1
|
Yokoi S, Suno R, Mitsutake A. Structural and Computational Insights into Dynamics and Intermediate States of Orexin 2 Receptor Signaling. J Phys Chem B 2024; 128:6082-6096. [PMID: 38722794 DOI: 10.1021/acs.jpcb.4c00730] [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: 06/28/2024]
Abstract
Orexin 2 receptor (OX2R) is a G protein-coupled receptor (GPCR) whose activation is crucial to regulation of the sleep-wake cycle. Recently, inactive and active state structures were determined from X-ray crystallography and cryo-electron microscopy single particle analysis, and the activation mechanisms have been discussed based on these static data. GPCRs have multiscale intermediate states during activation, and insights into these dynamics and intermediate states may aid the precise control of intracellular signaling by ligands in drug discovery. Molecular dynamics (MD) simulations are used to investigate dynamics induced in response to thermal perturbations, such as structural fluctuations of main and side chains. In this study, we proposed collective motions of the TM domain during activation by performing 30 independent microsecond-scale MD simulations for various OX2R systems and applying relaxation mode analysis. The analysis results suggested that TM3 had a vertical structural movement relative to the membrane surface during activation. In addition, we extracted three characteristic amino acid residues on TM3, i.e., Q1343.32, V1423.40, and R1523.50, which exhibited large conformational fluctuations. We quantitatively evaluated the changes in their equilibrium during activation in relation to the movement of TM3. We also discuss the regulation of ligand binding recognition and intracellular signal selectivity by changes in the equilibrium of Q1343.32 and R1523.50, respectively, according to MD simulations and GPCR database. Additionally, the OX2R-Gi signaling complex is stabilized in the conformation resembling a non-canonical (NC) state, which was previously proposed as an intermediate state during activation of neurotensin 1 receptor. Insights into the dynamics and intermediate states during activation gained from this study may be useful for developing biased agonists for OX2R.
Collapse
Affiliation(s)
- Shun Yokoi
- Department of Physics, School of Science and Technology, Meiji University, 1-1-1 Higashi-Mita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Ryoji Suno
- Department of Medical Chemistry, Kansai Medical University, 2-5-1 Shin-Machi, Hirakata, Osaka 573-1010, Japan
| | - Ayori Mitsutake
- Department of Physics, School of Science and Technology, Meiji University, 1-1-1 Higashi-Mita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| |
Collapse
|
2
|
Bandarabadi M, Li S, Aeschlimann L, Colombo G, Tzanoulinou S, Tafti M, Becchetti A, Boutrel B, Vassalli A. Inactivation of hypocretin receptor-2 signaling in dopaminergic neurons induces hyperarousal and enhanced cognition but impaired inhibitory control. Mol Psychiatry 2024; 29:327-341. [PMID: 38123729 PMCID: PMC11116111 DOI: 10.1038/s41380-023-02329-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023]
Abstract
Hypocretin/Orexin (HCRT/OX) and dopamine (DA) are both key effectors of salience processing, reward and stress-related behaviors and motivational states, yet their respective roles and interactions are poorly delineated. We inactivated HCRT-to-DA connectivity by genetic disruption of Hypocretin receptor-1 (Hcrtr1), Hypocretin receptor-2 (Hcrtr2), or both receptors (Hcrtr1&2) in DA neurons and analyzed the consequences on vigilance states, brain oscillations and cognitive performance in freely behaving mice. Unexpectedly, loss of Hcrtr2, but not Hcrtr1 or Hcrtr1&2, induced a dramatic increase in theta (7-11 Hz) electroencephalographic (EEG) activity in both wakefulness and rapid-eye-movement sleep (REMS). DAHcrtr2-deficient mice spent more time in an active (or theta activity-enriched) substate of wakefulness, and exhibited prolonged REMS. Additionally, both wake and REMS displayed enhanced theta-gamma phase-amplitude coupling. The baseline waking EEG of DAHcrtr2-deficient mice exhibited diminished infra-theta, but increased theta power, two hallmarks of EEG hyperarousal, that were however uncoupled from locomotor activity. Upon exposure to novel, either rewarding or stress-inducing environments, DAHcrtr2-deficient mice featured more pronounced waking theta and fast-gamma (52-80 Hz) EEG activity surges compared to littermate controls, further suggesting increased alertness. Cognitive performance was evaluated in an operant conditioning paradigm, which revealed that DAHcrtr2-ablated mice manifest faster task acquisition and higher choice accuracy under increasingly demanding task contingencies. However, the mice concurrently displayed maladaptive patterns of reward-seeking, with behavioral indices of enhanced impulsivity and compulsivity. None of the EEG changes observed in DAHcrtr2-deficient mice were seen in DAHcrtr1-ablated mice, which tended to show opposite EEG phenotypes. Our findings establish a clear genetically-defined link between monosynaptic HCRT-to-DA neurotransmission and theta oscillations, with a differential and novel role of HCRTR2 in theta-gamma cross-frequency coupling, attentional processes, and executive functions, relevant to disorders including narcolepsy, attention-deficit/hyperactivity disorder, and Parkinson's disease.
Collapse
Affiliation(s)
- Mojtaba Bandarabadi
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Sha Li
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Lea Aeschlimann
- Centre for Psychiatric Neuroscience, Department of Psychiatry, The Lausanne University Hospital, Lausanne, Switzerland
| | - Giulia Colombo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | | | - Mehdi Tafti
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Benjamin Boutrel
- Centre for Psychiatric Neuroscience, Department of Psychiatry, The Lausanne University Hospital, Lausanne, Switzerland
| | - Anne Vassalli
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
3
|
Tsuneoka Y, Funato H. Whole Brain Mapping of Orexin Receptor mRNA Expression Visualized by Branched In Situ Hybridization Chain Reaction. eNeuro 2024; 11:ENEURO.0474-23.2024. [PMID: 38199807 PMCID: PMC10883752 DOI: 10.1523/eneuro.0474-23.2024] [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: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024] Open
Abstract
Orexins, which are produced within neurons of the lateral hypothalamic area, play a pivotal role in the regulation of various behaviors, including sleep/wakefulness, reward behavior, and energy metabolism, via orexin receptor type 1 (OX1R) and type 2 (OX2R). Despite the advanced understanding of orexinergic regulation of behavior at the circuit level, the precise distribution of orexin receptors in the brain remains unknown. Here, we develop a new branched in situ hybridization chain reaction (bHCR) technique to visualize multiple target mRNAs in a semiquantitative manner, combined with immunohistochemistry, which provided comprehensive distribution of orexin receptor mRNA and neuron subtypes expressing orexin receptors in mouse brains. Only a limited number of cells expressing both Ox1r and Ox2r were observed in specific brain regions, such as the dorsal raphe nucleus and ventromedial hypothalamic nucleus. In many brain regions, Ox1r-expressing cells and Ox2r-expressing cells belong to different cell types, such as glutamatergic and GABAergic neurons. Moreover, our findings demonstrated considerable heterogeneity in Ox1r- or Ox2r-expressing populations of serotonergic, dopaminergic, noradrenergic, cholinergic, and histaminergic neurons. The majority of orexin neurons did not express orexin receptors. This study provides valuable insights into the mechanism underlying the physiological and behavioral regulation mediated by the orexin system, as well as the development of therapeutic agents targeting orexin receptors.
Collapse
Affiliation(s)
- Yousuke Tsuneoka
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo 145-854, Japan
| | - Hiromasa Funato
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo 145-854, Japan
- International Institutes for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
4
|
Bonifazi A, Del Bello F, Giorgioni G, Piergentili A, Saab E, Botticelli L, Cifani C, Micioni Di Bonaventura E, Micioni Di Bonaventura MV, Quaglia W. Targeting orexin receptors: Recent advances in the development of subtype selective or dual ligands for the treatment of neuropsychiatric disorders. Med Res Rev 2023; 43:1607-1667. [PMID: 37036052 DOI: 10.1002/med.21959] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/08/2023] [Accepted: 03/28/2023] [Indexed: 04/11/2023]
Abstract
Orexin-A and orexin-B, also named hypocretin-1 and hypocretin-2, are two hypothalamic neuropeptides highly conserved across mammalian species. Their effects are mediated by two distinct G protein-coupled receptors, namely orexin receptor type 1 (OX1-R) and type 2 (OX2-R), which share 64% amino acid identity. Given the wide expression of OX-Rs in different central nervous system and peripheral areas and the several pathophysiological functions in which they are involved, including sleep-wake cycle regulation (mainly mediated by OX2-R), emotion, panic-like behaviors, anxiety/stress, food intake, and energy homeostasis (mainly mediated by OX1-R), both subtypes represent targets of interest for many structure-activity relationship (SAR) campaigns carried out by pharmaceutical companies and academies. However, before 2017 the research was predominantly directed towards dual-orexin ligands, and limited chemotypes were investigated. Analytical characterizations, including resolved structures for both OX1-R and OX2-R in complex with agonists and antagonists, have improved the understanding of the molecular basis of receptor recognition and are assets for medicinal chemists in the design of subtype-selective ligands. This review is focused on the medicinal chemistry aspects of small molecules acting as dual or subtype selective OX1-R/OX2-R agonists and antagonists belonging to different chemotypes and developed in the last years, including radiolabeled OX-R ligands for molecular imaging. Moreover, the pharmacological effects of the most studied ligands in different neuropsychiatric diseases, such as sleep, mood, substance use, and eating disorders, as well as pain, have been discussed. Poly-pharmacology applications and multitarget ligands have also been considered.
Collapse
Affiliation(s)
- Alessandro Bonifazi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, Maryland, United States
| | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Camerino, Italy
| | - Gianfabio Giorgioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Camerino, Italy
| | | | - Elizabeth Saab
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, Maryland, United States
| | - Luca Botticelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | | | | | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Camerino, Italy
| |
Collapse
|
5
|
Lucey BP, Liu H, Toedebusch CD, Freund D, Redrick T, Chahin SL, Mawuenyega KG, Bollinger JG, Ovod V, Barthélemy NR, Bateman RJ. Suvorexant Acutely Decreases Tau Phosphorylation and Aβ in the Human CNS. Ann Neurol 2023; 94:27-40. [PMID: 36897120 PMCID: PMC10330114 DOI: 10.1002/ana.26641] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/11/2023] [Accepted: 03/08/2023] [Indexed: 03/11/2023]
Abstract
OBJECTIVE In Alzheimer's disease, hyperphosphorylated tau is associated with formation of insoluble paired helical filaments that aggregate as neurofibrillary tau tangles and are associated with neuronal loss and cognitive symptoms. Dual orexin receptor antagonists decrease soluble amyloid-β levels and amyloid plaques in mouse models overexpressing amyloid-β, but have not been reported to affect tau phosphorylation. In this randomized controlled trial, we tested the acute effect of suvorexant, a dual orexin receptor antagonist, on amyloid-β, tau, and phospho-tau. METHODS Thirty-eight cognitively unimpaired participants aged 45 to 65 years were randomized to placebo (N = 13), suvorexant 10 mg (N = 13), and suvorexant 20 mg (N = 12). Six milliliters of cerebrospinal fluid were collected via an indwelling lumbar catheter every 2 hours for 36 hours starting at 20:00. Participants received placebo or suvorexant at 21:00. All samples were processed and measured for multiple forms of amyloid-β, tau, and phospho-tau via immunoprecipitation and liquid chromatography-mass spectrometry. RESULTS The ratio of phosphorylated-tau-threonine-181 to unphosphorylated-tau-threonine-181, a measure of phosphorylation at this tau phosphosite, decreased ~10% to 15% in participants treated with suvorexant 20 mg compared to placebo. However, phosphorylation at tau-serine-202 and tau-threonine-217 were not decreased by suvorexant. Suvorexant decreased amyloid-β ~10% to 20% compared to placebo starting 5 hours after drug administration. INTERPRETATION In this study, suvorexant acutely decreased tau phosphorylation and amyloid-β concentrations in the central nervous system. Suvorexant is approved by the US Food and Drug Administration to treatment insomnia and may have potential as a repurposed drug for the prevention of Alzheimer's disease, however, future studies with chronic treatment are needed. ANN NEUROL 2023;94:27-40.
Collapse
Affiliation(s)
- Brendan P. Lucey
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
- Center on Biological Rhythms and Sleep, Washington University School of Medicine, St Louis, MO
| | - Haiyan Liu
- Department of Neurology, Washington University School of Medicine, St Louis, MO
| | | | - David Freund
- Department of Neurology, Washington University School of Medicine, St Louis, MO
| | - Tiara Redrick
- Department of Neurology, Washington University School of Medicine, St Louis, MO
| | - Samir L. Chahin
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
| | - Kwasi G. Mawuenyega
- Biomolecular Analytical Research and Development, MilliporeSigma, St Louis, MO
| | - James G. Bollinger
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
| | - Vitaliy Ovod
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
| | - Nicolas R. Barthélemy
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
| | - Randall J. Bateman
- Department of Neurology, Washington University School of Medicine, St Louis, MO
- Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
6
|
Kukkonen JP. The G protein preference of orexin receptors is currently an unresolved issue. Nat Commun 2023; 14:3162. [PMID: 37264034 PMCID: PMC10235037 DOI: 10.1038/s41467-023-38764-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 05/15/2023] [Indexed: 06/03/2023] Open
Affiliation(s)
- Jyrki P Kukkonen
- Department of Pharmacology, Institute of Biomedicine, Faculty of Medicine, University of Helsinki, POB 63, FI-00014, Helsinki, Finland.
| |
Collapse
|
7
|
Ball HL, Said H, Chapman K, Fu R, Xiong Y, Burk JA, Rosenbaum D, Veneziano R, Cotten ML. Orexin A, an amphipathic α-helical neuropeptide involved in pleiotropic functions in the nervous and immune systems: Synthetic approach and biophysical studies of the membrane-bound state. Biophys Chem 2023; 297:107007. [PMID: 37037119 DOI: 10.1016/j.bpc.2023.107007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/11/2023] [Accepted: 03/12/2023] [Indexed: 03/16/2023]
Abstract
This research reports on the membrane interactions of orexin A (OXA), an α-helical and amphipathic neuropeptide that contains 33 residues and two disulfide bonds in the N-terminal region. OXA, which activates the orexins 1 and 2 receptors in neural and immune cell membranes, has essential pleiotropic physiological effects, including at the levels of arousal, sleep/wakefulness, energy balance, neuroprotection, lipid signaling, the inflammatory response, and pain. As a result, the orexin system has become a prominent target to treat diseases such as sleep disorders, drug addiction, and inflammation. While the high-resolution structure of OXA has been investigated in water and bound to micelles, there is a lack of information about its conformation bound to phospholipid membranes and its receptors. NMR is a powerful method to investigate peptide structures in a membrane environment. To facilitate the NMR structural studies of OXA exposed to membranes, we present a novel synthetic scheme, leading to the production of isotopically-labeled material at high purity. A receptor activation assay shows that the 15N-labeled peptide is biologically active. Biophysical studies are performed using surface plasmon resonance, circular dichroism, and NMR to investigate the interactions of OXA with phospholipid bilayers. The results demonstrate a strong interaction between the peptide and phospholipids, an increase in α-helical content upon membrane binding, and an in-plane orientation of the C-terminal region critical to function. This new knowledge about structure-activity relationships in OXA could inspire the design of novel therapeutics that leverage the anti-inflammatory and neuro-protective functions of OXA, and therefore could help address neuroinflammation, a major issue associated with neurological disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Haydn L Ball
- Department of Chemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hooda Said
- Department of Bioengineering, College of Engineering and Computing, George Mason University, Fairfax, VA 22030, USA
| | - Karen Chapman
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Riqiang Fu
- National High Magnetic Field Laboratory, Tallahassee, FL 32310, USA
| | - Yawei Xiong
- Department of Applied Science, William & Mary, Williamsburg, VA 23185, USA
| | - Joshua A Burk
- Department of Psychological Sciences, William & Mary, Williamsburg, VA 23185, USA
| | - Daniel Rosenbaum
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Remi Veneziano
- Department of Bioengineering, College of Engineering and Computing, George Mason University, Fairfax, VA 22030, USA
| | - Myriam L Cotten
- Department of Applied Science, William & Mary, Williamsburg, VA 23185, USA.
| |
Collapse
|
8
|
Chatterjee O, Gopalakrishnan L, Pullimamidi D, Raj C, Yelamanchi S, Gangadharappa BS, Nair B, Mahadevan A, Raju R, Keshava Prasad TS. A molecular network map of orexin-orexin receptor signaling system. J Cell Commun Signal 2023; 17:217-227. [PMID: 36480100 PMCID: PMC10030760 DOI: 10.1007/s12079-022-00700-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/04/2022] [Accepted: 09/15/2022] [Indexed: 12/13/2022] Open
Abstract
Orexins are excitatory neuropeptides, which are predominantly associated with feeding behavior, sleep-wake cycle and energy homeostasis. The orexinergic system comprises of HCRTR1 and HCRTR2, G-protein-coupled receptors of rhodopsin family and the endogenous ligands processed from HCRT pro-hormone, Orexin A and Orexin B. These neuropeptides are biosynthesized by the orexin neurons present in the lateral hypothalamus area, with dense projections to other brain regions. The orexin-receptor signaling is implicated in various metabolic as well as neurological disorders, making it a promising target for pharmacological interventions. However, there is limited information available on the collective representation of the signal transduction pathways pertaining to the orexin-orexin receptor signaling system. Here, we depict a compendium of the Orexin A/B stimulated reactions in the form of a basic signaling pathway map. This map catalogs the reactions into five categories: molecular association, activation/inhibition, catalysis, transport, and gene regulation. A total of 318 downstream molecules were annotated adhering to the guidelines of NetPath curation. This pathway map can be utilized for further assessment of signaling events associated with orexin-mediated physiological functions and is freely available on WikiPathways, an open-source pathway database ( https://www.wikipathways.org/index.php/Pathway:WP5094 ).
Collapse
Affiliation(s)
- Oishi Chatterjee
- Institute of Bioinformatics, International Tech Park, 560 066, Bangalore, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, 690 525, Kollam, India
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), 575 018, Mangalore, India
| | - Lathika Gopalakrishnan
- Institute of Bioinformatics, International Tech Park, 560 066, Bangalore, India
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), 575 018, Mangalore, India
- Manipal Academy of Higher Education (MAHE), 576 104, Manipal, India
| | | | - Chinmayi Raj
- Institute of Bioinformatics, International Tech Park, 560 066, Bangalore, India
| | - Soujanya Yelamanchi
- Institute of Bioinformatics, International Tech Park, 560 066, Bangalore, India
| | | | - Bipin Nair
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, 690 525, Kollam, India
| | - Anita Mahadevan
- Human Brain Tissue Repository, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, 560 029, Bangalore, India
- Department of Neuropathology, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, 560 029, Bangalore, India
| | - Rajesh Raju
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), 575 018, Mangalore, India.
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), 575 018, Mangalore, India.
| |
Collapse
|
9
|
Knez R, Stevanovic D, Fernell E, Gillberg C. Orexin/Hypocretin System Dysfunction in ESSENCE (Early Symptomatic Syndromes Eliciting Neurodevelopmental Clinical Examinations). Neuropsychiatr Dis Treat 2022; 18:2683-2702. [PMID: 36411777 PMCID: PMC9675327 DOI: 10.2147/ndt.s358373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022] Open
Abstract
Early Symptomatic Syndromes Eliciting Neurodevelopmental Clinical Examinations (ESSENCE) is an umbrella term covering a wide range of neurodevelopmental difficulties and disorders. Thus, ESSENCE includes attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorder (ASD), and other neurodevelopmental disorders (NDDs) and difficulties, with a variety of symptoms in cognitive, motor, sensory, social, arousal, regulatory, emotional, and behavioral developmental domains, frequently co-occurring and likely having partly common neurobiological substrates. The ESSENCE concept is a clinical paradigm that promotes organizing NDDs in everyday clinical practice according to their coexistence, symptom dimensions overlapping, and treatment possibilities. Despite increased knowledge regarding NDDs, the neurobiological mechanisms that underlie them and other ESSENCE-related problems, are not well understood. With its wide range of neural circuits and interactions with numerous neurotransmitters, the orexin/hypocretin system (Orx-S) is possibly associated with a variety of neurocognitive, psychobiological, neuroendocrine, and physiological functions and behaviors. Dysfunction of Orx-S has been implicated in various psychiatric and neurological disorders. This article provides an overview of Orx-S dysfunctions' possible involvement in the development, presentation, and maintenance of ESSENCE. We provide a focused review of current research evidence linking orexin neuropeptides with specific clinical NDDs symptoms, mostly in ADHD and ASD, within the Research Domain Criteria (RDoC) framework. We propose that Orx-S dysfunction might have an important role in some of these neurodevelopmental symptom domains, such as arousal, wakefulness, sleep, motor and sensory processing, mood and emotional regulation, fear processing, reward, feeding, attention, executive functions, and sociability. Our perspective is presented from a clinical point of view. Further, more thorough systematic reviews are needed as well as planning of extensive new research into the Orx-S's role in ESSENCE, especially considering RDoC elements.
Collapse
Affiliation(s)
- Rajna Knez
- Gillberg Neuropsychiatry Centre, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, Skaraborg Hospital, Skövde, Sweden
- School of Health Sciences, University of Skövde, Skövde, Sweden
| | - Dejan Stevanovic
- Gillberg Neuropsychiatry Centre, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elisabeth Fernell
- Gillberg Neuropsychiatry Centre, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christopher Gillberg
- Gillberg Neuropsychiatry Centre, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
10
|
Raïch I, Rebassa JB, Lillo J, Cordomi A, Rivas-Santisteban R, Lillo A, Reyes-Resina I, Franco R, Navarro G. Antagonization of OX 1 Receptor Potentiates CB 2 Receptor Function in Microglia from APP Sw/Ind Mice Model. Int J Mol Sci 2022; 23:12801. [PMID: 36361598 PMCID: PMC9656664 DOI: 10.3390/ijms232112801] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/04/2022] [Accepted: 10/14/2022] [Indexed: 08/01/2023] Open
Abstract
Microdialysis assays demonstrated a possible role of orexin in the regulation of amyloid beta peptide (Aß) levels in the hippocampal interstitial fluid in the APP transgenic model. CB2R is overexpressed in activated microglia, showing a neuroprotective effect. These two receptors may interact, forming CB2-OX1-Hets and becoming a new target to combat Alzheimer's disease. Aims: Demonstrate the potential role of CB2-OX1-Hets expression and function in microglia from animal models of Alzheimer's disease. Receptor heteromer expression was detected by immunocytochemistry, bioluminescence resonance energy transfer (BRET) and proximity ligation assay (PLA) in transfected HEK-293T cells and microglia primary cultures. Quantitation of signal transduction events in a heterologous system and in microglia cells was performed using the AlphaScreen® SureFire® kit, western blot, the GCaMP6 calcium sensor and the Lance Ultra cAMP kit (PerkinElmer). The formation of CB2-OX1 receptor complexes in transfected HEK-293T cells has been demonstrated. The tetrameric complex is constituted by one CB2R homodimer, one OX1R homodimer and two G proteins, a Gi and a Gq. The use of TAT interfering peptides showed that the CB2-OX1 receptor complex interface is TM4-TM5. At the functional level it has been observed that the OX1R antagonist, SB334867, potentiates the action induced by CB2R agonist JWH133. This effect is observed in transfected HEK-293T cells and microglia, and it is stronger in the Alzheimer's disease (AD) animal model APPSw/Ind where the expression of the complex assessed by the proximity ligation assay indicates an increase in the number of complexes compared to resting microglia. The CB2-OX1 receptor complex is overexpressed in microglia from AD animal models where OX1R antagonists potentiate the neuroprotective actions of CB2R activation. Taken together, these results point to OX1R antagonists as drugs with therapeutic potential to combat AD. Data access statement: Raw data will be provided by the corresponding author upon reasonable requirement.
Collapse
Affiliation(s)
- Iu Raïch
- Molecular Neuropharmacology Laboratory, Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, 08007 Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, 28029 Madrid, Spain
- Neurosciences Institut, University of Barcelona (NeuroUB), 08028 Barcelona, Spain
| | - Joan Biel Rebassa
- Molecular Neuropharmacology Laboratory, Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, 08007 Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, 28029 Madrid, Spain
- Neurosciences Institut, University of Barcelona (NeuroUB), 08028 Barcelona, Spain
| | - Jaume Lillo
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, 28029 Madrid, Spain
- Neurosciences Institut, University of Barcelona (NeuroUB), 08028 Barcelona, Spain
| | | | - Rafael Rivas-Santisteban
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, 28029 Madrid, Spain
- Neurosciences Institut, University of Barcelona (NeuroUB), 08028 Barcelona, Spain
| | - Alejandro Lillo
- Molecular Neuropharmacology Laboratory, Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, 08007 Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, 28029 Madrid, Spain
- Neurosciences Institut, University of Barcelona (NeuroUB), 08028 Barcelona, Spain
| | - Irene Reyes-Resina
- Molecular Neuropharmacology Laboratory, Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, 08007 Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, 28029 Madrid, Spain
- Neurosciences Institut, University of Barcelona (NeuroUB), 08028 Barcelona, Spain
| | - Rafael Franco
- Molecular Neuropharmacology Laboratory, Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, 08007 Barcelona, Spain
- School of Chemistry, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Gemma Navarro
- Molecular Neuropharmacology Laboratory, Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, 08007 Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, 28029 Madrid, Spain
- Neurosciences Institut, University of Barcelona (NeuroUB), 08028 Barcelona, Spain
| |
Collapse
|
11
|
López JM, Carballeira P, Pozo J, León-Espinosa G, Muñoz A. Hypothalamic orexinergic neuron changes during the hibernation of the Syrian hamster. Front Neuroanat 2022; 16:993421. [PMID: 36157325 PMCID: PMC9501701 DOI: 10.3389/fnana.2022.993421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
Hibernation in small mammals is a highly regulated process with periods of torpor involving drops in body temperature and metabolic rate, as well as a general decrease in neural activity, all of which proceed alongside complex brain adaptive changes that appear to protect the brain from extreme hypoxia and low temperatures. All these changes are rapidly reversed, with no apparent brain damage occurring, during the short periods of arousal, interspersed during torpor—characterized by transitory and partial rewarming and activity, including sleep activation, and feeding in some species. The orexins are neuropeptides synthesized in hypothalamic neurons that project to multiple brain regions and are known to participate in the regulation of a variety of processes including feeding behavior, the sleep-wake cycle, and autonomic functions such as brown adipose tissue thermogenesis. Using multiple immunohistochemical techniques and quantitative analysis, we have characterized the orexinergic system in the brain of the Syrian hamster—a facultative hibernator. Our results revealed that orexinergic neurons in this species consisted of a neuronal population restricted to the lateral hypothalamic area, whereas orexinergic fibers distribute throughout the rostrocaudal extent of the brain, particularly innervating catecholaminergic and serotonergic neuronal populations. We characterized the changes of orexinergic cells in the different phases of hibernation based on the intensity of immunostaining for the neuronal activity marker C-Fos and orexin A (OXA). During torpor, we found an increase in C-Fos immunostaining intensity in orexinergic neurons, accompanied by a decrease in OXA immunostaining. These changes were accompanied by a volume reduction and a fragmentation of the Golgi apparatus (GA) as well as a decrease in the colocalization of OXA and the GA marker GM-130. Importantly, during arousal, C-Fos and OXA expression in orexinergic neurons was highest and the structural appearance and the volume of the GA along with the colocalization of OXA/GM-130 reverted to euthermic levels. We discuss the involvement of orexinergic cells in the regulation of mammalian hibernation and, in particular, the possibility that the high activation of orexinergic cells during the arousal stage guides the rewarming as well as the feeding and sleep behaviors characteristic of this phase.
Collapse
Affiliation(s)
- Jesús M. López
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
| | - Paula Carballeira
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
| | - Javier Pozo
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
| | - Gonzalo León-Espinosa
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-Centro de Estudios Universitarios (CEU), Madrid, Spain
| | - Alberto Muñoz
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica (CTB), Universidad Politécnica de Madrid, Madrid, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- *Correspondence: Alberto Muñoz,
| |
Collapse
|
12
|
Dale NC, Hoyer D, Jacobson LH, Pfleger KDG, Johnstone EKM. Orexin Signaling: A Complex, Multifaceted Process. Front Cell Neurosci 2022; 16:812359. [PMID: 35496914 PMCID: PMC9044999 DOI: 10.3389/fncel.2022.812359] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/07/2022] [Indexed: 11/15/2022] Open
Abstract
The orexin system comprises two G protein-coupled receptors, OX1 and OX2 receptors (OX1R and OX2R, respectively), along with two endogenous agonists cleaved from a common precursor (prepro-orexin), orexin-A (OX-A) and orexin-B (OX-B). For the receptors, a complex array of signaling behaviors has been reported. In particular, it becomes obvious that orexin receptor coupling is very diverse and can be tissue-, cell- and context-dependent. Here, the early signal transduction interactions of the orexin receptors will be discussed in depth, with particular emphasis on the direct G protein interactions of each receptor. In doing so, it is evident that ligands, additional receptor-protein interactions and cellular environment all play important roles in the G protein coupling profiles of the orexin receptors. This has potential implications for our understanding of the orexin system's function in vivo in both central and peripheral environments, as well as the development of novel agonists, antagonists and possibly allosteric modulators targeting the orexin system.
Collapse
Affiliation(s)
- Natasha C. Dale
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, WA, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, WA, Australia
| | - Daniel Hoyer
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Laura H. Jacobson
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Kevin D. G. Pfleger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, WA, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, WA, Australia
- Dimerix Limited, Nedlands, WA, Australia
| | - Elizabeth K. M. Johnstone
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, WA, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
13
|
Ramser A, Dridi S. Avian Orexin: Feed Intake Regulator or Something Else? Vet Sci 2022; 9:vetsci9030112. [PMID: 35324840 PMCID: PMC8950792 DOI: 10.3390/vetsci9030112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
Originally named for its expression in the posterior hypothalamus in rats and after the Greek word for “appetite”, hypocretin, or orexin, as it is known today, gained notoriety as a neuropeptide regulating feeding behavior, energy homeostasis, and sleep. Orexin has been proven to be involved in both central and peripheral control of neuroendocrine functions, energy balance, and metabolism. Since its discovery, its ability to increase appetite as well as regulate feeding behavior has been widely explored in mammalian food production animals such as cattle, pigs, and sheep. It is also linked to neurological disorders, leading to its intensive investigation in humans regarding narcolepsy, depression, and Alzheimer’s disease. However, in non-mammalian species, research is limited. In the case of avian species, orexin has been shown to have no central effect on feed-intake, however it was found to be involved in muscle energy metabolism and hepatic lipogenesis. This review provides current knowledge and summarizes orexin’s physiological roles in livestock and pinpoints the present lacuna to facilitate further investigations.
Collapse
Affiliation(s)
- Alison Ramser
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
- Cell and Molecular Biology Program, Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Sami Dridi
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA;
- Cell and Molecular Biology Program, Department of Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
- Correspondence: ; Tel.: +1-(479)-575-2583; Fax: +1-(479)-575-7139
| |
Collapse
|
14
|
Orexin receptor blockers: A tool for lowering alcohol intake and alcohol addictive behavior in the light of preclinical studies. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Alcohol use disorder (AUD) is a severe and globally widespread neurological and psychiatric problem. The treatment with currently used drugs often does not bring the expected effect. New optimization methods or directions in pharmacotherapy are still being sought. The group of bioactive ligands, targeted at neuropeptides called orexins (OXs) and their receptors (OXRs), affects a number of functions including ingestion, sleep-wake regulation, as well as the brain reward system which is the basis of addiction.
The purpose of this paper is to systematize the knowledge in the field of preclinical behavioral studies on rodents (rats and mice) in several models of alcohol consumption using the OXRs antagonists.
The results of the experiments indicated a potential efficacy of particular OXRs antagonists in the AUD treatment, especially those selectively blocking the OX1R. Among them, SB-334867 in the lowest effective dose of 3 mg/kg i.p. was most studied, as shown in the model of two-bottle choice using C57BL/6 mice. Moreover, this compound did not affect the reduction of cognitive functions. GSK1059865 was also involved in the selective reduction of ethanol intake, and simultaneously did not alter the consumption of sugar solution. The other group of selective OX2R antagonists, such as TCS-OX2-29 and LSN2424100, was less efficient.
In summary, the OX1R antagonists proved to have the potential in AUD therapy, not only through the reduction of ethanol consumption but also in the treatment of coexisting behavioral and physiological disorders, such as insomnia and anxiety.
Collapse
|
15
|
Coleman P, de Lecea L, Gotter A, Hagan J, Hoyer D, Kilduff T, Kukkonen JP, Porter R, Renger J, Siegel JM, Sutcliffe G, Upton N, Winrow CJ. Orexin receptors in GtoPdb v.2021.3. IUPHAR/BPS GUIDE TO PHARMACOLOGY CITE 2021; 2021. [PMID: 34927075 DOI: 10.2218/gtopdb/f51/2021.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Orexin receptors (nomenclature as agreed by the NC-IUPHAR Subcommittee on Orexin receptors [42]) are activated by the endogenous polypeptides orexin-A and orexin-B (also known as hypocretin-1 and -2; 33 and 28 aa) derived from a common precursor, preproorexin or orexin precursor, by proteolytic cleavage and some typical peptide modifications [109]. Currently the only orexin receptor ligands in clinical use are suvorexant and lemborexant, which are used as hypnotics. Orexin receptor crystal structures have been solved [134, 133, 54, 117, 46].
Collapse
|
16
|
Hsu CW, Wang S. Changes in the Orexin System in Rats Exhibiting Learned Helplessness Behaviors. Brain Sci 2021; 11:brainsci11121634. [PMID: 34942932 PMCID: PMC8699801 DOI: 10.3390/brainsci11121634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022] Open
Abstract
Orexin-A (OX-A) and orexin-B (OX-B) are neuropeptides produced in the hypothalamus. Preclinical and clinical studies suggest that depression and anxiety are associated with the orexin system. In the current study, we used the learned helplessness (LH) animal model of depression to identify rats displaying LH behaviors (LH rats) and those that did not (No-LH rats). We compared the number of orexin-containing neurons in the hypothalamus of LH, No-LH, and control rats. Orexin peptides, orexin receptor 1 (OXR1) and 2 (OXR2) in brain areas involved in major depression and serum OX-A and corticosterone (CORT) concentrations were quantified and compared between rat groups. We found that LH and No-LH rats displayed higher serum OX-A concentrations compared with control rats. Comparison between LH and No-LH rats revealed that No-LH rats had significantly higher OX-A levels in the brain, more OX-A neurons, and more OX-A neuron activation. LH rats had more OX-B neurons and more OX-B neuron activation. Orexin peptides and receptors in the brain areas involved in major depression exhibited different patterns in LH and NoLH rats. Our findings revealed that activation of OX-A neurons could promote resilient behaviors under stressful situations and OX-A and OX-B neuropeptides exhibit dissimilar functions in LH behaviors.
Collapse
|
17
|
Salaberry NL, Mendoza J. The circadian clock in the mouse habenula is set by catecholamines. Cell Tissue Res 2021; 387:261-274. [PMID: 34816282 DOI: 10.1007/s00441-021-03557-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 11/12/2021] [Indexed: 12/15/2022]
Abstract
Circadian rhythms are those variations in behavioral and molecular processes of organisms that follow roughly 24 h cycles in the absence of any external cue. The hypothalamic suprachiasmatic nucleus (SCN) harbors the principal brain pacemaker driving circadian rhythms. The epithalamic habenula (Hb) contains a self-sustained circadian clock functionally coupled to the SCN. Anatomically, the Hb projects to the midbrain dopamine (DA) and serotonin (5-HT) systems, and it receives inputs from the forebrain, midbrain, and brainstem. The SCN is set by internal signals such as 5-HT or melatonin from the raphe nuclei and pineal gland, respectively. However, how the Hb clock is set by internal cues is not well characterized. Hence, in the present study, we determined whether DA, noradrenaline (NA), 5-HT, and the neuropeptides orexin (ORX) and vasopressin influence the Hb circadian clock. Using PER2::Luciferase transgenic mice, we found that the amplitude of the PER2 protein circadian oscillations from Hb explants was strongly affected by DA and NA. Importantly, these effects were dose-and region (rostral vs. caudal) dependent for NA, with a main effect in the caudal part of the Hb. Furthermore, ORX also induced a significant change in the amplitude of PER2 protein oscillations in the caudal Hb. In conclusion, catecholaminergic (DA, NA) and ORXergic transmission impacts the clock properties of the Hb clock likely contributing to the circadian regulation of motivated behaviors. Accordingly, pathological conditions that lead in alterations of catecholamine or ORX activity (drug intake, compulsive feeding) might affect the Hb clock and conduct to circadian disturbances.
Collapse
Affiliation(s)
- Nora L Salaberry
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, 8 Allée du Général Rouvillois, Strasbourg, 67000, France
| | - Jorge Mendoza
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, 8 Allée du Général Rouvillois, Strasbourg, 67000, France.
| |
Collapse
|
18
|
Yokoi S, Mitsutake A. Characteristic structural difference between inactive and active states of orexin 2 receptor determined using molecular dynamics simulations. Biophys Rev 2021; 14:221-231. [DOI: 10.1007/s12551-021-00862-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
|
19
|
Goldstein N, Tsuneki H, Bhandarkar N, Aimaretti E, Haim Y, Kon K, Sato K, Wada T, Liberty IF, Kirshtein B, Dukhno O, Maixner N, Gepner Y, Sasaoka T, Rudich A. Human adipose tissue is a putative direct target of daytime orexin with favorable metabolic effects: A cross-sectional study. Obesity (Silver Spring) 2021; 29:1857-1867. [PMID: 34472713 DOI: 10.1002/oby.23262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/03/2021] [Accepted: 07/03/2021] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Orexin/hypocretin (Ox) and its receptors (OxR), a neuroendocrine system centrally regulating sleep/wakefulness, were implicated in the regulation of peripheral metabolism. It was hypothesized that human adipose tissue constitutes a direct target of the OxA/OxR system that associates with distinct metabolic profile(s). METHODS Serum Ox levels and abdominal subcutaneous and visceral adipose tissue expression of Ox/HCRT, OxR1/HCRTR1, and OxR2/HCRTR2 were measured in n = 81 patients. RESULTS Higher morning circulating Ox levels were associated with improved lipid profile and insulin sensitivity, independently of BMI (β = -0.363, p = 0.018 for BMI-adjusted homeostatic model of insulin resistance). Adipose HCRT mRNA was detectable in <20% of patients. Visceral HCRT expressers were mostly (80%) males and, compared with nonexpressers, had lower total and LDL cholesterol. HCRTR1 was readily detectable, and HCRTR2 was undetectable. HCRTR1 mRNA and OxR1 protein expression were higher in subcutaneous than visceral adipose tissue, and among nonobese patients, patients with obesity, and patients with obesity and T2DM were 3.4 (1.0), 0.7 (0.1), 0.6 (0.1) (AU) (p < 0.001) and 1.0 (0.2), 0.5 (0.1), 0.4 (0.1) (AU) (p = NS), respectively. Higher visceral HCRTR1 expression was associated with lower fasting insulin and homeostatic model of insulin resistance, also after adjusting for BMI. In human adipocytes, HCRTR1 expression did not exhibit significant oscillation. CONCLUSIONS Human adipose tissue is a putative direct target of the OxA-OxR1 system, with higher morning input being associated with improved metabolic profile.
Collapse
Affiliation(s)
- Nir Goldstein
- Department of Clinical Biochemistry and Pharmacology and the National Institute of Biotechnology in the Negev, Ben-Gurion University, Beer-Sheva, Israel
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Sylvan Adams Sports Institute, Tel-Aviv University, Tel-Aviv, Israel
| | - Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, Sugitani, Toyama, Japan
| | - Nikhil Bhandarkar
- Department of Clinical Biochemistry and Pharmacology and the National Institute of Biotechnology in the Negev, Ben-Gurion University, Beer-Sheva, Israel
| | - Eleonora Aimaretti
- Department of Clinical Biochemistry and Pharmacology and the National Institute of Biotechnology in the Negev, Ben-Gurion University, Beer-Sheva, Israel
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Yulia Haim
- Department of Clinical Biochemistry and Pharmacology and the National Institute of Biotechnology in the Negev, Ben-Gurion University, Beer-Sheva, Israel
| | - Kanta Kon
- Department of Clinical Pharmacology, University of Toyama, Sugitani, Toyama, Japan
| | - Kiyofumi Sato
- Department of Clinical Pharmacology, University of Toyama, Sugitani, Toyama, Japan
| | - Tsutomu Wada
- Department of Clinical Pharmacology, University of Toyama, Sugitani, Toyama, Japan
| | - Idit F Liberty
- Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Boris Kirshtein
- Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Oleg Dukhno
- Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nitzan Maixner
- Department of Clinical Biochemistry and Pharmacology and the National Institute of Biotechnology in the Negev, Ben-Gurion University, Beer-Sheva, Israel
| | - Yftach Gepner
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Sylvan Adams Sports Institute, Tel-Aviv University, Tel-Aviv, Israel
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University of Toyama, Sugitani, Toyama, Japan
| | - Assaf Rudich
- Department of Clinical Biochemistry and Pharmacology and the National Institute of Biotechnology in the Negev, Ben-Gurion University, Beer-Sheva, Israel
| |
Collapse
|
20
|
Exploring the Role of Orexinergic Neurons in Parkinson's Disease. Neurotox Res 2021; 39:2141-2153. [PMID: 34495449 DOI: 10.1007/s12640-021-00411-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 07/31/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease affecting about 2% of the population. A neuropeptide, orexin, is linked with sleep abnormalities in the parkinsonian patient. This study aimed to review the changes in the orexinergic system in parkinsonian subjects and the effects of orexin. A number of search techniques were used and presumed during the search, including cloud databank searches of PubMed and Medline using title words, keywords, and MeSH terms. PD is characterised by motor dysfunctions (postural instability, rigidity, tremor) and cognitive disorders, sleep-wake abnormalities grouped under non-motor disorders. The Orexinergic system found in the hypothalamus is linked with autonomic function, neuroprotection, learning and memory, and the sleep-wake cycle. Prepro-orexin, a precursor peptide (130 amino acids), gives rise to orexins (Orx-A and Orx-B). Serum orexin level measurement is vital for evaluating several neurological disorders (Alzheimer's disease, Huntington's disease, and PD). Orexinergic neurons are activated by hypoglycemia and ghrelin, while they are restrained by food consumption and leptin. Orexinergic system dysfunctioning was found to be linked with non-motor symptoms (sleep abnormalities) in PD. Orexinergic neuron's behaviour may be either inhibitory or excitatory depending on the environment in which they are present. As well, orexin antagonists are found to improve the abnormal sleep pattern. Since the orexinergic system plays a role in several psychological and neurological disorders, therefore, these disorders can be managed by targeting this system.
Collapse
|
21
|
Subramanian S, Ravichandran M. Orexin receptors: Targets and applications. Fundam Clin Pharmacol 2021; 36:72-80. [PMID: 34464995 DOI: 10.1111/fcp.12723] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/18/2021] [Accepted: 08/26/2021] [Indexed: 01/02/2023]
Abstract
Over the years, elucidating targets from the neural circuits that can be used to treat disorders pertaining to the nervous system and extending their scope to other systems have always proved interesting to researchers. The role of various peptides and neurotransmitters has been elucidated and is being developed as therapeutic targets. Out of these, orexins are neuropeptides produced in the hypothalamus that stimulate a specific type of G-Protein coupled receptors (GPCR) called orexin receptors and bring about various physiological and pathological roles. Orexin receptors are of interest not only because of their wide applications such as insomnia, obesity, and inflammatory disorders but also because of their contribution to promising aspects of drug discovery such as optogenetics and their tremendous growth from the stage of being orphans to orexins. This review will discuss in detail the structure of orexin receptors, their physiological role, and various applications in disease states adding a note on agonists and antagonists and finally summarizing the recent drug approvals in the field.
Collapse
Affiliation(s)
- Subhiksha Subramanian
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Mirunalini Ravichandran
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| |
Collapse
|
22
|
Focus on the Complex Interconnection between Cancer, Narcolepsy and Other Neurodegenerative Diseases: A Possible Case of Orexin-Dependent Inverse Comorbidity. Cancers (Basel) 2021; 13:cancers13112612. [PMID: 34073579 PMCID: PMC8198883 DOI: 10.3390/cancers13112612] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary This narrative review first describes from several points of view the complex interrelationship between cancer and neurodegeneration, with special attention to the mechanisms that might underlie an inverse relationship between them. In particular, the mechanisms that might induce an imbalance between cell apoptotic and proliferative stimuli are discussed. Second, the review summarizes findings on orexins and their involvement in narcolepsy, neurodegenerative diseases, and cancer, starting from epidemiological data then addressing laboratory findings, animal models, and human clinical observational and interventional investigations. Important research efforts are warranted on these topics, as they might lead to novel therapeutic approaches to both neurodegenerative diseases and cancer. Abstract Conditions such as Alzheimer’s (AD) and Parkinson’s diseases (PD) are less prevalent in cancer survivors and, overall, cancer is less prevalent in subjects with these neurodegenerative disorders. This seems to suggest that a propensity towards one type of disease may decrease the risk of the other. In addition to epidemiologic data, there is also evidence of a complex biological interconnection, with genes, proteins, and pathways often showing opposite dysregulation in cancer and neurodegenerative diseases. In this narrative review, we focus on the possible role played by orexin signaling, which is altered in patients with narcolepsy type 1 and in those with AD and PD, and which has been linked to β-amyloid brain levels and inflammation in mouse models and to cancer in cell lines. Taken together, these lines of evidence depict a possible case of inverse comorbidity between cancer and neurodegenerative disorders, with a role played by orexins. These considerations suggest a therapeutic potential of orexin modulation in diverse pathologies such as narcolepsy, neurodegenerative disorders, and cancer.
Collapse
|
23
|
Li TL, Lee YH, Wu FH, Hwang LL. Orexin-A directly depolarizes dorsomedial hypothalamic neurons, including those innervating the rostral ventrolateral medulla. Eur J Pharmacol 2021; 899:174033. [PMID: 33727058 DOI: 10.1016/j.ejphar.2021.174033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 02/10/2021] [Accepted: 03/10/2021] [Indexed: 02/04/2023]
Abstract
The dorsomedial hypothalamus (DMH) receives dense orexinergic innervation. Intra-DMH application of orexins increases arterial pressure and heart rate in rats. We studied the effects of orexin-A on DMH neurons, including those innervating the medullary cardiovascular center, the rostral ventrolateral medulla (RVLM), by using whole-cell recordings in brain slices. In the presence of tetrodotoxin, orexin-A (30-1000 nM) depolarized 56% of DMH neurons (EC50 82.4 ± 4.4 nM). Under voltage-clamp recording, orexin-A (300 nM) induced three types of responses characterized by different current-voltage relationships, namely unchanged, increased, and decreased slope conductance in 68%, 14%, and 18% of orexin-A-responsive neurons, respectively. The reversal potential of the decreased-conductance response was near the equilibrium potential of K+ and became more positive in a high-K+ solution, suggesting that K+ conductance blockade is the underlying mechanism. In a low-Na+ solution, unchanged-, increased-, and decreased-conductance responses were observed in 56%, 11%, and 33% of orexin-A-responsive neurons, respectively, implying that a non-selective cation current (NSCC) underlies orexin-A-induced responses in a small population of DMH neurons. KBR-7943 (70 μM), an inhibitor of Na+-Ca2+ exchanger (NCX), suppressed orexin-A-induced depolarization in 7 of 10 neurons. In the presence of KBR-7943, the majority of orexin-A-responsive neurons exhibited decreased-conductance responses. These findings suggest that NCX activation may underlie orexin-A-induced depolarization in the majority of orexin-responsive DMH neurons. Of 19 RVLM-projecting DMH neurons identified by retrograde labeling, 17 (90%) were orexin-A responsive. In conclusion, orexin-A directly excited over half of DMH neurons, including those innervating the RVLM, through decreasing K+ conductance, activating NCX, and/or increasing NSCC.
Collapse
Affiliation(s)
- Tzu-Ling Li
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Yen-Hsien Lee
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, Taiwan.
| | - Feng-Hsu Wu
- Division of General Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan.
| | - Ling-Ling Hwang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
24
|
Erichsen JM, Calva CB, Reagan LP, Fadel JR. Intranasal insulin and orexins to treat age-related cognitive decline. Physiol Behav 2021; 234:113370. [PMID: 33621561 PMCID: PMC8053680 DOI: 10.1016/j.physbeh.2021.113370] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/19/2021] [Indexed: 02/06/2023]
Abstract
The intranasal (IN) administration of neuropeptides, such as insulin and orexins, has been suggested as a treatment strategy for age-related cognitive decline (ARCD). Because dysfunctional neuropeptide signaling is an observed characteristic of ARCD, it has been suggested that IN delivery of insulin and/or orexins may restore endogenous peptide signaling and thereby preserve cognition. IN administration is particularly alluring as it is a relatively non-invasive method that directly targets peptides to the brain. Several laboratories have examined the behavioral effects of IN insulin in young, aged, and cognitively impaired rodents and humans. These studies demonstrated improved performance on various cognitive tasks following IN insulin administration. Fewer laboratories have assessed the effects of IN orexins; however, this peptide also holds promise as an effective treatment for ARCD through the activation of the cholinergic system and/or the reduction of neuroinflammation. Here, we provide a brief overview of the advantages of IN administration and the delivery pathway, then summarize the current literature on IN insulin and orexins. Additional preclinical studies will be useful to ultimately uncover the mechanisms underlying the pro-cognitive effects of IN insulin and orexins, whereas future clinical studies will aid in the determination of the most efficacious dose and dosing paradigm. Eventually, IN insulin and/or orexin administration may be a widely used treatment strategy in the clinic for ARCD.
Collapse
Affiliation(s)
- Jennifer M Erichsen
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States.
| | - Coleman B Calva
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States
| | - Lawrence P Reagan
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States; Columbia VA Health Care System, Columbia, SC, 29208, United States
| | - Jim R Fadel
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States
| |
Collapse
|
25
|
Yokoi S, Mitsutake A. Molecular Dynamics Simulations for the Determination of the Characteristic Structural Differences between Inactive and Active States of Wild Type and Mutants of the Orexin2 Receptor. J Phys Chem B 2021; 125:4286-4298. [PMID: 33885321 DOI: 10.1021/acs.jpcb.0c10985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The orexin2 receptor (OX2R), which is classified as a class A G protein-coupled receptor (GPCR), is the target of our study. We performed over 20 several-microsecond-scale molecular dynamics simulations of the wild type and mutants of OX2R to extract the characteristics of the structural changes taking place in the active state. We introduced mutations that exhibited the stable inactive state and the constitutively active state in class A GPCRs. In these simulations, significant characteristic structural changes were observed in the V3096.40Y mutant, which corresponded to a constitutively active mutant. These conformational changes include the outward movement of the transmembrane helix 6 (TM6) and the inward movement of TM7, which are common structural changes in the activation of GPCRs. In addition, we extracted a suitable index for the quantitative evaluation of the active and inactive states of GPCRs, namely, the inter-atomic distance of Cα atoms between x(3.46) and Y(7.53). The structures of the inactive and active states solved by X-ray crystallography and cryo-electron microscopy can be classified using the inter-atomic distance. Furthermore, we clarified that the inward movement of TM7 requires the swapping of M3056.36 on TM6 and L3677.56 on TM7. Finally, we discussed the structural advantages of TM7 inward movement for GPCR activation.
Collapse
Affiliation(s)
- Shun Yokoi
- Department of Physics, School of Science and Technology, Meiji University, 1-1-1 Higashi-Mita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Ayori Mitsutake
- Department of Physics, School of Science and Technology, Meiji University, 1-1-1 Higashi-Mita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| |
Collapse
|
26
|
Nakajo H, Chou MY, Kinoshita M, Appelbaum L, Shimazaki H, Tsuboi T, Okamoto H. Hunger Potentiates the Habenular Winner Pathway for Social Conflict by Orexin-Promoted Biased Alternative Splicing of the AMPA Receptor Gene. Cell Rep 2021; 31:107790. [PMID: 32579920 DOI: 10.1016/j.celrep.2020.107790] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/13/2020] [Accepted: 05/29/2020] [Indexed: 12/31/2022] Open
Abstract
Many animals fight for dominance between conspecifics. Because winners could obtain more resources than losers, fighting outcomes are important for the animal's survival, especially in a situation with insufficient resources, such as hunger. However, it remains unclear whether and how hunger affects fighting outcomes. Herein, we investigate the effects of food deprivation on brain activity and fighting behaviors in zebrafish. We report that starvation induces winning in social conflicts. Before the fights, starved fish show potentiation of the lateral subregion of the dorsal habenula (dHbL)-dorsal/intermediate interpeduncular nucleus (d/iIPN) pathway, which is known to be essential for and potentiated after winning fights. Circuit potentiation is mediated by hypothalamic orexin/hypocretin neuropeptides, which prolong AMPA-type glutamate receptor (AMPAR) activity by increasing the expression of a flip type of alternative splicing variant of the AMPAR subunit. This mechanism may underlie how hungry vertebrates win fights and may be commonly shared across animal phylogeny.
Collapse
Affiliation(s)
- Haruna Nakajo
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Center for Brain Science, Saitama 351-0198, Japan; Department of Life Sciences, Graduate School of Arts and Science, The University of Tokyo, Tokyo 153-8902, Japan
| | - Ming-Yi Chou
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Center for Brain Science, Saitama 351-0198, Japan; Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Masae Kinoshita
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Lior Appelbaum
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Hideaki Shimazaki
- Graduate School of Informatics, Kyoto University, Kyoto 606-8501, Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Science, The University of Tokyo, Tokyo 153-8902, Japan
| | - Hitoshi Okamoto
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Center for Brain Science, Saitama 351-0198, Japan; RIKEN CBS-Kao Collaboration Center, Saitama 351-0198, Japan; Department of Life Sciences, Graduate School of Arts and Science, The University of Tokyo, Tokyo 153-8902, Japan.
| |
Collapse
|
27
|
Orexin-A differentially modulates inhibitory and excitatory synaptic transmission in rat inner retina. Neuropharmacology 2021; 187:108492. [PMID: 33582153 DOI: 10.1016/j.neuropharm.2021.108492] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/08/2021] [Accepted: 02/06/2021] [Indexed: 11/21/2022]
Abstract
In this work, modulation by orexin-A of the release of glutamate and GABA from bipolar and amacrine cells respectively was studied by examining the effects of the neuropeptide on miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs) of rat retinal ganglion cells (GCs). Using RNAscope in situ hybridization in combination with immunohistochemistry, we showed positive signals for orexin receptor-1 (OX1R) mRNA in the bipolar cell terminals and those for orexin receptor-2 (OX2R) mRNA in the amacrine cell terminals. With whole-cell patch-clamp recordings in rat retinal slices, we demonstrated that application of orexin-A reduced the interevent interval of mEPSCs of GCs through OX1R. However, it increased the interevent interval of mIPSCs, mediated by GABAA receptors, through OX2R. Furthermore, orexin-A-induced reduction of mEPSC interevent interval was abolished by the application of PI-PLC inhibitors or PKC inhibitors. In contrast, orexin-A-induced increase of GABAergic mIPSC interevent interval was mimicked by 8-Br-cAMP or an adenylyl cyclase activator, but was eliminated by PKA antagonists. Finally, application of nimodipine, an L-type Ca2+ channel blocker, increased both mEPSC and mIPSC interevent interval, and co-application of orexin-A no longer changed the mEPSCs and mIPSCs. We conclude that orexin-A increases presynaptic glutamate release onto GCs by activating L-type Ca2+ channels in bipolar cells, a process that is mediated by an OX1R/PI-PLC/PKC signaling pathway. However, orexin-A decreases presynaptic GABA release onto GCs by inhibiting L-type Ca2+ channels in amacrine cells, a process that is mediated by an OX2R/cAMP-PKA signaling pathway.
Collapse
|
28
|
Alzugaray ME, Gavazzi MV, Ronderos JR. G protein-coupled receptor signal transduction and Ca 2+ signaling pathways of the allatotropin/orexin system in Hydra. Gen Comp Endocrinol 2021; 300:113637. [PMID: 33017583 DOI: 10.1016/j.ygcen.2020.113637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/16/2020] [Accepted: 09/28/2020] [Indexed: 11/24/2022]
Abstract
Allatotropin is a pleiotropic peptide originally characterized in insects. The existence of AT neuropeptide signaling was proposed in other invertebrates. In fact, we previously proposed the presence of an AT-like system regulating feeding behavior in Hydra sp. Even in insects, the information about the AT signaling pathway is incomplete. The aim of this study is to analyze the signaling cascade activated by AT in Hydra plagiodesmica using a pharmacological approach. The results show the involvement of Ca2+ and IP3 signaling in the transduction pathway of the peptide. Furthermore, we confirm the existence of a GPCR system involved in this pathway, that would be coupled to a Gq subfamily of Gα protein, which activates a PLC, inducing an increase in IP3 and cytosolic Ca2+. To the best of our knowledge, this work represents the first in vivo approach to study the overall signaling pathway and intracellular events involved in the myoregulatory effect of AT in Hydra sp.
Collapse
Affiliation(s)
- María Eugenia Alzugaray
- Cátedra de Histología y Embriología Animal. Facultad de Ciencias Naturales y Museo, Universidad Nacional de La Plata (FCNyM-UNLP), Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - María Victoria Gavazzi
- Cátedra de Histología y Embriología Animal. Facultad de Ciencias Naturales y Museo, Universidad Nacional de La Plata (FCNyM-UNLP), Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Jorge Rafael Ronderos
- Cátedra de Histología y Embriología Animal. Facultad de Ciencias Naturales y Museo, Universidad Nacional de La Plata (FCNyM-UNLP), Argentina.
| |
Collapse
|
29
|
Yaeger JD, Krupp KT, Gale JJ, Summers CH. Counterbalanced microcircuits for Orx1 and Orx2 regulation of stress reactivity. MEDICINE IN DRUG DISCOVERY 2020. [DOI: 10.1016/j.medidd.2020.100059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
30
|
Hwang YT, Piguet O, Hodges JR, Grunstein R, Burrell JR. Sleep and orexin: A new paradigm for understanding behavioural-variant frontotemporal dementia? Sleep Med Rev 2020; 54:101361. [DOI: 10.1016/j.smrv.2020.101361] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022]
|
31
|
Khairuddin S, Aquili L, Heng BC, Hoo TLC, Wong KH, Lim LW. Dysregulation of the orexinergic system: A potential neuropeptide target in depression. Neurosci Biobehav Rev 2020; 118:384-396. [DOI: 10.1016/j.neubiorev.2020.07.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 07/19/2020] [Accepted: 07/31/2020] [Indexed: 12/20/2022]
|
32
|
Mediavilla C. Bidirectional gut-brain communication: A role for orexin-A. Neurochem Int 2020; 141:104882. [PMID: 33068686 DOI: 10.1016/j.neuint.2020.104882] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 10/02/2020] [Accepted: 10/11/2020] [Indexed: 02/06/2023]
Abstract
It is increasingly evident that bidirectional gut-brain signaling provides a communication pathway that uses neural, hormonal, and immunological routes to regulate homeostatic mechanisms such as hunger/satiety as well as emotions and inflammation. Hence, disruption of the gut-brain axis can cause numerous pathophysiologies, including obesity and intestinal inflammatory diseases. One chemical mediator in the gut-brain axis is orexin-A, given that hypothalamic orexin-A affects gastrointestinal motility and secretion, and peripheral orexin in the intestinal mucosa can modulate brain functions, making possible an orexinergic gut-brain network. It has been proposed that orexin-A acts on this axis to regulate nutritional processes, such as short-term intake, gastric acid secretion, and motor activity associated with the cephalic phase of feeding. Orexin-A has also been related to stress systems and stress responses via the hypothalamic-pituitary-adrenal axis. Recent studies on the relationship of orexin with immune system-brain communications in an animal model of colitis suggested an immunomodulatory role for orexin-A in signaling and responding to infection by reducing the production of pro-inflammatory cytokines (e.g., tumor necrosis factor α, interleukin-6, and monocyte chemoattractant protein-1). These studies suggested that orexin administration might be of potential therapeutic value in irritable bowel syndrome or chronic intestinal inflammatory diseases, in which gastrointestinal symptoms frequently coexist with behavioral disorders, including loss of appetite, anxiety, depression, and sleeping disorders. Interventions in the orexinergic system have been proposed as a therapeutic approach to these diseases and for the treatment of chemotherapeutic drug-related hyperalgesia and fatigue in cancer patients.
Collapse
Affiliation(s)
- Cristina Mediavilla
- Department of Psychobiology, and Mind, Brain, and Behavior Research Center (CIMCYC), University of Granada, Spain.
| |
Collapse
|
33
|
Inhibition of orexin receptor 1 contributes to the development of morphine dependence via attenuation of cAMP response element-binding protein and phospholipase Cβ3. J Chem Neuroanat 2020; 108:101801. [DOI: 10.1016/j.jchemneu.2020.101801] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/05/2020] [Accepted: 05/05/2020] [Indexed: 11/21/2022]
|
34
|
Sadat-Shirazi MS, Soltani H, Nikpour N, Haghshenas M, Khalifeh S, Mokri A, Zarrindast MR. Alteration of orexin-A and PKCα in the postmortem brain of pure-opioid and multi-drug abusers. Neuropeptides 2020; 83:102074. [PMID: 32741526 DOI: 10.1016/j.npep.2020.102074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/18/2020] [Accepted: 07/19/2020] [Indexed: 02/09/2023]
Abstract
Finding changes induced by the drug of abuse is one of the most important approaches to design new drugs for the treatment of substance use disorders (SUD). Postmortem study is the most reliable method for detecting alteration in the brain of SUD patients. Recently, the role of orexinergic system in SUD is in consideration. In the current study, we evaluated the level of orexin-A in the CSF and protein kinase Cα (PKCα) in the brain of pure-opioid (POA) and multi-drug abusers (MDA). A total of 56 POA, 45 MDA, and 13 matched control brains were collected from the legal medicine center, Tehran, Iran. The CSF was gathered from the third ventricle immediately after opening the skull and kept at -80 °C. The medial prefrontal cortex (mPFC), lateral prefrontal cortex (lPFC), orbitofrontal cortex (OFC), nucleus accumbens (NAc), and amygdala were dissected from fresh brain, frozen with liquid nitrogen and kept at -80 °C. The level of orexin-A evaluated in the CSF. Using western blotting, the level of PKCα assessed in the brain. Obtained data revealed that the level of orexin-A increased in POA and MDA compared with the control group (p < 0.05). In addition, the level of PKCα increased in the prefrontal cortex and amygdala of the abusers compared with the control group, although we did not detect changes in the level of PKCα in the NAc. Along with animal studies, the current results showed that the level of orexin increased in the CSF of drug abusers, which might be related to increases in the activation of lateral hypothalamic orexinergic neurons faced with the drug of abuse. Enhancement in the level of PKCα in the drug reward circuits might be adaptational changes induced by orexin and drugs of abuse.
Collapse
Affiliation(s)
| | - Haniyeh Soltani
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Nikpour
- Department of Microbiology, Pasteur Institute of Iran, Tehran, Iran
| | - Masoud Haghshenas
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Khalifeh
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Amir-Almomenin Hospital, Islamic Azad University, Tehran, Iran
| | - Azarakhsh Mokri
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran; Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran; Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Amir-Almomenin Hospital, Islamic Azad University, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Endocrinology and Metabolism Research Institute, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
35
|
Phospholipase Cβ3 in the hippocampus may mediate impairment of memory by long-term blockade of orexin 1 receptors assessed by the Morris water maze. Life Sci 2020; 257:118046. [PMID: 32622948 DOI: 10.1016/j.lfs.2020.118046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/19/2020] [Accepted: 06/30/2020] [Indexed: 11/23/2022]
Abstract
Orexin-A is an endogenous peptide with receptors throughout the brain. According to some recent research, learning and memory are affected by the central administration of orexin; however, no study so far has investigated the long-term inhibition of the orexinergic system. The present study has evaluated the effect of pretraining administration of orexin 1 receptor (OXR1) antagonist, SB-334867, on the acquisition of memory. The Morris water maze (MWM) task was used for training and trial purposes in all groups. Memory performance was analyzed by measuring escape latency, traveled distance, and time spent in the target quadrant. Moreover, the effect of SB-334867 on phospholipase Cβ3 (PLCβ3) levels in the CA1 region of hippocampus slices was examined. Hippocampus slices were prepared using an immunohistochemistry (IHC) approach. SB-334867 (20 mg/kg) increased escape latency in SB-treated rats compared to SB-vehicle group (P < 0.01). SB-treated rats spent less time in the target quadrant compared to the SB-vehicle group (P < 0.001). Distance traveled in the target quadrant was significantly more in SB-treated rats compared to the SB-vehicle group (P < 0.001). Furthermore, SB-334867 decreased PLCβ3 levels in the CA1 of the hippocampus (P < 0.01 and P < 0.05, respectively). Put together, our results suggest that the long-term inhibition of OXR1 plays a prominent role in spatial learning and memory, probably by attenuating PLCβ3 in CA1 neurons.
Collapse
|
36
|
Cai X, Wang H, Wang M, Wang D, Zhang Z, Wei R, Gao X, Zhang R, Wang C, Chen J. A novel phosphorylation site on orexin receptor 1 regulating orexinA-induced GRK2-biased signaling. Cell Signal 2020; 75:109743. [PMID: 32827691 DOI: 10.1016/j.cellsig.2020.109743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 11/24/2022]
Abstract
Drug discovery efforts targeting G protein-coupled receptors (GPCRs) have succeeded in developing multiple medications for treating various human diseases including cancer, metabolic disorders, and inflammatory disorders. These medications are broadly classified as either agonists or antagonists that respectively promote or inhibit receptor activation by endogenous stimuli. However, there has been a growing appreciation that GPCR biased signaling between G protein- and β-arrestin-dependent signaling in particular is a promising method for improving drug efficacy and therapy. Orexin receptor 1 (OX1R), a member of the GPCRs, is an important drug target in the central nervous system. In this study, we identified a novel regulatory phosphorylation site (Ser-262) on OX1R that abolished its capability to interact with GRK2, but did not affect its interaction with G proteins, GRK5, or β-arrestin1/2 activation, indicating that Ser-262 is a key amino acid for OX1R internalization that contributes to induction of GRK2-dependent biased signaling via orexin A. Our findings could potentially lead to the development of new drug targets for the prevention and treatment of insomnia, narcolepsy, and substance abuse, with fewer side effects than existing therapies.
Collapse
Affiliation(s)
- Xin Cai
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, 261042, PR China
| | - Huannan Wang
- Neurobiology Institute, Jining Medical University, Jining, Shandong, 272067, PR China; School of Pharmacy, Jining Medical University, Rizhao, Shandong, 276800, PR China
| | - Maochang Wang
- Shouguang Agricultural Development Group Co., Ltd,Shouguang, Shandong, 262700, PR China
| | - Dexiu Wang
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, 261042, PR China
| | - Zhen Zhang
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, 276800, PR China
| | - Ruotong Wei
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, 261042, PR China
| | - Xiang Gao
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, 261042, PR China
| | - Rumin Zhang
- Neurobiology Institute, Jining Medical University, Jining, Shandong, 272067, PR China
| | - Chunmei Wang
- Neurobiology Institute, Jining Medical University, Jining, Shandong, 272067, PR China
| | - Jing Chen
- Neurobiology Institute, Jining Medical University, Jining, Shandong, 272067, PR China; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
37
|
Pang YJ, Feng H, Wen SY, Qiao QC, Zhang J, Yang N. Orexin enhances firing activities in the gigantocellular reticular nucleus through the activation of non-selective cationic conductance. Neurosci Lett 2020; 733:135088. [PMID: 32464262 DOI: 10.1016/j.neulet.2020.135088] [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: 02/22/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 10/24/2022]
Abstract
Orexin/hypocretin has been implicated in central motor control. The gigantocellular reticular nucleus (Gi), a key element of the brainstem motor inhibitory system, also receives orexinergic innervations. However, the modulations of orexin on the neuronal activities and the underlying cellular mechanisms in Gi neurons remain unknown. Here, through whole-cell patch-clamp recordings, we first observed that orexin increased the firing frequency in Gi neurons. Interestingly, a postsynaptic depolarization elicited by orexin was observed in the presence of tetrodotoxin, without altering the input resistance of Gi neurons at around -60 mV. Moreover, through comparing the current-frequency curves constructed by identical current injections from equal membrane potentials, we found that orexin also increased the repetitive firing ability of Gi neurons. This action appeared to be caused by the shortening of inter-spike intervals, without altering the waveform of individual action potentials. We finally revealed that activation of the non-selective cationic conductance contributed to the orexin-elicited excitation in Gi neurons. Together, these results suggest that orexin may facilitate Gi-mediated motor functions through enhancing the neuronal activities of Gi neurons.
Collapse
Affiliation(s)
- Yu-Jie Pang
- Department of Physiology, Army Medical University, Chongqing 400038, PR China
| | - Hui Feng
- Department of Physiology, Army Medical University, Chongqing 400038, PR China
| | - Si-Yi Wen
- Department of Physiology, Army Medical University, Chongqing 400038, PR China
| | - Qi-Cheng Qiao
- Department of Physiology, Army Medical University, Chongqing 400038, PR China
| | - Jun Zhang
- Department of Physiology, Army Medical University, Chongqing 400038, PR China.
| | - Nian Yang
- Department of Physiology, Army Medical University, Chongqing 400038, PR China.
| |
Collapse
|
38
|
Ahmadi-Soleimani SM, Mianbandi V, Azizi H, Azhdari-Zarmehri H, Ghaemi-Jandabi M, Abbasi-Mazar A, Mohajer Y, Darana SP. Coregulation of sleep-pain physiological interplay by orexin system: An unprecedented review. Behav Brain Res 2020; 391:112650. [DOI: 10.1016/j.bbr.2020.112650] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/28/2020] [Accepted: 04/08/2020] [Indexed: 12/14/2022]
|
39
|
Calva CB, Fadel JR. Intranasal administration of orexin peptides: Mechanisms and therapeutic potential for age-related cognitive dysfunction. Brain Res 2020; 1731:145921. [PMID: 30148983 PMCID: PMC6387866 DOI: 10.1016/j.brainres.2018.08.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/03/2018] [Accepted: 08/23/2018] [Indexed: 12/11/2022]
Abstract
Cognitive impairment is a core feature of several neuropsychiatric and neurological disorders, including narcolepsy and age-related dementias. Current pharmacotherapeutic approaches to cognitive enhancement are few in number and limited in efficacy. Thus, novel treatment strategies are needed. The hypothalamic orexin (hypocretin) system, a central integrator of physiological function, plays an important role in modulating cognition. Several single- and dual-orexin receptor antagonists are available for various clinical and preclinical applications, but the paucity of orexin agonists has limited the ability to research their therapeutic potential. To circumvent this hurdle, direct intranasal administration of orexin peptides is being investigated as a prospective treatment for cognitive dysfunction, narcolepsy or other disorders in which deficient orexin signaling has been implicated. Here, we describe the possible mechanisms and therapeutic potential of intranasal orexin delivery. Combined with the behavioral evidence that intranasal orexin-A administration improves cognitive function in narcoleptic and sleep-deprived subjects, our neurochemical studies in young and aged animals highlights the capacity for intranasal orexin administration to improve age-related deficits in neurotransmission. In summary, we highlight prior and original work from our lab and from others that provides a framework for the use of intranasal orexin peptides in treating cognitive dysfunction, especially as it relates to age-related cognitive disorders.
Collapse
Affiliation(s)
- Coleman B Calva
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, 6311 Garners Ferry Road, Columbia, SC 29209, USA
| | - Jim R Fadel
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, 6311 Garners Ferry Road, Columbia, SC 29209, USA.
| |
Collapse
|
40
|
Lind A, Eriksson D, Akel O, Ramelius A, Palm L, Lernmark Å, Kämpe O, Elding Larsson H, Landegren N. Screening for autoantibody targets in post-vaccination narcolepsy using proteome arrays. Scand J Immunol 2020; 91:e12864. [PMID: 32056243 DOI: 10.1111/sji.12864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 01/01/2023]
Abstract
Narcolepsy type 1 (NT1) is a chronic sleep disorder caused by a specific loss of hypocretin-producing neurons. The incidence of NT1 increased in Sweden, Finland and Norway following Pandemrix®-vaccination, initiated to prevent the 2009 influenza pandemic. The pathogenesis of NT1 is poorly understood, and causal links to vaccination are yet to be clarified. The strong association with Human leukocyte antigen (HLA) DQB1*06:02 suggests an autoimmune pathogenesis, but proposed autoantigens remain controversial. We used a two-step approach to identify autoantigens in patients that acquired NT1 after Pandemrix®-vaccination. Using arrays of more than 9000 full-length human proteins, we screened the sera of 10 patients and 24 healthy subjects for autoantibodies. Identified candidate antigens were expressed in vitro to enable validation studies with radiobinding assays (RBA). The validation cohort included NT1 patients (n = 39), their first-degree relatives (FDR) (n = 66), population controls (n = 188), and disease controls representing multiple sclerosis (n = 100) and FDR to type 1 diabetes patients (n = 41). Reactivity towards previously suggested NT1 autoantigen candidates including Tribbles homolog 2, Prostaglandin D2 receptor, Hypocretin receptor 2 and α-MSH/proopiomelanocortin was not replicated in the protein array screen. By comparing case to control signals, three novel candidate autoantigens were identified in the protein array screen; LOC401464, PARP3 and FAM63B. However, the RBA did not confirm elevated reactivity towards either of these proteins. In summary, three putative autoantigens in NT1 were identified by protein array screening. Autoantibodies against these candidates could not be verified with independent methods. Further studies are warranted to identify hypothetical autoantigens related to the pathogenesis of Pandemrix®-induced NT1.
Collapse
Affiliation(s)
- Alexander Lind
- Department of Clinical Sciences Malmö, Lund University/CRC, Skåne University Hospital SUS, Malmö, Sweden
| | - Daniel Eriksson
- Department of Medicine (Solna), Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden
| | - Omar Akel
- Department of Clinical Sciences Malmö, Lund University/CRC, Skåne University Hospital SUS, Malmö, Sweden
| | - Anita Ramelius
- Department of Clinical Sciences Malmö, Lund University/CRC, Skåne University Hospital SUS, Malmö, Sweden
| | - Lars Palm
- Section for Paediatric Neurology, Department of Paediatrics, Skåne University Hospital SUS, Malmö, Sweden
| | - Åke Lernmark
- Department of Clinical Sciences Malmö, Lund University/CRC, Skåne University Hospital SUS, Malmö, Sweden
| | - Olle Kämpe
- Department of Medicine (Solna), Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden.,Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Helena Elding Larsson
- Department of Clinical Sciences Malmö, Lund University/CRC, Skåne University Hospital SUS, Malmö, Sweden
| | - Nils Landegren
- Department of Medicine (Solna), Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,K.G. Jebsen Center for Autoimmune Disorders, Bergen, Norway
| |
Collapse
|
41
|
Dai YWE, Lee YH, Li TL, Hwang LL. Mechanisms of orexin 2 receptor-mediated depolarization in the rat paraventricular nucleus of the hypothalamus. Eur J Pharmacol 2020; 869:172802. [DOI: 10.1016/j.ejphar.2019.172802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 12/18/2022]
|
42
|
Alteration of Phospholipase C Expression in Rat Visual Cortical Neurons by Chronic Blockade of Orexin Receptor 1. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09943-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
43
|
Kong T, Qiu K, Liu M, Cheng B, Pan Y, Yang C, Chen J, Wang C. Orexin-A protects against oxygen-glucose deprivation/reoxygenation-induced cell damage by inhibiting endoplasmic reticulum stress-mediated apoptosis via the Gi and PI3K signaling pathways. Cell Signal 2019; 62:109348. [DOI: 10.1016/j.cellsig.2019.109348] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 10/26/2022]
|
44
|
Cellular Mechanisms for Antinociception Produced by Oxytocin and Orexins in the Rat Spinal Lamina II-Comparison with Those of Other Endogenous Pain Modulators. Pharmaceuticals (Basel) 2019; 12:ph12030136. [PMID: 31527474 PMCID: PMC6789548 DOI: 10.3390/ph12030136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/31/2019] [Accepted: 09/12/2019] [Indexed: 01/23/2023] Open
Abstract
Much evidence indicates that hypothalamus-derived neuropeptides, oxytocin, orexins A and B, inhibit nociceptive transmission in the rat spinal dorsal horn. In order to unveil cellular mechanisms for this antinociception, the effects of the neuropeptides on synaptic transmission were examined in spinal lamina II neurons that play a crucial role in antinociception produced by various analgesics by using the whole-cell patch-clamp technique and adult rat spinal cord slices. Oxytocin had no effect on glutamatergic excitatory transmission while producing a membrane depolarization, γ-aminobutyric acid (GABA)-ergic and glycinergic spontaneous inhibitory transmission enhancement. On the other hand, orexins A and B produced a membrane depolarization and/or a presynaptic spontaneous excitatory transmission enhancement. Like oxytocin, orexin A enhanced both GABAergic and glycinergic transmission, whereas orexin B facilitated glycinergic but not GABAergic transmission. These inhibitory transmission enhancements were due to action potential production. Oxytocin, orexins A and B activities were mediated by oxytocin, orexin-1 and orexin-2 receptors, respectively. This review article will mention cellular mechanisms for antinociception produced by oxytocin, orexins A and B, and discuss similarity and difference in antinociceptive mechanisms among the hypothalamic neuropeptides and other endogenous pain modulators (opioids, nociceptin, adenosine, adenosine 5’-triphosphate (ATP), noradrenaline, serotonin, dopamine, somatostatin, cannabinoids, galanin, substance P, bradykinin, neuropeptide Y and acetylcholine) exhibiting a change in membrane potential, excitatory or inhibitory transmission in the spinal lamina II neurons.
Collapse
|
45
|
Li GY, Zhuang QX, Zhang XY, Wang JJ, Zhu JN. Ionic Mechanisms Underlying the Excitatory Effect of Orexin on Rat Subthalamic Nucleus Neurons. Front Cell Neurosci 2019; 13:153. [PMID: 31105528 PMCID: PMC6499184 DOI: 10.3389/fncel.2019.00153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/08/2019] [Indexed: 11/24/2022] Open
Abstract
Central orexinergic system deficiency results in cataplexy, a motor deficit characterized with a sudden loss of muscle tone, highlighting a direct modulatory role of orexin in motor control. However, the neural mechanisms underlying the regulation of orexin on motor function are still largely unknown. The subthalamic nucleus (STN), the only excitatory structure of the basal ganglia, holds a key position in the basal ganglia circuitry and motor control. Previous study has revealed a wide distribution of orexinergic fibers as well as orexin receptors in the basal ganglia including the STN. Therefore, in the present study, by using whole-cell patch clamp recording and immunostaining techniques, the direct effect of orexin on the STN neurons in brain slices, especially the underlying receptor and ionic mechanisms, were investigated. Our results show that orexin-A elicits an excitatory effect on STN neurons in rats. Tetrodotoxin (TTX) does not block the orexin-induced excitation on STN neurons, suggesting a direct postsynaptic action of the neuropeptide. The orexin-A-induced inward current on STN neurons is mediated by the activation of both OX1 and OX2 receptors. Immunofluorescence result shows that OX1 and OX2 receptors are co-expressed and co-localized in STN neurons. Furthermore, Na+-Ca2+ exchangers (NCXs) and inward rectifier K+ channels co-mediate the excitatory effect of orexin-A on STN neurons. These results demonstrate a dual receptor in conjunction with the downstream ionic mechanisms underlying the excitatory action of orexin on STN neurons, suggesting a potential modulation of the central orexinergic system on basal ganglia circuitry as well as its related motor control and motor diseases.
Collapse
Affiliation(s)
- Guang-Ying Li
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qian-Xing Zhuang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xiao-Yang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Jian-Jun Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Jing-Ning Zhu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
46
|
Wang L, He T, Wan B, Wang X, Zhang L. Orexin A ameliorates HBV X protein-induced cytotoxicity and inflammatory response in human hepatocytes. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2003-2009. [PMID: 31106596 DOI: 10.1080/21691401.2019.1614014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Li Wang
- Department of Hepatopancreatobiliary Surgery, He’nan Tumor Hospital, Affiliated to Zhengzhou University, Zhengzhou, China
| | - Tao He
- Department of Hepatopancreatobiliary Surgery, He’nan Tumor Hospital, Affiliated to Zhengzhou University, Zhengzhou, China
| | - Baishun Wan
- Department of Hepatopancreatobiliary Surgery, He’nan Tumor Hospital, Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xiaoqian Wang
- Department of Hepatopancreatobiliary Surgery, He’nan Tumor Hospital, Affiliated to Zhengzhou University, Zhengzhou, China
| | - Ling Zhang
- Department of Hepatopancreatobiliary Surgery, He’nan Tumor Hospital, Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
47
|
Palus-Chramiec K, Chrobok L, Kepczynski M, Lewandowski MH. Orexin A depolarises rat intergeniculate leaflet neurons through non-selective cation channels. Eur J Neurosci 2019; 50:2683-2693. [PMID: 30803080 DOI: 10.1111/ejn.14394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 02/12/2019] [Accepted: 02/15/2019] [Indexed: 12/22/2022]
Abstract
Orexins/hypocretins are hypothalamic neuropeptides that have a variety of functions, including maintenance of arousal, control over the sleep/wake cycle, reward and feeding. Accumulating evidence links orexins to the time-keeping system with a documented action in the master clock-the suprachiasmatic nucleus. The intergeniculate leaflet (IGL) is a thalamic structure with the well-known function of collecting photic and non-photic cues to adjust the rhythm of the suprachiasmatic nucleus to changing environmental conditions. The IGL consists of GABAergic neurons that are intrinsically active, even in slice preparations. Our previous studies revealed the excitatory postsynaptic effects of orexins on single IGL neurons, even though the ionic mechanism underlying this effect remained elusive. Therefore, in this study, we used patch clamp electrophysiology to identify the ions and distinct ion channels responsible for the observed depolarisations. The major finding of this article is that the orexin A-evoked depolarisation of IGL neurons depends on non-selective cation channels, implicating the orexinergic tone in establishing the basal firing rate in these cells. The data presented here strengthen the mutual connections between the time-keeping and orexinergic systems.
Collapse
Affiliation(s)
- Katarzyna Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Krakow, Poland
| | - Lukasz Chrobok
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Krakow, Poland
| | - Mariusz Kepczynski
- Department of Physical Chemistry and Electrochemistry, Faculty of Chemistry, Jagiellonian University in Krakow, Krakow, Poland
| | - Marian Henryk Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
48
|
Gottschalk MG, Richter J, Ziegler C, Schiele MA, Mann J, Geiger MJ, Schartner C, Homola GA, Alpers GW, Büchel C, Fehm L, Fydrich T, Gerlach AL, Gloster AT, Helbig-Lang S, Kalisch R, Kircher T, Lang T, Lonsdorf TB, Pané-Farré CA, Ströhle A, Weber H, Zwanzger P, Arolt V, Romanos M, Wittchen HU, Hamm A, Pauli P, Reif A, Deckert J, Neufang S, Höfler M, Domschke K. Orexin in the anxiety spectrum: association of a HCRTR1 polymorphism with panic disorder/agoraphobia, CBT treatment response and fear-related intermediate phenotypes. Transl Psychiatry 2019; 9:75. [PMID: 30718541 PMCID: PMC6361931 DOI: 10.1038/s41398-019-0415-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 01/06/2019] [Accepted: 01/17/2019] [Indexed: 12/23/2022] Open
Abstract
Preclinical studies point to a pivotal role of the orexin 1 (OX1) receptor in arousal and fear learning and therefore suggest the HCRTR1 gene as a prime candidate in panic disorder (PD) with/without agoraphobia (AG), PD/AG treatment response, and PD/AG-related intermediate phenotypes. Here, a multilevel approach was applied to test the non-synonymous HCRTR1 C/T Ile408Val gene variant (rs2271933) for association with PD/AG in two independent case-control samples (total n = 613 cases, 1839 healthy subjects), as an outcome predictor of a six-weeks exposure-based cognitive behavioral therapy (CBT) in PD/AG patients (n = 189), as well as with respect to agoraphobic cognitions (ACQ) (n = 483 patients, n = 2382 healthy subjects), fMRI alerting network activation in healthy subjects (n = 94), and a behavioral avoidance task in PD/AG pre- and post-CBT (n = 271). The HCRTR1 rs2271933 T allele was associated with PD/AG in both samples independently, and in their meta-analysis (p = 4.2 × 10-7), particularly in the female subsample (p = 9.8 × 10-9). T allele carriers displayed a significantly poorer CBT outcome (e.g., Hamilton anxiety rating scale: p = 7.5 × 10-4). The T allele count was linked to higher ACQ sores in PD/AG and healthy subjects, decreased inferior frontal gyrus and increased locus coeruleus activation in the alerting network. Finally, the T allele count was associated with increased pre-CBT exposure avoidance and autonomic arousal as well as decreased post-CBT improvement. In sum, the present results provide converging evidence for an involvement of HCRTR1 gene variation in the etiology of PD/AG and PD/AG-related traits as well as treatment response to CBT, supporting future therapeutic approaches targeting the orexin-related arousal system.
Collapse
Affiliation(s)
- Michael G. Gottschalk
- Department of Psychiatry and Psychotherapy, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany ,0000 0001 1378 7891grid.411760.5Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Jan Richter
- grid.5603.0Department of Biological and Clinical Psychology/Psychotherapy, University of Greifswald, Greifswald, Germany
| | - Christiane Ziegler
- Department of Psychiatry and Psychotherapy, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Miriam A. Schiele
- Department of Psychiatry and Psychotherapy, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Mann
- 0000 0001 1378 7891grid.411760.5Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Maximilian J. Geiger
- 0000 0001 1378 7891grid.411760.5Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany ,grid.5963.9Epilepsy Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Schartner
- 0000 0001 1378 7891grid.411760.5Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany ,0000 0001 2297 6811grid.266102.1Department of Physiology, University of California San Francisco, San Francisco, CA USA
| | - György A. Homola
- 0000 0001 1958 8658grid.8379.5Department of Neuroradiology, University of Würzburg, Würzburg, Germany
| | - Georg W. Alpers
- 0000 0001 0943 599Xgrid.5601.2Department of Psychology, School of Social Sciences, University of Mannheim, Mannheim, Germany
| | - Christian Büchel
- 0000 0001 2180 3484grid.13648.38Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lydia Fehm
- 0000 0001 2248 7639grid.7468.dDepartment of Psychology, Humboldt University, Berlin, Germany
| | - Thomas Fydrich
- 0000 0001 2248 7639grid.7468.dDepartment of Psychology, Humboldt University, Berlin, Germany
| | - Alexander L. Gerlach
- 0000 0000 8580 3777grid.6190.eDepartment of Clinical Psychology and Psychotherapy, University of Cologne, Cologne, Germany
| | - Andrew T. Gloster
- 0000 0001 2111 7257grid.4488.0Department of Psychology, Institute of Clinical Psychology and Psychotherapy, Technische Universität Dresden, Dresden, Germany ,0000 0004 1937 0642grid.6612.3Division of Clinical Psychology and Intervention Science, University of Basel, Basel, Switzerland
| | - Sylvia Helbig-Lang
- 0000 0001 2111 7257grid.4488.0Department of Psychology, Institute of Clinical Psychology and Psychotherapy, Technische Universität Dresden, Dresden, Germany ,0000 0001 2287 2617grid.9026.dDepartment of Psychology and Psychotherapy, University of Hamburg, Hamburg, Germany
| | - Raffael Kalisch
- grid.410607.4Neuroimaging Center (NIC) und Deutsches Resilienz-Zentrum (DRZ), Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Tilo Kircher
- 0000 0004 1936 9756grid.10253.35Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - Thomas Lang
- 0000 0001 2111 7257grid.4488.0Department of Psychology, Institute of Clinical Psychology and Psychotherapy, Technische Universität Dresden, Dresden, Germany ,0000 0001 2287 2617grid.9026.dDepartment of Psychology and Psychotherapy, University of Hamburg, Hamburg, Germany ,Christoph-Dornier-Foundation for Clinical Psychology, Bremen, Germany
| | - Tina B. Lonsdorf
- 0000 0001 2180 3484grid.13648.38Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christiane A. Pané-Farré
- grid.5603.0Department of Biological and Clinical Psychology/Psychotherapy, University of Greifswald, Greifswald, Germany
| | - Andreas Ströhle
- Department of Psychiatry and Psychotherapy, Campus Charité Mitte, Charité – Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Heike Weber
- 0000 0001 1378 7891grid.411760.5Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany ,0000 0004 0578 8220grid.411088.4Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt, Germany
| | - Peter Zwanzger
- 0000 0004 0551 4246grid.16149.3bDepartment of Psychiatry and Psychotherapy, University Hospital of Münster, Münster, Germany ,kbo-Inn-Salzach-Hospital, Wasserburg, Germany ,0000 0004 1936 973Xgrid.5252.0Department of Psychiatry und Psychotherapy, Ludwig Maximilians University, Munich, Germany
| | - Volker Arolt
- 0000 0004 0551 4246grid.16149.3bDepartment of Psychiatry and Psychotherapy, University Hospital of Münster, Münster, Germany
| | - Marcel Romanos
- 0000 0001 1378 7891grid.411760.5Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Hans-Ulrich Wittchen
- 0000 0001 2111 7257grid.4488.0Department of Psychology, Institute of Clinical Psychology and Psychotherapy, Technische Universität Dresden, Dresden, Germany ,0000 0004 1936 973Xgrid.5252.0Department of Psychiatry und Psychotherapy, Ludwig Maximilians University, Munich, Germany
| | - Alfons Hamm
- grid.5603.0Department of Biological and Clinical Psychology/Psychotherapy, University of Greifswald, Greifswald, Germany
| | - Paul Pauli
- 0000 0001 1958 8658grid.8379.5Department of Psychology, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Andreas Reif
- 0000 0004 0578 8220grid.411088.4Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt, Germany
| | - Jürgen Deckert
- 0000 0001 1378 7891grid.411760.5Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Susanne Neufang
- 0000 0001 1378 7891grid.411760.5Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany ,0000 0001 2176 9917grid.411327.2Department of Psychiatry and Psychotherapy, Medical Faculty Heinrich-Heine University, Duesseldorf, Germany
| | - Michael Höfler
- 0000 0001 2111 7257grid.4488.0Department of Psychology, Institute of Clinical Psychology and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Center for NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
49
|
Couvineau A, Voisin T, Nicole P, Gratio V, Abad C, Tan YV. Orexins as Novel Therapeutic Targets in Inflammatory and Neurodegenerative Diseases. Front Endocrinol (Lausanne) 2019; 10:709. [PMID: 31695678 PMCID: PMC6817618 DOI: 10.3389/fendo.2019.00709] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/02/2019] [Indexed: 02/05/2023] Open
Abstract
Orexins [orexin-A (OXA) and orexin-B (OXB)] are two isoforms of neuropeptides produced by the hypothalamus. The main biological actions of orexins, focused on the central nervous system, are to control the sleep/wake process, appetite and feeding, energy homeostasis, drug addiction, and cognitive processes. These effects are mediated by two G protein-coupled receptor (GPCR) subtypes named OX1R and OX2R. In accordance with the synergic and dynamic relationship between the nervous and immune systems, orexins also have neuroprotective and immuno-regulatory (i.e., anti-inflammatory) properties. The present review gathers recent data demonstrating that orexins may have a therapeutic potential in several pathologies with an immune component including multiple sclerosis, Alzheimer's disease, narcolepsy, obesity, intestinal bowel diseases, septic shock, and cancers.
Collapse
Affiliation(s)
- Alain Couvineau
- INSERM UMR1149/Inflammation Research Center (CRI), Team “From Inflammation to Cancer in Digestive Diseases” Labeled by “la Ligue Nationale Contre le Cancer”, University of Paris, Paris, France
- *Correspondence: Alain Couvineau
| | - Thierry Voisin
- INSERM UMR1149/Inflammation Research Center (CRI), Team “From Inflammation to Cancer in Digestive Diseases” Labeled by “la Ligue Nationale Contre le Cancer”, University of Paris, Paris, France
| | - Pascal Nicole
- INSERM UMR1149/Inflammation Research Center (CRI), Team “From Inflammation to Cancer in Digestive Diseases” Labeled by “la Ligue Nationale Contre le Cancer”, University of Paris, Paris, France
| | - Valérie Gratio
- INSERM UMR1149/Inflammation Research Center (CRI), Team “From Inflammation to Cancer in Digestive Diseases” Labeled by “la Ligue Nationale Contre le Cancer”, University of Paris, Paris, France
| | - Catalina Abad
- University of Rouen Normandy, INSERM U1234 PANTHER, IRIB, Rouen, France
| | - Yossan-Var Tan
- University of Rouen Normandy, INSERM U1234 PANTHER, IRIB, Rouen, France
- Yossan-Var Tan
| |
Collapse
|
50
|
Messal N, Fernandez N, Dayot S, Gratio V, Nicole P, Prochasson C, Chantret I, LeGuilloux G, Jarry A, Couvelard A, Tréton X, Voisin T, Ogier-Denis E, Couvineau A. Ectopic expression of OX1R in ulcerative colitis mediates anti-inflammatory effect of orexin-A. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3618-3628. [PMID: 30251681 DOI: 10.1016/j.bbadis.2018.08.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/07/2018] [Accepted: 08/17/2018] [Indexed: 02/08/2023]
Abstract
Orexins (orexin-A and orexin-B) are hypothalamic peptides that are produced by the same precursor and are involved in sleep/wake control, which is mediated by two G protein-coupled receptor subtypes, OX1R and OX2R. Ulcerative colitis (UC) is an inflammatory bowel disease, (IBD) which is characterized by long-lasting inflammation and ulcers that affect the colon and rectum mucosa and is known to be a significant risk factor for colon cancer development. Based on our recent studies showing that OX1R is aberrantly expressed in colon cancer, we wondered whether orexin-A could play a role in UC. Immunohistochemistry studies revealed that OX1R is highly expressed in the affected colonic epithelium of most UC patients, but not in the non-affected colonic mucosa. Injection of exogenous orexin-A specifically improved the inflammatory symptoms in the two colitis murine models. Conversely, injection of inactive orexin-A analog, OxB7-28 or OX1R specific antagonist SB-408124 did not have anti-inflammatory effect. Moreover, treatment with orexin-A in DSS-colitis induced OX1R-/- knockout mice did not have any protective effect. The orexin-A anti-inflammatory effect was due to the decreased expression of pro-inflammatory cytokines in immune cells and specifically in T-cells isolated from colonic mucosa. Moreover, orexin-A inhibited canonical NFκB activation in an immune cell line and in intestinal epithelial cell line. These results suggest that orexin-A might represent a promising alternative to current UC therapies.
Collapse
Affiliation(s)
- N Messal
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From inflammation to cancer in digestive diseases" labeled by "la Ligue Nationale contre le Cancer", Paris-Diderot University, DHU UNITY, 75018 Paris, France
| | - N Fernandez
- INSERM UMR1149/Inflammation Research Center (CRI), Team "Intestinal inflammation", Paris-Diderot University, DHU UNITY, 75018 Paris, France
| | - S Dayot
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From inflammation to cancer in digestive diseases" labeled by "la Ligue Nationale contre le Cancer", Paris-Diderot University, DHU UNITY, 75018 Paris, France
| | - V Gratio
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From inflammation to cancer in digestive diseases" labeled by "la Ligue Nationale contre le Cancer", Paris-Diderot University, DHU UNITY, 75018 Paris, France
| | - P Nicole
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From inflammation to cancer in digestive diseases" labeled by "la Ligue Nationale contre le Cancer", Paris-Diderot University, DHU UNITY, 75018 Paris, France
| | - C Prochasson
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From inflammation to cancer in digestive diseases" labeled by "la Ligue Nationale contre le Cancer", Paris-Diderot University, DHU UNITY, 75018 Paris, France
| | - I Chantret
- INSERM UMR1149/Inflammation Research Center (CRI), Team "Inflammatory and stress responses in chronic liver diseases", Paris-Diderot University, DHU UNITY, 75018 Paris, France
| | - G LeGuilloux
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From inflammation to cancer in digestive diseases" labeled by "la Ligue Nationale contre le Cancer", Paris-Diderot University, DHU UNITY, 75018 Paris, France
| | - A Jarry
- EA4273 Biometadys, Faculté de Médecine, Université de Nantes, 1 Rue Gaston Veil, 44035 Nantes, France
| | - A Couvelard
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From inflammation to cancer in digestive diseases" labeled by "la Ligue Nationale contre le Cancer", Paris-Diderot University, DHU UNITY, 75018 Paris, France
| | - X Tréton
- INSERM UMR1149/Inflammation Research Center (CRI), Team "Intestinal inflammation", Paris-Diderot University, DHU UNITY, 75018 Paris, France
| | - T Voisin
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From inflammation to cancer in digestive diseases" labeled by "la Ligue Nationale contre le Cancer", Paris-Diderot University, DHU UNITY, 75018 Paris, France
| | - E Ogier-Denis
- INSERM UMR1149/Inflammation Research Center (CRI), Team "Intestinal inflammation", Paris-Diderot University, DHU UNITY, 75018 Paris, France
| | - A Couvineau
- INSERM UMR1149/Inflammation Research Center (CRI), Team "From inflammation to cancer in digestive diseases" labeled by "la Ligue Nationale contre le Cancer", Paris-Diderot University, DHU UNITY, 75018 Paris, France.
| |
Collapse
|