosci.1979-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/27/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Affiliation(s)
- Nathalie Krauth
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
27
|
Xiao X, Yeghiazaryan G, Eggersmann F, Cremer AL, Backes H, Kloppenburg P, Hausen AC. Deficiency of orexin receptor type 1 in dopaminergic neurons increases novelty-induced locomotion and exploration. eLife 2025; 12:RP91716. [PMID: 39841059 PMCID: PMC11753781 DOI: 10.7554/elife.91716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025] Open
Abstract
Orexin signaling in the ventral tegmental area and substantia nigra promotes locomotion and reward processing, but it is not clear whether dopaminergic neurons directly mediate these effects. We show that dopaminergic neurons in these areas mainly express orexin receptor subtype 1 (Ox1R). In contrast, only a minor population in the medial ventral tegmental area express orexin receptor subtype 2 (Ox2R). To analyze the functional role of Ox1R signaling in dopaminergic neurons, we deleted Ox1R specifically in dopamine transporter-expressing neurons of mice and investigated the functional consequences. Deletion of Ox1R increased locomotor activity and exploration during exposure to novel environments or when intracerebroventricularely injected with orexin A. Spontaneous activity in home cages, anxiety, reward processing, and energy metabolism did not change. Positron emission tomography imaging revealed that Ox1R signaling in dopaminergic neurons affected distinct neural circuits depending on the stimulation mode. In line with an increase of neural activity in the lateral paragigantocellular nucleus (LPGi) of Ox1RΔDAT mice, we found that dopaminergic projections innervate the LPGi in regions where the inhibitory dopamine receptor subtype D2 but not the excitatory D1 subtype resides. These data suggest a crucial regulatory role of Ox1R signaling in dopaminergic neurons in novelty-induced locomotion and exploration.
Collapse
Affiliation(s)
- Xing Xiao
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of MetabolismCologneGermany
| | - Gagik Yeghiazaryan
- Department of Biology, Institute for Zoology, University of CologneCologneGermany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of CologneCologneGermany
| | - Fynn Eggersmann
- Department of Biology, Institute for Zoology, University of CologneCologneGermany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of CologneCologneGermany
| | - Anna Lena Cremer
- Max Planck Institute for Metabolism Research, Multimodal Imaging of Brain Metabolism GroupCologneGermany
| | - Heiko Backes
- Max Planck Institute for Metabolism Research, Multimodal Imaging of Brain Metabolism GroupCologneGermany
| | - Peter Kloppenburg
- Department of Biology, Institute for Zoology, University of CologneCologneGermany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of CologneCologneGermany
| | - Anne Christine Hausen
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of MetabolismCologneGermany
| |
Collapse
|
28
|
Huang Q, Hu B, Zhang P, Yuan Y, Yue S, Chen X, Liang J, Tang Z, Zhang B. Neuroscience of cancer: unraveling the complex interplay between the nervous system, the tumor and the tumor immune microenvironment. Mol Cancer 2025; 24:24. [PMID: 39825376 PMCID: PMC11740516 DOI: 10.1186/s12943-024-02219-0] [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: 11/21/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025] Open
Abstract
The study of the multifaceted interactions between neuroscience and cancer is an emerging field with significant implications for understanding tumor biology and the innovation in therapeutic approaches. Increasing evidence suggests that neurological functions are connected with tumorigenesis. In particular, the peripheral and central nervous systems, synapse, neurotransmitters, and neurotrophins affect tumor progression and metastasis through various regulatory approaches and the tumor immune microenvironment. In this review, we summarized the neurological functions that affect tumorigenesis and metastasis, which are controlled by the central and peripheral nervous systems. We also explored the roles of neurotransmitters and neurotrophins in cancer progression. Moreover, we examined the interplay between the nervous system and the tumor immune microenvironment. We have also identified drugs that target the nervous system for cancer treatment. In this review we present the work supporting that therapeutic agent targeting the nervous system could have significant potential to improve cancer therapy.
Collapse
Affiliation(s)
- Qibo Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China
| | - Bai Hu
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ye Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Shiwei Yue
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China.
| | - Junnan Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China.
| |
Collapse
|
29
|
Yu J, Liu H, Gao R, Wang TV, Li C, Liu Y, Yang L, Xu Y, Cui Y, Jia C, Huang J, Chen PR, Rao Y. Calcineurin: An essential regulator of sleep revealed by biochemical, chemical biological, and genetic approaches. Cell Chem Biol 2025; 32:157-173.e7. [PMID: 39740665 DOI: 10.1016/j.chembiol.2024.12.003] [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: 07/09/2024] [Revised: 10/29/2024] [Accepted: 12/09/2024] [Indexed: 01/02/2025]
Abstract
Research into mechanisms underlying sleep traditionally relies on electrophysiology and genetics. Because sleep can only be measured on whole animals by behavioral observations and physical means, no sleep research was initiated by biochemical and chemical biological approaches. We used phosphorylation sites of kinases important for sleep as targets for biochemical and chemical biological approaches. Sleep was increased in mice carrying a threonine-to-alanine substitution at residue T469 of salt-inducible kinase 3 (SIK3). Our biochemical purification and photo-crosslinking revealed calcineurin (CaN) dephosphorylation, both in vitro and in vivo, of SIK3 at T469 and S551, but not T221. Knocking down CaN regulatory subunit reduced daily sleep by more than 5 h, exceeding all known mouse mutants. Our work uncovered a critical physiological role for CaN in sleep and pioneered biochemical purification and chemical biology as effective approaches to study sleep.
Collapse
Affiliation(s)
- Jianjun Yu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Huijie Liu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Rui Gao
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Tao V Wang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Chenggang Li
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Yuxiang Liu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Lu Yang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Ying Xu
- National Center for Protein Sciences Phoenix, Beijing, China
| | - Yunfeng Cui
- Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Chenxi Jia
- National Center for Protein Sciences Phoenix, Beijing, China
| | - Juan Huang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Peng R Chen
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Yi Rao
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China.
| |
Collapse
|
30
|
Luo PX, Trainor BC. Hypocretin modulation of behavioral coping strategies for social stress. Neuroscience 2025; 564:126-134. [PMID: 39547335 DOI: 10.1016/j.neuroscience.2024.11.031] [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: 08/21/2024] [Revised: 10/30/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Best known for promoting wakefulness and arousal, the neuropeptide hypocretin (Hcrt) also plays an important role in mediating stress responses, including social stress. However, central and systemic manipulation of the Hcrt system has produced diverse behavioral outcomes in animal models. In this review, we first focus on studies where similar manipulations of the Hcrt system led to divergent coping behaviors. We hypothesize that Hcrt differentially facilitates active and passive coping behaviors in response to social stress by acting in different brain regions and on different cell types. We then focus on region and cell type-specific effects of Hcrt in the ventral pallidum, lateral habenula, ventral tegmental area, nucleus accumbens, amygdala, and bed nucleus of the stria terminalis. Overall, the evidence suggests that rather than enhancing or inhibiting behavioral responses to social stress, Hcrt may signal the heightened arousal associated with stressful contexts. The resulting behavioral effects depend on which circuits Hcrt release occurs in and which receptor types are activated. Further study is needed to determine how and why circuit specific activation of Hcrt neurons occurs.
Collapse
Affiliation(s)
- Pei X Luo
- Department of Psychology, University of California - Davis, Davis, CA 95616, USA
| | - Brian C Trainor
- Department of Psychology, University of California - Davis, Davis, CA 95616, USA.
| |
Collapse
|
31
|
Palagini L, Geoffroy PA, Manni R, Gemignani A. Circadian aspects in nonpharmacologic and pharmacologic treatment of insomnia. HANDBOOK OF CLINICAL NEUROLOGY 2025; 206:161-179. [PMID: 39864924 DOI: 10.1016/b978-0-323-90918-1.00010-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Insomnia disorder is a frequent sleep disorder leading to significant health and economic consequences. It has been proposed that individuals with insomnia may experience compromised deactivation systems of arousal, leading to a chronic state of hyperactivation of arousal known as hyperarousal, along with instability in the flip-flop system. Such disruptions may have a primarily impact on the sleep homeostatic drive process. Insomnia may indeed be associated with a disruption in the body's internal clock, known as chronodisruption. Despite the differentiation established in diagnostic nosology between insomnia disorder and circadian rhythm disorders, there is a significant body of evidence suggesting a complex interplay and frequent co-occurrence between these two conditions. In particular, circadian factors can predispose individuals to insomnia disorders, as well as precipitate and perpetuate their symptoms. Accordingly numerous pieces of evidence suggest that both pharmacologic and nonpharmacologic options for treating insomnia can have a resynchronization effect on circadian rhythms. The first-line treatment for chronic insomnia, according to current guidelines, is cognitive behavioral therapy for insomnia while pharmacologic interventions comprise of benzodiazepine receptor agonists also known as Z-drugs and short- to medium-acting benzodiazepines, melatonergic agonists such as ramelteon and melatonin 2mg prolonged release, and dual orexin receptor antagonists such as daridorexant, suvorexant, and lemborexant. At the same time, certain therapies recommended for circadian rhythm disorders can be utilized as adjunctive treatments for insomnia. Therefore, this chapter will discuss the circadian aspects of insomnia disorder and of its therapeutic approach. Furthermore, the effects of chronobiologic interventions, recommended for the treatment of circadian rhythm sleep-wake disorders, will be examined in individuals afflicted with chronic insomnia.
Collapse
Affiliation(s)
- Laura Palagini
- Department of Neuroscience, Psychiatric Section, Azienda Ospedaliera Universitaria Pisana (AUOP), Pisa, Italy.
| | - Pierre-Alexis Geoffroy
- Département de psychiatrie et d'addictologie, AP-HP, GHU Paris Nord, DMU Neurosciences, Hopital Bichat-Claude Bernard, Paris, France; Centre ChronoS, GHU Paris-Psychiatry & Neurosciences, Paris, France; Université Paris Cité, Inserm, NeuroDiderot, Paris, France
| | - Raffaele Manni
- Sleep Disorder Center, Mondino Hospital Pavia, Pavia, Italy
| | - Angelo Gemignani
- Department of Surgical Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy; Department of Neuroscience, Psychology Unit, University of Pisa Azienda Ospedaliera Universitaria Pisana (AUOP), Pisa, Italy
| |
Collapse
|
32
|
Moran KM, Enstrom AE, Jarrell L, Khashchuluun M, Tran A, Delville Y. Adolescent social stress alters the role of orexin innervation in the hindbrain in male hamsters. J Neuroendocrinol 2025; 37:e13457. [PMID: 39462511 DOI: 10.1111/jne.13457] [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: 05/06/2024] [Revised: 09/05/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024]
Abstract
Juvenile male hamsters exposed to chronic social stress eat more, gain weight, and have larger fat pads. The purpose of the present study was to address possible changes in food hoarding and orexin/hypocretin innervation in response to social stress. Male hamsters in early adolescence were exposed to a resident-intruder social stress paradigm or control condition daily for 2 weeks. Metabolism-related physiological measures and behaviors were tracked, and brains were immunocytochemically labeled for orexin-A. Our data confirm our previous observations on appetite, weight gain, and obesity, and showed a strong trend toward enhanced food hoarding as in prior studies. In addition, there were no statistically significant differences in orexin innervation in any brain area analyzed. However, unique correlation patterns were observed between orexin innervation and appetite or metabolic outcome. In particular, opposite correlations were observed between groups within the dorsal raphe nucleus, lateral parabrachial nucleus, and nucleus of the solitary tract. These opposite patterns of correlations suggest chronic social stress causes site-specific alterations in synaptic activity in relation with these behaviors.
Collapse
Affiliation(s)
- Kevin M Moran
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| | - Ava Elana Enstrom
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| | - Leah Jarrell
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| | | | - Anna Tran
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| | - Yvon Delville
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
33
|
Gratio V, Dragan P, Garcia L, Saveanu L, Nicole P, Voisin T, Latek D, Couvineau A. Pharmacodynamics of the orexin type 1 (OX 1) receptor in colon cancer cell models: A two-sided nature of antagonistic ligands resulting from partial dissociation of Gq. Br J Pharmacol 2024. [PMID: 39675769 DOI: 10.1111/bph.17422] [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: 01/23/2024] [Revised: 10/24/2024] [Accepted: 11/10/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND AND PURPOSE Orexins have important biological effects on the central and peripheral nervous systems. Their primary ability is to regulate the sleep-wake cycle. Orexins and their antagonists, via OX1 receptor have been shown to have proapoptotic and antitumor effects on various digestive cancers cell models. We investigated, (1) the ability of orexin-A and its antagonists to regulate OX1 receptor expression at the cell surface and (2), how OX1 antagonists induced proapoptotic effect in cancer cells models. EXPERIMENTAL APPROACH The OX1 receptor internalisation is determined by imaging flow cytometry in colon cancer cell models and the OX1 receptor coupling to G proteins via bioluminescence resonance energy transfer and molecular dynamic simulation. KEY RESULTS Orexin-A induced rapid receptor internalisation within 15 min via β-arrestin 2 recruitment, whereas antagonists had no effect. Furthermore, Gq is critical for receptor internalisation and signalling pathways, and no other G proteins appear to be recruited. Surprisingly, antagonists induced recruitment and conformational changes in Gq protein. Simulated molecular dynamics of agonists/orexin receptor/Gq complexes show that antagonists exhibits a similar binding mode, stable at the binding site and show conformational changes of ECL2, similar to that of the agonists. CONCLUSION AND IMPLICATIONS OX1 receptor activation induced orexin/β-arrestin-dependent internalisation, which was independent of the apoptotic pathway induced by orexins and antagonists. In addition, antagonists activate the Gq protein, suggesting its putative partial dissociation. These results suggest that the development of OX1 receptor targeting molecules, including orexin antagonists with antitumor properties, may pave the way for innovative cancer therapies.
Collapse
Affiliation(s)
- Valérie Gratio
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From Inflammation to Cancer in Digestive diseases (INDiD)", DHU UNITY, Université Paris Cité, Paris, France
- INSERM UMR1149/Inflammation Research Center (CRI), Flow Cytometry Platform (CytoCRI), DHU UNITY, Université Paris Cité, Paris, France
| | - Paulina Dragan
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland
| | - Laurine Garcia
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From Inflammation to Cancer in Digestive diseases (INDiD)", DHU UNITY, Université Paris Cité, Paris, France
| | - Loredana Saveanu
- INSERM UMR1149/Inflammation Research Center (CRI), Team "Antigen Presentation by Dendritic Cells to T cells (APreT)", DHU UNITY, Université Paris Cité, Paris, France
| | - Pascal Nicole
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From Inflammation to Cancer in Digestive diseases (INDiD)", DHU UNITY, Université Paris Cité, Paris, France
| | - Thierry Voisin
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From Inflammation to Cancer in Digestive diseases (INDiD)", DHU UNITY, Université Paris Cité, Paris, France
| | - Dorota Latek
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland
| | - Alain Couvineau
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From Inflammation to Cancer in Digestive diseases (INDiD)", DHU UNITY, Université Paris Cité, Paris, France
| |
Collapse
|
34
|
Jászberényi M, Thurzó B, Jayakumar AR, Schally AV. The Aggravating Role of Failing Neuropeptide Networks in the Development of Sporadic Alzheimer's Disease. Int J Mol Sci 2024; 25:13086. [PMID: 39684795 DOI: 10.3390/ijms252313086] [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: 10/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease imposes an increasing burden on aging Western societies. The disorder most frequently appears in its sporadic form, which can be caused by environmental and polygenic factors or monogenic conditions of incomplete penetrance. According to the authors, in the majority of cases, Alzheimer's disease represents an aggravated form of the natural aging of the central nervous system. It can be characterized by the decreased elimination of amyloid β1-42 and the concomitant accumulation of degradation-resistant amyloid plaques. In the present paper, the dysfunction of neuropeptide regulators, which contributes to the pathophysiologic acceleration of senile dementia, is reviewed. However, in the present review, exclusively those neuropeptides or neuropeptide families are scrutinized, and the authors' investigations into their physiologic and pathophysiologic activities have made significant contributions to the literature. Therefore, the pathophysiologic role of orexins, neuromedins, RFamides, corticotrope-releasing hormone family, growth hormone-releasing hormone, gonadotropin-releasing hormone, ghrelin, apelin, and natriuretic peptides are discussed in detail. Finally, the therapeutic potential of neuropeptide antagonists and agonists in the inhibition of disease progression is discussed here.
Collapse
Affiliation(s)
- Miklós Jászberényi
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
| | - Balázs Thurzó
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
- Emergency Patient Care Unit, Albert Szent-Györgyi Health Centre, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew V Schally
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
35
|
Luo PX, Serna Godoy A, Zakharenkov HC, Vang N, Wright EC, Balantac TA, Archdeacon SC, Black AM, Lake AA, Ramirez AV, Lozier LE, Perez MD, Bhangal I, Desta NM, Trainor BC. Hypocretin in the nucleus accumbens shell modulates social approach in female but not male California mice. Neuropsychopharmacology 2024; 49:2000-2010. [PMID: 39117901 PMCID: PMC11480414 DOI: 10.1038/s41386-024-01937-9] [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: 04/18/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024]
Abstract
The hypocretin (Hcrt) system modulates arousal and anxiety-related behaviors and has been considered as a novel treatment target for stress-related affective disorders. We examined the effects of Hcrt acting in the nucleus accumbens shell (NAcSh) and anterodorsal bed nucleus of the stria terminalis (adBNST) on social behavior in male and female California mice (Peromyscus californicus). In female but not male California mice, infusion of Hcrt1 into NAcSh decreased social approach. Weak effects of Hcrt1 on social vigilance were observed in both females and males. No behavioral effects of Hcrt1 infused into the adBNST were observed. Analyses of sequencing data from California mice and Mus musculus NAc showed that Hcrtr2 was more abundant than Hcrtr1, so we infused the selective Hcrt receptor 2 antagonist into the NAcSh, which increased social approach in females previously exposed to social defeat. A calcium imaging study in the NAcSh of females before and after stress exposure showed that neural activity increased immediately following the expression of social avoidance but not during freezing behavior. This observation is consistent with previous studies that identified populations of neurons in the NAc that drive avoidance. Intriguingly, calcium transients were not affected by stress. These data suggest that hypocretin acting in the NAcSh plays a key role in modulating stress-induced social avoidance.
Collapse
Affiliation(s)
- Pei X Luo
- Department of Psychology, University of California, Davis, CA, USA
| | | | | | - Nou Vang
- Department of Psychology, University of California, Davis, CA, USA
| | - Emily C Wright
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | | | | | - Alexis M Black
- Department of Psychology, University of California, Davis, CA, USA
| | - Alyssa A Lake
- Department of Psychology, University of California, Davis, CA, USA
| | - Alison V Ramirez
- Department of Psychology, University of California, Davis, CA, USA
| | - Lauren E Lozier
- Department of Psychology, University of California, Davis, CA, USA
| | - Melvin D Perez
- Department of Psychology, University of California, Davis, CA, USA
| | - Irvin Bhangal
- Department of Psychology, University of California, Davis, CA, USA
| | - Nile M Desta
- Department of Psychology, University of California, Davis, CA, USA
| | - Brian C Trainor
- Department of Psychology, University of California, Davis, CA, USA.
| |
Collapse
|
36
|
Christensen J, Vlassopoulos E, Barlow CK, Schittenhelm RB, Li CN, Sgro M, Warren S, Semple BD, Yamakawa GR, Shultz SR, Mychasiuk R. The beneficial effects of modafinil administration on repeat mild traumatic brain injury (RmTBI) pathology in adolescent male rats are not dependent upon the orexinergic system. Exp Neurol 2024; 382:114969. [PMID: 39332798 DOI: 10.1016/j.expneurol.2024.114969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/22/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
Abstract
The sleep-wake cycle plays an influential role in the development and progression of repeat mild traumatic brain injury (RmTBI)-related pathology. Therefore, we first aimed to manipulate the sleep-wake cycle post-RmTBI using modafinil, a wake-promoting substance used for the treatment of narcolepsy. We hypothesized that modafinil would exacerbate RmTBI-induced deficits. Chronic behavioural analyses were completed along with a 27-plex serum cytokine array, metabolomic and proteomic analyses of cerebrospinal fluid (CSF), as well as immunohistochemical staining in structures important for sleep/wake cycles, to examine orexin, melanin-concentrating hormone, tyrosine hydroxylase, and choline acetyltransferase, in the lateral hypothalamus, locus coeruleus, and basal forebrain, respectively. Contrary to expectation, modafinil administration attenuated behavioural deficits, metabolomic changes, and neuropathological modifications. Therefore, the second aim was to determine if the beneficial effects of modafinil treatment were driven by the orexinergic system. The same experimental protocol was used; however, RmTBI rats received chronic orexin-A administration instead of modafinil. Orexin-A administration produced drastically different outcomes, exacerbating anxiety-related and motor deficits, while also significantly disrupting their metabolomic and neuropathological profiles. These results suggest that the beneficial effects of modafinil administration post-RmTBI, work independently of its wake-promoting properties, as activation of the orexinergic wake-promoting system with orexin-A was detrimental. Overall, these findings highlight the complexity of sleep-wake changes in the injured brain and showcase the potential of the arousal and sleep systems in its treatment.
Collapse
Affiliation(s)
- Jennaya Christensen
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Elaina Vlassopoulos
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Christopher K Barlow
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Crystal N Li
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Marissa Sgro
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Samantha Warren
- Monash Micro Imaging, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Bridgette D Semple
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Centre for Trauma and Mental Health Research, Vancouver Island University, Nanaimo, B.C., Canada
| | - Richelle Mychasiuk
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
37
|
Vringer M, Zhou J, Gool JK, Bijlenga D, Lammers GJ, Fronczek R, Schinkelshoek MS. Recent insights into the pathophysiology of narcolepsy type 1. Sleep Med Rev 2024; 78:101993. [PMID: 39241492 DOI: 10.1016/j.smrv.2024.101993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/09/2024]
Abstract
Narcolepsy type 1 (NT1) is a sleep-wake disorder in which people typically experience excessive daytime sleepiness, cataplexy and other sleep-wake disturbances impairing daily life activities. NT1 symptoms are due to hypocretin deficiency. The cause for the observed hypocretin deficiency remains unclear, even though the most likely hypothesis is that this is due to an auto-immune process. The search for autoantibodies and autoreactive T-cells has not yet produced conclusive evidence for or against the auto-immune hypothesis. Other mechanisms, such as reduced corticotrophin-releasing hormone production in the paraventricular nucleus have recently been suggested. There is no reversive treatment, and the therapeutic approach is symptomatic. Early diagnosis and appropriate NT1 treatment is essential, especially in children to prevent impaired cognitive, emotional and social development. Hypocretin receptor agonists have been designed to replace the attenuated hypocretin signalling. Pre-clinical and clinical trials have shown encouraging initial results. A better understanding of NT1 pathophysiology may contribute to faster diagnosis or treatments, which may cure or prevent it.
Collapse
Affiliation(s)
- Marieke Vringer
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jingru Zhou
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jari K Gool
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands; Department of Anatomy & Neurosciences, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Compulsivity, Impulsivity and Attention, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Denise Bijlenga
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Gert Jan Lammers
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Rolf Fronczek
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Mink S Schinkelshoek
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
38
|
Ishizaka Y, Watanabe H, Ono M. Structure-Affinity-Pharmacokinetics Relationships of Novel 18F-Labeled 1,4-Diazepane Derivatives for Orexin 1 Receptor Imaging. J Med Chem 2024; 67:18781-18793. [PMID: 39431857 DOI: 10.1021/acs.jmedchem.4c01090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
The orexin 1 receptor (OX1R) has been suggested to be involved in the reward and autonomic nervous systems. Positron emission tomography (PET) of OX1R contributes to elucidating its role and developing new drugs. However, there are no useful PET probes for in vivo imaging of OX1R. Here, we newly designed and synthesized 18F-labeled 1,4-diazepane derivatives and evaluated their utilities as OX1R PET probes. In particular, BTF showed high and selective binding affinity for OX1R. In a biodistribution study using normal mice, [18F]BTF exhibited brain uptake, and radioactivity in the brain was significantly decreased by preinjection of unlabeled BTF. In a PET/CT study, it was suggested that [18F]BTF has the potential to visualize high-expression regions of OX1R in the normal mouse brain. Collectively, [18F]BTF has the fundamental features of an OX1R PET probe, and further studies may lead to the development of more useful probes.
Collapse
Affiliation(s)
- Yui Ishizaka
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
39
|
Di Marco T, Djonlagic I, Dauvilliers Y, Sadeghi K, Little D, Datta AN, Hubbard J, Hajak G, Krystal A, Olivieri A, Parrino L, Puryear CB, Zammit G, Donoghue J, Scammell TE. Effect of daridorexant on sleep architecture in patients with chronic insomnia disorder: a pooled post hoc analysis of two randomized phase 3 clinical studies. Sleep 2024; 47:zsae098. [PMID: 38644625 PMCID: PMC11543623 DOI: 10.1093/sleep/zsae098] [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] [Revised: 04/09/2024] [Indexed: 04/23/2024] Open
Abstract
STUDY OBJECTIVES Post hoc analysis to evaluate the effect of daridorexant on sleep architecture in people with insomnia, focusing on features associated with hyperarousal. METHODS We studied sleep architecture in adults with chronic insomnia disorder from two randomized phase 3 clinical studies (Clinicaltrials.gov: NCT03545191 and NCT03575104) investigating 3 months of daridorexant treatment (placebo, daridorexant 25 mg, daridorexant 50 mg). We analyzed sleep-wake transition probabilities, EEG spectra, and sleep spindle properties including density, dispersion, and slow oscillation phase coupling. The wake EEG similarity index (WESI) was determined using a machine learning algorithm analyzing the spectral profile of the EEG. RESULTS At month 3, daridorexant 50 mg decreased wake-to-wake transition probabilities (p < .05) and increased the probability of transitions from wake-to-N1 (p < .05), N2 (p < .05), and REM sleep (p < .05), as well as from N1-to-N2 (p < .05) compared to baseline and placebo. Daridorexant 50 mg decreased relative beta power during wake (p = .011) and N1 (p < .001) compared to baseline and placebo. During the wake, relative alpha power decreased (p < .001) and relative delta power increased (p < .001) compared to placebo. Daridorexant did not alter EEG spectra bands in N2, N3, and REM stages or in sleep spindle activity. Daridorexant decreased the WESI score during wake compared to baseline (p = .004). Effects with 50 mg were consistent between months 1 and 3 and less pronounced with 25 mg. CONCLUSIONS Daridorexant reduced EEG features associated with hyperarousal as indicated by reduced wake-to-wake transition probabilities and enhanced spectral features associated with drowsiness and sleep during wake and N1. CLINICAL TRIALS ClinicalTrials.gov NCT03545191: study to assess the efficacy and safety of ACT-541468 (daridorexant) in adult and elderly participants with insomnia disorder. URL: Study Details | study to assess the efficacy and safety of ACT-541468 (daridorexant) in adult and elderly participants with insomnia disorder | ClinicalTrials.gov ClinicalTrials.gov NCT03575104:study to assess the efficacy and safety of ACT-541468 (daridorexant) in adult and elderly participants who experience difficulties sleeping. URL: study details | study to assess the efficacy and safety of ACT-541468 (daridorexant) in adult and elderly participants who experience difficulties sleeping | ClinicalTrials.gov.
Collapse
Affiliation(s)
- Tobias Di Marco
- Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
- Department of Clinical Research, University of Basel, Schanzenstrasse, Basel
| | - Ina Djonlagic
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yves Dauvilliers
- Centre National de Référence Narcolepsie, Unité du Sommeil, CHU Montpellier, Hôpital Gui-de-Chauliac, Université de Montpellier, INSERM INM, Montpellier, France
| | | | | | - Alexandre N Datta
- Zentrum für Schlaf- und Chronomedizin der Basler Universitätskliniken, University Children’s Hospital Basel, Basel, Switzerland
| | | | - Göran Hajak
- Social Foundation Bamberg, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Bamberg, Germany
| | - Andrew Krystal
- Departments of Psychiatry and Neurology, University of California, San Francisco, CA, USA
| | | | - Liborio Parrino
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Gary Zammit
- Clinilabs Drug Development Corporation, New York, NY, USA
| | | | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
40
|
Vanini G. Are orexins-hypocretins the culprit for rapid eye movement sleep behavior disorder in narcolepsy? Sleep 2024; 47:zsae208. [PMID: 39238164 PMCID: PMC11543618 DOI: 10.1093/sleep/zsae208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Indexed: 09/07/2024] Open
Affiliation(s)
- Giancarlo Vanini
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
41
|
Podder S, Murata Y, Taniguchi M, Shimizu S, Yamaguchi M. Synaptic plasticity and roles of orexin in distinct domains of the olfactory tubercle. Front Neural Circuits 2024; 18:1473403. [PMID: 39574931 PMCID: PMC11578722 DOI: 10.3389/fncir.2024.1473403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024] Open
Abstract
Olfactory behavior is highly plastic, and the olfactory tubercle (OT), a component of the olfactory cortex and ventral striatum, includes anteromedial (amOT) and lateral (lOT) domains with roles in attractive and aversive olfactory behavioral learning, respectively. However, the underlying properties of synaptic plasticity in these domains are incompletely understood. Synaptic plasticity is regulated by multiple signals including synaptic inputs and neuromodulators. Interestingly, the amOT domain exhibits high expression of various receptors for neuromodulators. We investigated synaptic plasticity in mouse OT slices by combining electrical stimulation and treatment with the appetite-promoting neuropeptide orexin, the receptors of which are highly expressed in the amOT. In both the amOT and lOT, one round of 2-Hz burst stimulation elicited short-term potentiation of the field excitatory postsynaptic potential, whereas three rounds of stimulation induced long-term potentiation (LTP) that persisted for 150 min. In the amOT, orexin-A induced LTP was blocked by the orexin receptor type 1 antagonist SB334867. Orexin-A also facilitated LTP induction in the amOT by one round of 2-Hz burst stimulation. By contrast, these effects were not observed in the lOT. These results highlighted the similarity and difference in synaptic plasticity between the OT domains and suggested that orexin facilitates synaptic plasticity in the amOT during olfactory learning processes such as food odor learning.
Collapse
Affiliation(s)
| | - Yoshihiro Murata
- Department of Physiology, Kochi Medical School, Kochi University, Kochi, Japan
| | | | | | - Masahiro Yamaguchi
- Department of Physiology, Kochi Medical School, Kochi University, Kochi, Japan
| |
Collapse
|
42
|
Xu W, Ding W, Zhang Y, Wang S, Yan X, Xu Y, Zhi X, Liu R. The Role of T Cells in the Pathogenesis of Narcolepsy Type 1: A Narrative Review. Int J Mol Sci 2024; 25:11914. [PMID: 39595997 PMCID: PMC11593411 DOI: 10.3390/ijms252211914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Narcolepsy type 1 (NT1) is an uncommon, persistent sleep disorder distinguished by significant daytime sleepiness, episodes of cataplexy, and irregularities in rapid eye movement sleep. The etiology of NT1 is linked to the destruction of hypothalamic neurons responsible for the synthesis of the wake-promoting neuropeptide known as hypothalamic orexin. The pathophysiological mechanisms underlying NT1 remain inadequately elucidated; however, a model that incorporates the interplay of genetic predisposition, environmental influences, immune system factors, and a deficiency in hypocretin (HCRT) provides a framework for elucidating the pathogenesis of NT1. The prevalence of NT1 has been observed to rise following influenza A (H1N1) pdm09 and the administration of the Pandemrix influenza vaccine. The strong association between narcolepsy and the HLA-DQB1*06:02 allele strongly indicates an autoimmune etiology for this condition. Increasing evidence suggests that T cells play a critical role in this autoimmune-mediated HCRT neuronal loss. Studies have identified specific T cell subsets, including CD4+ and CD8+ T cells, that target HCRT neurons, contributing to their destruction. Clarifying the pathogenesis of NT1 driven by autoimmune T cells is crucial for the development of effective therapeutic interventions for this disorder. This review examines the risk factors associated with the pathogenesis of NT1, explores the role of T cells within the immune system in the progression of NT1, and evaluates immune-mediated animal models alongside prospective immunotherapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rongzeng Liu
- Department of Immunology, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471003, China
| |
Collapse
|
43
|
Mohammadkhani A, Mitchell C, James MH, Borgland SL, Dayas CV. Contribution of hypothalamic orexin (hypocretin) circuits to pathologies of motivation. Br J Pharmacol 2024; 181:4430-4449. [PMID: 39317446 PMCID: PMC11458361 DOI: 10.1111/bph.17325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 06/17/2024] [Accepted: 06/28/2024] [Indexed: 09/26/2024] Open
Abstract
The orexin (also known as hypocretin) system, consisting of neuropeptides orexin-A and orexin-B, was discovered over 25 years ago and was immediately identified as a central regulator of sleep and wakefulness. These peptides interact with two G-protein coupled receptors, orexin 1 (OX1) and orexin 2 (OX2) receptors which are capable of coupling to all heterotrimeric G-protein subfamilies, but primarily transduce increases in calcium signalling. Orexin neurons are regulated by a variety of transmitter systems and environmental stimuli that signal reward availability, including food and drug related cues. Orexin neurons are also activated by anticipation, stress, cues predicting motivationally relevant information, including those predicting drugs of abuse, and engage neuromodulatory systems, including dopamine neurons of the ventral tegmental area (VTA) to respond to these signals. As such, orexin neurons have been characterized as motivational activators that coordinate a range of functions, including feeding and arousal, that allow the individual to respond to motivationally relevant information, critical for survival. This review focuses on the role of orexins in appetitive motivation and highlights a role for these neuropeptides in pathologies characterized by inappropriately high levels of motivated arousal (overeating, anxiety and substance use disorders) versus those in which motivation is impaired (depression).
Collapse
Affiliation(s)
- Aida Mohammadkhani
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Alberta, Canada
| | - Caitlin Mitchell
- School of Biomedical Sciences and Pharmacy, University of Newcastle, University Drive, Callaghan, New South Wales, Australia
- The Hunter Medical Research, New Lambton Heights, New South Wales, Australia
| | - Morgan H James
- Department of Psychiatry and Brain Health Institute, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Alberta, Canada
| | - Christopher V Dayas
- School of Biomedical Sciences and Pharmacy, University of Newcastle, University Drive, Callaghan, New South Wales, Australia
- The Hunter Medical Research, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
44
|
Flores-Ramirez FJ, Illenberger JM, Martin-Fardon R. Interaction between corticotropin-releasing factor, orexin, and dynorphin in the infralimbic cortex may mediate exacerbated alcohol-seeking behavior. Neurobiol Stress 2024; 33:100695. [PMID: 39640001 PMCID: PMC11617300 DOI: 10.1016/j.ynstr.2024.100695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
A major challenge for the treatment of alcohol use disorder (AUD) is relapse to alcohol use, even after protracted periods of self-imposed abstinence. Stress significantly contributes to the chronic relapsing nature of AUD, given its long-lasting ability to elicit intense craving and precipitate relapse. As individuals transition to alcohol dependence, compensatory allostatic mechanisms result in insults to hypothalamic-pituitary-adrenal axis function, mediated by corticotropin-releasing factor (CRF), which is subsequently hypothesized to alter brain reward pathways, influence affect, elicit craving, and ultimately perpetuate problematic drinking and relapse vulnerability. Orexin (OX; also called hypocretin) plays a well-established role in regulating diverse physiological processes, including stress, and has been shown to interact with CRF. Interestingly, most hypothalamic cells that express Ox mRNA also express Pdyn mRNA. Both dynorphin and OX are located in the same synaptic vesicles, and they are co-released. The infralimbic cortex (IL) of the medial prefrontal cortex (mPFC) has emerged as being directly involved in the compulsive nature of alcohol consumption during dependence. The IL is a CRF-rich region that receives OX projections from the hypothalamus and where OX receptor mRNA has been detected. Although not thoroughly understood, anatomical and behavioral pharmacology data suggest that CRF, OX, and dynorphin may interact, particularly in the IL, and that functional interactions between these three systems in the IL may be critical for the etiology and pervasiveness of compulsive alcohol seeking in dependent subjects that may render them vulnerable to relapse. The present review presents evidence of the role of the IL in AUD and discusses functional interactions between CRF, OX, and dynorphin in this structure and how they are related to exacerbated alcohol drinking and seeking.
Collapse
Affiliation(s)
- Francisco J. Flores-Ramirez
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Department of Psychology, California State University, San Marcos, CA, USA
| | | | - Rémi Martin-Fardon
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
45
|
Carpi M, Mercuri NB, Liguori C. Orexin Receptor Antagonists for the Prevention and Treatment of Alzheimer's Disease and Associated Sleep Disorders. Drugs 2024; 84:1365-1378. [PMID: 39365407 PMCID: PMC11602839 DOI: 10.1007/s40265-024-02096-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/05/2024]
Abstract
Orexins/hypocretins are neuropeptides produced by the hypothalamic neurons, binding two G-protein coupled receptors (orexin 1 and orexin 2 receptors) and playing a critical role in regulating arousal, wakefulness, and various physiological functions. Given the high prevalence of sleep disturbances in Alzheimer's disease (AD) and their reported involvement in AD pathophysiology, the orexin system is hypothesized to contribute to the disease pathogenesis. Specifically, recent evidence suggests that orexin's influence may extend beyond sleep regulation, potentially affecting amyloid-β and tau pathologies. Dual orexin receptor antagonists (DORAs), namely suvorexant, lemborexant, and daridorexant, demonstrated efficacy in treating chronic insomnia disorder across diverse clinical populations. Considering their stabilizing effects on sleep parameters and emerging evidence of a possible neuroprotective role, these agents represent a promising strategy for AD management. This leading article reviews the potential use of orexin receptor antagonists in AD, particularly focusing on their effect in modulating disease-associated sleep disturbances and clinical outcomes. Overall, clinical studies support the use of DORAs to enhance sleep quality in patients with AD with comorbid sleep and circadian sleep-wake rhythm disorders. Preliminary results also suggest that these compounds might influence AD pathology, potentially affecting disease progression. Conversely, research on selective orexin receptor antagonists in AD is currently limited. Further investigation is needed to explore orexin antagonism not only as a symptomatic treatment for sleep disturbances, but also for its broader implications in modifying AD neurodegeneration, emphasizing mechanisms of action and long-term outcomes.
Collapse
Affiliation(s)
- Matteo Carpi
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome "Tor Vergata", Viale Oxford 81, 00133, Rome, Italy
| | - Nicola Biagio Mercuri
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome "Tor Vergata", Viale Oxford 81, 00133, Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", Viale Oxford 81, 00133, Rome, Italy
| | - Claudio Liguori
- Sleep Medicine Centre, Neurology Unit, University Hospital of Rome "Tor Vergata", Viale Oxford 81, 00133, Rome, Italy.
- Department of Systems Medicine, University of Rome "Tor Vergata", Viale Oxford 81, 00133, Rome, Italy.
| |
Collapse
|
46
|
Xiang X, Wang F, Chen C, Guan Z, Zhou W. Orexinergic projections to substantia innominata mediate arousal and analgesia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620973. [PMID: 39554139 PMCID: PMC11565723 DOI: 10.1101/2024.10.29.620973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Understanding neural circuits involved in anesthesia is crucial for improving its safety and efficacy. Hypothalamic orexin neurons (LHA OX ), projecting broadly, are essential in regulating arousal and pain. However, the precise targets remain unclear. Here we investigated the orexin projections to the substantia innominata (SI). Combining optogenetics, fiber photometry, and EEG/EMG allowed us to manipulate orexin activities, while simultaneously recording local ligand release and global cortical activities during anesthesia. Brain slice electrophysiology revealed the synaptic connections in the SI, while RNAscope was employed to examine the distribution of orexin receptors and downstream neuronal types. Presynaptic vesicles were identified in the orexin terminals in the SI, where 49.16% of cells expressed OX2R and 6.8% expressed OX1R. Orexin release in the SI was reversibly suppressed by isoflurane. Optogenetic activation of the LHA OX →SI circuit significantly increased orexin release and promoted arousal from various anesthesia stages, including reanimation during 0.75% isoflurane (p < 0.0001), prolongation of 3% isoflurane induction (p = 0.0033), and acceleration of emergence from 2% isoflurane (p < 0.0001). Furthermore, activating this circuit induced analgesia to both thermal (p = 0.0074) and inflammatory (p = 0.0127) pain. Patch-clamp recordings revealed that optogenetic activation of orexin terminals in the SI elicited excitatory postsynaptic currents, which were blocked by the OX2R antagonist. SI contains more GABAergic (28.17%) and glutamatergic (11.96%) neurons than cholinergic neurons (4.13%), all of which expressed OX2R. Thus, LHA OX neurons innervate SI neurons to regulate both arousal and pain predominantly through OX2R.
Collapse
|
47
|
Krause GM, Chirich Barreira LM, Albrecht A. Spatial mRNA expression patterns of orexin receptors in the dorsal hippocampus. Sci Rep 2024; 14:24788. [PMID: 39433837 PMCID: PMC11494061 DOI: 10.1038/s41598-024-76237-9] [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: 04/19/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
Orexins are wake-promoting neuropeptides that originate from hypothalamic neurons projecting to widespread brain areas throughout the central nervous system. They modulate various physiological functions via their orexin 1 (OXR1) and 2 (OXR2) receptors, including sleep-wake rhythm but also cognitive functions such as memory formation. Here, we provide a detailed analysis of OXR1 and OXR2 mRNA expression profiles in the dorsal hippocampus as a key region for memory formation, using RNAscope multiplex in situ hybridization. Interconnected subareas relevant for cognition and memory such as the medial prefrontal cortex and the nucleus reuniens of the thalamus were assessed as well. Both receptor types display distinct profiles, with the highest percentage of OXR1 mRNA-positive cells in the hilus of the dentate gyrus. Here, the content of OXR1 mRNA per cell was slightly modulated at selected time points over a 12 h light/ 12 dark light phase. Using RNAScope and quantitative polymerase chain reaction approaches, we began to address a cell-type specific expression of OXR1 in hilar GABAergic interneurons. The distinct expression profiles of both receptor subtypes within hippocampal subareas and circuits provide an interesting basis for future interventional studies on orexin receptor function in spatial and contextual memory.
Collapse
Affiliation(s)
- Gina Marie Krause
- Institute of Anatomy, Otto-von-Guericke-University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | | | - Anne Albrecht
- Institute of Anatomy, Otto-von-Guericke-University, Leipziger Str. 44, 39120, Magdeburg, Germany.
- Center for Behavioral Brain Sciences (CBBS), Universitätsplatz 2, 39106, Magdeburg, Germany.
- German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Magdeburg, Germany.
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany.
| |
Collapse
|
48
|
Hitrec T, Del Vecchio F, Alberti L, Luppi M, Martelli D, Occhinegro A, Piscitiello E, Taddei L, Tupone D, Amici R, Cerri M. Activation of orexin-A (hypocretin-1) receptors in the Raphe Pallidus at different ambient temperatures in the rat: effects on thermoregulation, cardiovascular control, sleep, and feeding behavior. Front Neurosci 2024; 18:1458437. [PMID: 39429700 PMCID: PMC11486763 DOI: 10.3389/fnins.2024.1458437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
The Raphe Pallidus (RPa) is a brainstem nucleus containing sympathetic premotor neurons that control thermogenesis and modulate cardiovascular function. It receives inputs from various hypothalamic areas, including the Lateral Hypothalamus (LH), a heterogeneous region intricately involved in several autonomic and behavioral functions. A key subpopulation of neurons in the LH expresses orexin/hypocretin, a neuropeptide which is crucially involved in the regulation of the wake-sleep states and feeding behavior. The RPa receives orexinergic projections from the LH and orexinergic signalling in the RPa has been shown to enhance thermogenesis in the anaesthetized rat, but only in the presence of an already existing thermogenic drive, without significantly affecting cardiovascular function. The present work was aimed at exploring the effects on thermoregulation and autonomic function and the possible role in the modulation of the wake-sleep states and feeding behavior of orexin injection in the RPa in the free-behaving rat. In order to assess the influence of an already present thermogenic drive on orexinergic signalling in the RPa, animals were studied at three different ambient temperatures (Ta, 10°C, 24°C, and 32°C). We found that orexin injection into the RPa variably affected the wake-sleep states, autonomic functions, motor activity, and feeding behavior, at the different Tas. In particular, in the first post-injection hour, we observed an increase in wakefulness, which was large at Ta 24°C and Ta 10°C and rather mild at Ta 32°C. Deep brain temperature was increased by orexin injection at Ta 10°C, but not at either Ta 24°C or Ta 32°C. Moreover, an increase in mean arterial blood pressure occurred at Ta 24°C, which was probably masked by the high baseline levels at Ta 10°C and was completely absent at Ta 32°C. Finally, an enhancement in feeding behavior was observed at Ta 24°C and 10°C only. In accordance with what observed in anaesthetized rats, orexinergic signalling in the RPa seems to be ineffective in the absence of any thermogenic drive. Moreover, the effects observed on the wake-sleep states and feeding behavior introduce the RPa as a novel player in the central neural network promoting wakefulness and feeding.
Collapse
Affiliation(s)
- Timna Hitrec
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Flavia Del Vecchio
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Luca Alberti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marco Luppi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Davide Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandra Occhinegro
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Emiliana Piscitiello
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Ludovico Taddei
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Domenico Tupone
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR, United States
| | - Roberto Amici
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Matteo Cerri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Italian Institute of Technology (IIT), Genova, Italy
- National Institute of Nuclear Physics of Bologna, Bologna, Italy
| |
Collapse
|
49
|
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
|
50
|
Thorpy MJ, Siegel JM, Dauvilliers Y. REM sleep in narcolepsy. Sleep Med Rev 2024; 77:101976. [PMID: 39186901 DOI: 10.1016/j.smrv.2024.101976] [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: 11/16/2023] [Revised: 06/28/2024] [Accepted: 07/09/2024] [Indexed: 08/28/2024]
Abstract
Narcolepsy is mainly associated with excessive daytime sleepiness, but the characteristic feature is abnormal rapid eye movement (REM) sleep phenomena. REM sleep disturbances can manifest as cataplexy (in narcolepsy type 1), sleep paralysis, sleep-related hallucinations, REM sleep behavior disorder, abnormal dreams, polysomnographic evidence of REM sleep disruption with sleep-onset REM periods, and fragmented REM sleep. Characterization of REM sleep and related symptoms facilitates the differentiation of narcolepsy from other central hypersomnolence disorders and aids in distinguishing between narcolepsy types 1 and 2. A circuit comprising regions within the brainstem, forebrain, and hypothalamus is involved in generating and regulating REM sleep, which is influenced by changes in monoamines, acetylcholine, and neuropeptides. REM sleep is associated with brainstem functions, including autonomic control, and REM sleep disturbances may be associated with increased cardiovascular risk. Medications used to treat narcolepsy (and REM-related symptoms of narcolepsy) include stimulants/wake-promoting agents, pitolisant, oxybates, and antidepressants; hypocretin agonists are a potential new class of therapeutics. The role of REM sleep disturbances in narcolepsy remains an area of active research in pathophysiology, symptom management, and treatment. This review summarizes the current understanding of the role of REM sleep and its dysfunction in narcolepsy.
Collapse
Affiliation(s)
| | - Jerome M Siegel
- Department of Psychiatry and Brain Research Institute, University of California, Los Angeles, CA, USA; Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Yves Dauvilliers
- Sleep and Wake Disorders Centre, Department of Neurology, Gui de Chauliac Hospital, University Montpellier, INSERM INM, France
| |
Collapse
|