1
|
Cruise S, Secci ME, Kelley LK, Sharfman NM, Rodriguez-Graciani K, Wills TA, Gilpin NW, Avegno EM. Chronic alcohol withdrawal-associated increases in VTA Hcrtr1 expression are associated with heightened nociception and anxiety-like behavior in female rats. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2025; 5:14199. [PMID: 40247884 PMCID: PMC12004375 DOI: 10.3389/adar.2025.14199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/28/2025] [Indexed: 04/19/2025]
Abstract
Alcohol withdrawal is characterized by various symptoms that include pain and negative affect in the absence of the drug. The neural underpinnings of these behaviors are not entirely understood, but orexin has emerged as a candidate target for the treatment of substance use disorders. Here, we explored changes in orexin system-related gene expression in brain regions important for mediating reward and stress, including the ventral tegmental area (VTA) and extended amygdala (including the central amygdala, nucleus accumbens shell, and bed nucleus of the stria terminalis), in adolescent and adult female Wistar rats following chronic alcohol exposure. We observed higher numbers of Hcrtr1- (orexin receptor 1)-expressing neurons in the VTA of adolescent and adult female rats during withdrawal from chronic alcohol exposure. The number of Hcrt1+ VTA neurons was negatively correlated with thermal sensitivity in adolescent female rats and anxiety-like behavior in adult female rats. These data suggest that chronic alcohol effects on orexin receptor expression in the VTA are related to specific behaviors that manifest during withdrawal, highlighting potential avenues for targeting alcohol withdrawal-associated behaviors across development.
Collapse
Affiliation(s)
- Shealan Cruise
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Maria E. Secci
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Leslie K. Kelley
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Nathan M. Sharfman
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Keishla Rodriguez-Graciani
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Tiffany A. Wills
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Alcohol and Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Nicholas W. Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Alcohol and Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Southeast Louisiana VA Healthcare System (SLVHCS), New Orleans, LA, United States
| | - Elizabeth M. Avegno
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Alcohol and Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
2
|
Hosaini M, Abbasnejad M, Kooshki R, Esmaeili-Mahani S, Raoof M, Naderi R, Aarab G, Lobbezoo F. The involvement of orexin-1 receptors in modulation of feeding and anxiety-like behavior in rats with complete Freund's adjuvant-induced temporomandibular joint disorder. Odontology 2025; 113:764-775. [PMID: 39843662 PMCID: PMC11950102 DOI: 10.1007/s10266-024-01021-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 10/29/2024] [Indexed: 01/24/2025]
Abstract
Orexin-A (OXA), a neuropeptide produced in the hypothalamus, is recognized for its role in modulating orofacial nociception and regulating feeding behaviors, as well as its impact on psychophysiological responses. This study investigated the role of orexin-1 receptors (OX1R) in modulating nociceptive behaviors induced by noxious stimulation of the temporomandibular joint (TMJ) and the associated changes in mood and feeding behaviors in rats with complete Freund's adjuvant (CFA)-induced temporomandibular disorders (TMDs). Bilateral cannulation of the lateral ventricles was performed in rats. To induce nociception, CFA was injected unilaterally into the left TMJ of the rats. Nociceptive behaviors were assessed using the hot plate and tail flick tests, while anxiety-like behavior and food intake were evaluated using an elevated plus maze (EPM) and a food preference device, respectively. The results demonstrated a significant increase in nociceptive scores and anxiety-like behaviors, along with reductions in water and food consumption following CFA injection. However, post-treatment with OXA at concentrations of 50 and 100 pM/rat significantly decreased thermal nociceptive scores, alleviated anxiety-like behavior, and increased water and food intake. These beneficial effects were reversed when OXA was co-administered with SB-334867 (40 nM/rat), an OX1R antagonist. Collectively, our findings suggest that OX1R signaling plays a role in the modulation of anxiety-like behavior and abnormalities in food intake in CFA-treated rats. Understanding the involvement of OXA and its receptors in CFA-induced TMJ nociception and behavioral changes may pave the way for potential therapeutic interventions targeting OX1R signaling in the management of TMD-associated symptoms.
Collapse
Affiliation(s)
- Mojtaba Hosaini
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mehdi Abbasnejad
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Razieh Kooshki
- Department of Biology, Faculty of Sciences, Lorestan University, Khorramabad, Iran
| | - Saeed Esmaeili-Mahani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Maryam Raoof
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - Reyhaneh Naderi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Ghizlane Aarab
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Frank Lobbezoo
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Orofacial Pain and Jaw Function, Faculty of Odontology, Malmö University, Malmö, Sweden
| |
Collapse
|
3
|
Abstract
Narcolepsy is a rare, disabling, chronic neurologic disorder that requires lifelong management of symptoms with pharmacologic and nonpharmacologic methods. The pentad symptoms of narcolepsy include excessive daytime sleepiness, cataplexy, disrupted nighttime sleep, sleep paralysis, and hypnagogic/hypnopompic hallucinations. However, people with narcolepsy often experience additional symptoms and disability related to nonpentad symptoms and comorbidities, such as cognitive, psychiatric, metabolic, and sleep disturbances. Current treatment strategies have focused primarily on addressing two of the pentad symptoms, excessive daytime sleepiness, and cataplexy, mainly owing to medication options being approved by the US Food and Drug Administration for these specific indications, neglecting the full 24-h impact and spectrum of symptoms. Meanwhile, the burden of disease extends far beyond these symptoms, and optimal management should reflect a comprehensive, patient-specific approach that not only addresses the entire pentad, but also goes beyond it to include the complete clinical presentation and manifestations of the disease. Individualized treatment must consider the patient's age and stage of life, most debilitating symptoms, support system and structure, comorbid conditions, treatment goals, and overall health. This review discusses care considerations for people living with narcolepsy in the context of their clinical characteristics beyond the hallmark features of narcolepsy.
Collapse
Affiliation(s)
- Anne Marie Morse
- Geisinger Commonwealth School of Medicine, Geisinger Medical Center, Janet Weis Children's Hospital, 100 N. Academy Ave, Danville, PA, 17822, USA.
| | | | - Shelby Harris
- Albert Einstein College of Medicine, New York, NY, USA
| | - Monica Gow
- Wake Up Narcolepsy, Inc., Worcester, MA, USA
| |
Collapse
|
4
|
Zhu Z, Chen G, He J, Xu Y. The protective effects of orexin B in neuropathic pain by suppressing inflammatory response. Neuropeptides 2024; 108:102458. [PMID: 39255695 DOI: 10.1016/j.npep.2024.102458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/19/2024] [Accepted: 07/28/2024] [Indexed: 09/12/2024]
Abstract
Chronic pain induced by pathological insults to the sensorimotor system is a typical form of neuropathic pain (NP), and the underlying mechanism is complex. Currently, there are no successful therapeutic interventions for NP. Orexin B is a neuropeptide with a wide range of biological functions. However, the pharmacological function of orexin B in chronic neuropathic pain has been less studied. Here, we aim to examine the neuroprotective effects of orexin B in chronic constriction injury (CCI)- induced NP. Firstly, we found that orexin type 2 receptor (OX2R) but not orexin type 1 receptor (OX1R) was reduced in the spinal cord (SC) of CCI-treated rats. Mechanical withdrawal threshold and thermal withdrawal latency assays display that administration of orexin B clearly ameliorated CCI-evoked neuropathic pain dose-dependently. Notably, orexin B treatment also effectively prevented microglia activation by reducing the levels of IBA1. Additionally, orexin B was also found to suppress the inflammatory response in the SC tissue by reducing the levels of IL-6, TNF-α, iNOS, and COX-2 as well as the production of NO and PGE2 in CCI-treated rats. Furthermore, orexin B administration attenuated oxidative stress (OS) by increasing the activity of SOD and the levels of GSH. Mechanically, orexin B prevented activation of JNK/NF-κB signaling in the SC of CCI-treated rats. Based on these findings, we conclude that orexin B might have a promising role in ameliorating CCI-evoked neuropathic pain through the inhibition of microglial activation and inflammatory response.
Collapse
Affiliation(s)
- Zuqing Zhu
- Department of Anesthesiology, the First People's Hospital of Linping District, Hangzhou, Zhejiang 311100, China
| | - Gang Chen
- Department of Anesthesiology, Shaoyifu Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, Zhejiang 310018, China
| | - Jiangtao He
- Department of Anesthesiology, the First People's Hospital of Linping District, Hangzhou, Zhejiang 311100, China
| | - Yuanting Xu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 311100, China.
| |
Collapse
|
5
|
Xiang X, Wang F, Chen C, Guan Z, Zhou W. Orexinergic projections to substantia innominata mediate arousal and analgesia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620973. [PMID: 39554139 PMCID: PMC11565723 DOI: 10.1101/2024.10.29.620973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Understanding neural circuits involved in anesthesia is crucial for improving its safety and efficacy. Hypothalamic orexin neurons (LHA OX ), projecting broadly, are essential in regulating arousal and pain. However, the precise targets remain unclear. Here we investigated the orexin projections to the substantia innominata (SI). Combining optogenetics, fiber photometry, and EEG/EMG allowed us to manipulate orexin activities, while simultaneously recording local ligand release and global cortical activities during anesthesia. Brain slice electrophysiology revealed the synaptic connections in the SI, while RNAscope was employed to examine the distribution of orexin receptors and downstream neuronal types. Presynaptic vesicles were identified in the orexin terminals in the SI, where 49.16% of cells expressed OX2R and 6.8% expressed OX1R. Orexin release in the SI was reversibly suppressed by isoflurane. Optogenetic activation of the LHA OX →SI circuit significantly increased orexin release and promoted arousal from various anesthesia stages, including reanimation during 0.75% isoflurane (p < 0.0001), prolongation of 3% isoflurane induction (p = 0.0033), and acceleration of emergence from 2% isoflurane (p < 0.0001). Furthermore, activating this circuit induced analgesia to both thermal (p = 0.0074) and inflammatory (p = 0.0127) pain. Patch-clamp recordings revealed that optogenetic activation of orexin terminals in the SI elicited excitatory postsynaptic currents, which were blocked by the OX2R antagonist. SI contains more GABAergic (28.17%) and glutamatergic (11.96%) neurons than cholinergic neurons (4.13%), all of which expressed OX2R. Thus, LHA OX neurons innervate SI neurons to regulate both arousal and pain predominantly through OX2R.
Collapse
|
6
|
Danesh E, Hassanpour S, Vazir B, Saghafi M, Ghalandari-Shamami M, Haghparast A. The restraint stress-induced antinociceptive effects decreased by antagonism of both orexin receptors within the CA1 region of the hippocampus. Neuropeptides 2024; 107:102463. [PMID: 39180799 DOI: 10.1016/j.npep.2024.102463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
Studies have indicated that stress-related symptoms can lead to hormonal and neural changes, affecting the pain threshold and nociceptive behaviors. The precise role of orexin receptors (OX1r and OX2r) in stress-induced analgesia (SIA) remains an inquiry yet to be comprehensively elucidated. The current investigation aimed to assess the impact of acute immobilization restraint stress on pain-related behavioral responses after administering antagonists targeting OX1r and OX2r in a rat model using the tail-flick test. After a period of five to seven days post-stereotaxic surgery in CA1, the baseline tail-flick latency (TFL) was recorded for each animal. Subsequently, rats were unilaterally administered varying doses of the OX1r antagonist (SB334867; 1, 3, 10, and 30 nmol), the OX2r antagonist (TCS OX2 29; 1, 3, 10, and 30 nmol), or a vehicle (0.5 μl solution containing 12% DMSO) through an implanted cannula. Following a 5-min interval, the animals were subjected to a restraint stress (RS) lasting for 3 h. The tail-flick test was conducted after the stress exposure, and the TFLs were assessed at 60-min intervals. The findings of this study revealed that RS elicits antinociceptive responses in the tail-flick test. Microinjection of OX1r and OX2r antagonists into the CA1 attenuated RS-induced analgesia during the tail-flick test. Furthermore, the results underscored the preeminent role of OX2 receptors in modulating SIA. In conclusion, the orexin system localized within the hippocampal CA1 region may, in part, contribute to the manifestation of SIA in the context of acute pain.
Collapse
Affiliation(s)
- Elaheh Danesh
- Department of Basic Science, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Shahin Hassanpour
- Division of Physiology, Department of basic Science, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Bita Vazir
- Division of Physiology, Department of basic Science, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Saghafi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran; Department of Basic Sciences, Iranian Academy of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Kukkonen JP, Jacobson LH, Hoyer D, Rinne MK, Borgland SL. International Union of Basic and Clinical Pharmacology CXIV: Orexin Receptor Function, Nomenclature and Pharmacology. Pharmacol Rev 2024; 76:625-688. [PMID: 38902035 DOI: 10.1124/pharmrev.123.000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
The orexin system consists of the peptide transmitters orexin-A and -B and the G protein-coupled orexin receptors OX1 and OX2 Orexin receptors are capable of coupling to all four families of heterotrimeric G proteins, and there are also other complex features of the orexin receptor signaling. The system was discovered 25 years ago and was immediately identified as a central regulator of sleep and wakefulness; this is exemplified by the symptomatology of the disorder narcolepsy with cataplexy, in which orexinergic neurons degenerate. Subsequent translation of these findings into drug discovery and development has resulted to date in three clinically used orexin receptor antagonists to treat insomnia. In addition to sleep and wakefulness, the orexin system appears to be a central player at least in addiction and reward, and has a role in depression, anxiety and pain gating. Additional antagonists and agonists are in development to treat, for instance, insomnia, narcolepsy with or without cataplexy and other disorders with excessive daytime sleepiness, depression with insomnia, anxiety, schizophrenia, as well as eating and substance use disorders. The orexin system has thus proved an important regulator of numerous neural functions and a valuable drug target. Orexin prepro-peptide and orexin receptors are also expressed outside the central nervous system, but their potential physiological roles there remain unknown. SIGNIFICANCE STATEMENT: The orexin system was discovered 25 years ago and immediately emerged as an essential sleep-wakefulness regulator. This discovery has tremendously increased the understanding of these processes and has thus far resulted in the market approval of three orexin receptor antagonists, which promote more physiological aspects of sleep than previous hypnotics. Further, orexin receptor agonists and antagonists with different pharmacodynamic properties are in development since research has revealed additional potential therapeutic indications. Orexin receptor signaling is complex and may represent novel features.
Collapse
Affiliation(s)
- Jyrki P Kukkonen
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Laura H Jacobson
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Daniel Hoyer
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Maiju K Rinne
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Stephanie L Borgland
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| |
Collapse
|
8
|
Distler K, Maschauer S, Neu E, Hübner H, Einsiedel J, Prante O, Gmeiner P. Structure-guided discovery of orexin receptor-binding PET ligands. Bioorg Med Chem 2024; 110:117823. [PMID: 38964170 DOI: 10.1016/j.bmc.2024.117823] [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: 05/03/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024]
Abstract
Molecular imaging using positron emission tomography (PET) can serve as a promising tool for visualizing biological targets in the brain. Insights into the expression pattern and the in vivo imaging of the G protein-coupled orexin receptors OX1R and OX2R will further our understanding of the orexin system and its role in various physiological and pathophysiological processes. Guided by crystal structures of our lead compound JH112 and the approved hypnotic drug suvorexant bound to OX1R and OX2R, respectively, we herein describe the design and synthesis of two novel radioligands, [18F]KD23 and [18F]KD10. Key to the success of our structural modifications was a bioisosteric replacement of the triazole moiety with a fluorophenyl group. The 19F-substituted analog KD23 showed high affinity for the OX1R and selectivity over OX2R, while the high affinity ligand KD10 displayed similar Ki values for both subtypes. Radiolabeling starting from the respective pinacol ester precursors resulted in excellent radiochemical yields of 93% and 88% for [18F]KD23 and [18F]KD10, respectively, within 20 min. The new compounds will be useful in PET studies aimed at subtype-selective imaging of orexin receptors in brain tissue.
Collapse
Affiliation(s)
- Katharina Distler
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany; FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Simone Maschauer
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaulallee 12, 91054 Erlangen, Germany
| | - Eduard Neu
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany; FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany; FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Jürgen Einsiedel
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany; FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Olaf Prante
- FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany; Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Kussmaulallee 12, 91054 Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany; FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany.
| |
Collapse
|
9
|
Shiromani PJ, Vidal-Ortiz A. Most dynorphin neurons in the zona incerta-perifornical area are active in waking relative to non-rapid-eye movement and rapid-eye movement sleep. Sleep 2024; 47:zsae065. [PMID: 38447008 PMCID: PMC11494376 DOI: 10.1093/sleep/zsae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/26/2024] [Indexed: 03/08/2024] Open
Abstract
Dynorphin is an endogenous opiate localized in many brain regions and spinal cord, but the activity of dynorphin neurons during sleep is unknown. Dynorphin is an inhibitory neuropeptide that is coreleased with orexin, an excitatory neuropeptide. We used microendoscopy to test the hypothesis that, like orexin, the dynorphin neurons are wake-active. Dynorphin-cre mice (n = 3) were administered rAAV8-Ef1a-Con/Foff 2.0-GCaMP6M into the zona incerta-perifornical area, implanted with a GRIN lens (gradient reflective index), and electrodes to the skull that recorded sleep. One month later, a miniscope imaged calcium fluorescence in dynorphin neurons during multiple bouts of wake, non-rapid-eye movement (NREM), and rapid-eye movement (REM) sleep. Unbiased data analysis identified changes in calcium fluorescence in 64 dynorphin neurons. Most of the dynorphin neurons (72%) had the highest fluorescence during bouts of active and quiet waking compared to NREM or REM sleep; a subset (20%) were REM-max. Our results are consistent with the emerging evidence that the activity of orexin neurons can be classified as wake-max or REM-max. Since the two neuropeptides are coexpressed and coreleased, we suggest that dynorphin-cre-driven calcium sensors could increase understanding of the role of this endogenous opiate in pain and sleep.
Collapse
Affiliation(s)
- Priyattam J Shiromani
- Laboratory of Sleep Medicine and Chronobiology, Research Service, Ralph H. Johnson Veterans Healthcare System Charleston, SC, USA
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Aurelio Vidal-Ortiz
- Laboratory of Sleep Medicine and Chronobiology, Research Service, Ralph H. Johnson Veterans Healthcare System Charleston, SC, USA
| |
Collapse
|
10
|
Stanyer EC, Hoffmann J, Holland PR. Orexins and primary headaches: an overview of the neurobiology and clinical impact. Expert Rev Neurother 2024; 24:487-496. [PMID: 38517280 PMCID: PMC11034548 DOI: 10.1080/14737175.2024.2328728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/19/2024] [Indexed: 03/23/2024]
Abstract
INTRODUCTION Primary headaches, including migraines and cluster headaches, are highly prevalent disorders that significantly impact quality of life. Several factors suggest a key role for the hypothalamus, including neuroimaging studies, attack periodicity, and the presence of altered homeostatic regulation. The orexins are two neuropeptides synthesized almost exclusively in the lateral hypothalamus with widespread projections across the central nervous system. They are involved in an array of functions including homeostatic regulation and nociception, suggesting a potential role in primary headaches. AREAS COVERED This review summarizes current knowledge of the neurobiology of orexins, their involvement in sleep-wake regulation, nociception, and functions relevant to the associated symptomology of headache disorders. Preclinical reports of the antinociceptive effects of orexin-A in preclinical models are discussed, as well as clinical evidence for the potential involvement of the orexinergic system in headache. EXPERT OPINION Several lines of evidence support the targeted modulation of orexinergic signaling in primary headaches. Critically, orexins A and B, acting differentially via the orexin 1 and 2 receptors, respectively, demonstrate differential effects on trigeminal pain processing, indicating why dual-receptor antagonists failed to show clinical efficacy. The authors propose that orexin 1 receptor agonists or positive allosteric modulators should be the focus of future research.
Collapse
Affiliation(s)
- Emily C. Stanyer
- Headache Group, Wolfson Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- Sir Jules Thorne Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jan Hoffmann
- Headache Group, Wolfson Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Philip R. Holland
- Headache Group, Wolfson Sensory, Pain and Regeneration Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| |
Collapse
|
11
|
Sadeghi M, Zareie F, Gholami M, Nazari-Serenjeh F, Ghalandari-Shamami M, Haghparast A. Contribution of the intra-hippocampal orexin system in the regulation of restraint stress response to pain-related behaviors in the formalin test. Behav Pharmacol 2024; 35:103-113. [PMID: 37934654 DOI: 10.1097/fbp.0000000000000755] [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: 11/09/2023]
Abstract
Stress-induced antinociception (SIA) is due to the activation of several neural pathways and neurotransmitters that often suppress pain perception. Studies have shown that the orexin neuropeptide system is essential in pain modulation. Therefore, this study aimed to investigate the role of orexinergic receptors in the hippocampal CA1 region in modulating SIA response during the formalin test as an animal model of inflammatory pain. The orexin-1 receptor (OX1r) antagonist, SB334867, at 1, 3, 10, and 30 nmol or TCS OX2 29 as an orexin-2 receptor (OX2r) antagonist at the same doses were microinjected into the CA1 region in rats. Five minutes later, rats were exposed to restraint stress (RS) for 3 h, and pain-related behaviors were monitored in 5-min blocks for the 60-min test period in the formalin test. Results showed that applying RS for 3 h reduced pain responses in the early and late phases of the formalin test. The main findings showed that intra-CA1 injection of orexin receptor antagonists reduced the antinociception caused by stress in both phases of the formalin test. In addition, the contribution of OX2r in mediating the antinociceptive effect of stress was more prominent than that of OX1r in the early phase of the formalin test. However, in the late phase, both receptors worked similarly. Accordingly, the orexin system and its two receptors in the CA1 region of the hippocampus regulate SIA response to this animal model of pain in formalin test.
Collapse
Affiliation(s)
- Mehdi Sadeghi
- Department of Physiology, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr
| | - Fatemeh Zareie
- Neurobiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran
| | - Masoumeh Gholami
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak
| | | | | | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Bolouri-Roudsari A, Baghani M, Askari K, Mazaheri S, Haghparast A. The integrative role of orexin-1 and orexin-2 receptors within the hippocampal dentate gyrus in the modulation of the stress-induced antinociception in the formalin pain test in the rat. Behav Pharmacol 2024; 35:14-25. [PMID: 37578388 DOI: 10.1097/fbp.0000000000000737] [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: 08/15/2023]
Abstract
The stressful experiences, by triggering a cascade of hormonal and neural changes, can produce antinociception commonly referred to as stress-induced antinociception (SIA). Orexin neuropeptides have an essential role in stress responses and pain modulation. The dentate gyrus receives orexinergic projections and has been shown to be involved in pain processing. The current study investigated the possible role of orexin-1 and orexin-2 receptors (OX1r and OX2r, respectively) within the dentate gyrus in SIA in a rat model of formalin-induced pain behavior in one hind paw. Male Wistar rats weighing 230-250 g underwent stereotaxic surgery and a cannula was implanted in their brains, above the dentate gyrus region. Either SB334867 or TCS OX2 29 (OX1r and OX2r antagonists, respectively) was microinjected into the dentate gyrus region at a range of doses at 1, 3, 10, and 30 nmol (control group received DMSO 12% as vehicle), 5 min before the forced swim stress (FSS) exposure. The formalin test was performed to assess pain-related behaviors. The results indicated that FSS exposure relieves pain-related behavior in the early and late phases of the formalin test. Blockade of intra-dentate gyrus OX1 or OX2 receptors reduced the antinociceptive responses induced by FSS in the formalin test, with more impact during the late phase. Our findings support the potential role of intra-dentate gyrus orexin receptors as target sites of orexin neurons in painful and stressful situations. Therefore, understanding the exact mechanisms of SIA and the role of the orexinergic system in this phenomenon can lead to identifying the strategies to guide future research and offer a new approach to discovering new pain therapeutic agents.
Collapse
Affiliation(s)
- Arad Bolouri-Roudsari
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University
| | - Matin Baghani
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University
| | | | - Sajad Mazaheri
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences
| |
Collapse
|
13
|
Hastings LE, Frye EV, Carlson ER, Chuong V, Matthews AN, Koob GF, Vendruscolo LF, Marchette RCN. Cold nociception as a measure of hyperalgesia during spontaneous heroin withdrawal in mice. Pharmacol Biochem Behav 2024; 235:173694. [PMID: 38128767 PMCID: PMC10842911 DOI: 10.1016/j.pbb.2023.173694] [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: 08/10/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Opioids are powerful analgesic drugs that are used clinically to treat pain. However, chronic opioid use causes compensatory neuroadaptations that result in greater pain sensitivity during withdrawal, known as opioid withdrawal-induced hyperalgesia (OWIH). Cold nociception tests are commonly used in humans, but preclinical studies often use mechanical and heat stimuli to measure OWIH. Thus, further characterization of cold nociception stimuli is needed in preclinical models. We assessed three cold nociception tests-thermal gradient ring (5-30 °C, 5-50 °C, 15-40 °C, and 25-50 °C), dynamic cold plate (4 °C to -1 °C at -1 °C/min, -1 °C to 4 °C at +1 °C/min), and stable cold plate (10 °C, 6 °C, and 2 °C)-to measure hyperalgesia in a mouse protocol of heroin dependence. On the thermal gradient ring, mice in the heroin withdrawal group preferred warmer temperatures, and the results depended on the ring's temperature range. On the dynamic cold plate, heroin withdrawal increased the number of nociceptive responses, with a temperature ramp from 4 °C to -1 °C yielding the largest response. On the stable cold plate, heroin withdrawal increased the number of nociceptive responses, and a plate temperature of 2 °C yielded the most significant increase in responses. Among the three tests, the stable cold plate elicited the most robust change in behavior between heroin-dependent and nondependent mice and had the highest throughput. To pharmacologically characterize the stable cold plate test, we used μ-opioid and non-opioid receptor-targeting drugs that have been previously shown to reverse OWIH in mechanical and heat nociception assays. The full μ-opioid receptor agonist methadone and μ-opioid receptor partial agonist buprenorphine decreased OWIH, whereas the preferential μ-opioid receptor antagonist naltrexone increased OWIH. Two N-methyl-d-aspartate receptor antagonists (ketamine, MK-801), a corticotropin-releasing factor 1 receptor antagonist (R121919), a β2-adrenergic receptor antagonist (butoxamine), an α2-adrenergic receptor agonist (lofexidine), and a 5-hydroxytryptamine-3 receptor antagonist (ondansetron) had no effect on OWIH. These data demonstrate that the stable cold plate at 2 °C yields a robust, reliable, and concise measure of OWIH that is sensitive to opioid agonists.
Collapse
Affiliation(s)
- Lyndsay E Hastings
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Emma V Frye
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Erika R Carlson
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Vicky Chuong
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA; Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Intitute on Drug Abuse, Intramural Research Program, and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, Baltimore, MD, USA
| | - Aniah N Matthews
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Leandro F Vendruscolo
- Stress and Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, Baltimore, MD, USA
| | - Renata C N Marchette
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA.
| |
Collapse
|
14
|
Abdelmissih S. A Bitter Experience That Enlightens the Future: COVID-19 Neurological Affection and Perspectives on the Orexigenic System. Cureus 2022; 14:e30788. [DOI: 10.7759/cureus.30788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
|
15
|
Youn DH, Jun J, Kim TW, Park K. Spinal orexin A attenuates opioid-induced mechanical hypersensitivity in the rat. Korean J Pain 2022; 35:433-439. [PMID: 36175342 PMCID: PMC9530684 DOI: 10.3344/kjp.2022.35.4.433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/04/2022] [Accepted: 08/09/2022] [Indexed: 11/05/2022] Open
Abstract
Background Repeated administration of opioid analgesics for pain treatment can produce paradoxical hyperalgesia via peripheral and/or central mechanisms. Thus, this study investigated whether spinally (centrally) administered orexin A attenuates opioid-induced hyperalgesia (OIH). Methods [D-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO), a selective μ-opioid receptor agonist, was used to induce mechanical hypersensitivity and was administered intradermally (4 times, 1-hour intervals) on the rat hind paw dorsum. To determine whether post- or pretreatments with spinal orexin A, dynorphin A, and anti-dynorphin A were effective in OIH, the drugs were injected through an intrathecal catheter whose tip was positioned dorsally at the L3 segment of the spinal cord (5 μg for all). Mechanical hypersensitivity was assessed using von Frey monofilaments. Results Repeated intradermal injections of DAMGO resulted in mechanical hypersensitivity in rats, lasting more than 8 days. Although the first intrathecal treatment of orexin A on the 6th day after DAMGO exposure did not show any significant effect on the mechanical threshold, the second (on the 8th day) significantly attenuated the DAMGO-induced mechanical hypersensitivity, which disappeared when the type 1 orexin receptor (OX1R) was blocked. However, intrathecal administration of dynorphin or an anti-dynorphin antibody (dynorphin antagonists) had no effect on DAMGO-induced hypersensitivity. Lastly, pretreatment with orexin A, dynorphin, or anti-dynorphin did not prevent DAMGO-induced mechanical hypersensitivity. Conclusions Spinal orexin A attenuates mechanical hyperalgesia induced by repetitive intradermal injections of DAMGO through OX1R. These data suggest that OIH can be potentially treated by activating the orexin A-OX1R pathway in the spinal dorsal horn.
Collapse
Affiliation(s)
- Dong-Ho Youn
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Korea.,Advanced Dental Device Development Institute, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Jiyeon Jun
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Korea.,Advanced Dental Device Development Institute, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Tae Wan Kim
- Department of Physiology, College of Veterinary Medicine, Kyungpook National University, Daegu, Korea
| | - Kibeom Park
- Department of Anesthesiology and Pain Medicine, Keimyung University Dongsan Hospital, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
16
|
Deli SB, Bonab SI, Khakpay R, Khakpai F, Feyzi MH. An interaction between basolateral amygdala orexinergic and endocannabinoid systems in inducing anti-nociception in the rat formalin test. Psychopharmacology (Berl) 2022; 239:3171-3184. [PMID: 35918531 DOI: 10.1007/s00213-022-06199-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 07/22/2022] [Indexed: 11/29/2022]
Abstract
The amygdala has emerged as the main brain center for the emotional affective dimension of pain and pain modulation. In the amygdala, orexin and cannabinoid receptors are expressed in relatively high concentrations. To investigate the possible interaction between the amygdala orexin and cannabinoid systems on the modulation of inflammatory pain, we conducted formalin, rotarod, and plethysmometer tests, as well as analyzing mRNA expression of orexin and cannabinoid receptors in male rats. The basolateral amygdala (BLA) was unilaterally implanted by a guide cannula. Our results showed that, compared to saline and DMSO/saline, intra-BLA microinjection of orexin-A (50 and 100 µM) decreased flinch response in the early phase, but not in the late phase of the formalin test. However, these injections had no significant effect on the mRNA expression level of BLA, orexin receptor type-1 (Orx1), and cannabinoid receptor type-1 (Cb1). Moreover, intra-BLA administration of Orx1 receptor antagonist (SB-334867; 50 nM) and Cb1 receptor antagonist (AM251; 250 and 500 nM) decreased flinch response only in the early phase of the formalin test as compared to the DMSO group. Although the intra-BLA infusion of orexin-A alone and along with SB-334867 or AM251 decreased flinch response in the early phase of the formalin test, intra-BLA co-microinjection of SB-334867/AM251/OrxA increased flinch response in both early and late phases of the formalin test when compared to the DMSO/OrxA group. Interestingly, in the SB-334867/AM251/OrxA group, the Cb1 receptor was upregulated in all groups in comparison to Orx1 receptors. Our results revealed an interaction between BLA, orexin-A, and Cb1 receptors in inducing anti-nociception in the formalin test.
Collapse
Affiliation(s)
- Soghra Borneh Deli
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Samira Iman Bonab
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Roghaieh Khakpay
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Fatemeh Khakpai
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | |
Collapse
|
17
|
Moreno-Ajona D, Hoffmann J. From basic mechanisms to therapeutic perspectives in cluster headache. Curr Opin Neurol 2022; 35:336-342. [PMID: 35674077 DOI: 10.1097/wco.0000000000001055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The pathophysiological understanding of cluster headache has evolved significantly over the past years. Although it is now well known that the trigeminovascular system, the parasympathetic system and the hypothalamus play important roles in its pathomechanism, we increasingly understand the functional role several neurotransmitters and hormones play in the communication between these structures. RECENT FINDINGS This work will give an overview of the current understanding of the role of calcitonin gene-related peptide, vasoactive intestinal peptide, pituitary adenylate cyclase-activating peptide, melatonin and orexins in cluster headache. On the basis of recent evidence, this study will also review the relevance of the monoclonal calcitonin gene-related peptide antibody galcanezumab as well as the sleep-regulating hormone melatonin in the treatment of cluster headache. SUMMARY Herein, we aim to review the basic mechanisms implicated in the pathophysiology of cluster headache and how the increased mechanistic understanding may lead to the discovery of novel therapeutic targets.
Collapse
Affiliation(s)
- David Moreno-Ajona
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London.,NIHR-Wellcome Trust King's Clinical Research Facility/SLaM Biomedical Research Centre, King's College Hospital, London, UK
| | - Jan Hoffmann
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London.,NIHR-Wellcome Trust King's Clinical Research Facility/SLaM Biomedical Research Centre, King's College Hospital, London, UK
| |
Collapse
|
18
|
Wang C, Chen M, Qin C, Qu X, Shen X, Liu S. Lateral Hypothalamic Orexin Neurons Mediate the Reward Effects of Pain Relief Induced by Electroacupuncture. Front Mol Neurosci 2022; 15:812035. [PMID: 35299694 PMCID: PMC8923289 DOI: 10.3389/fnmol.2022.812035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/03/2022] [Indexed: 12/29/2022] Open
Abstract
The reward of pain relief caused by acupuncture has been found to be clinically significant. However, the molecular mechanisms underlying acupuncture-induced reward of pain relief in chronic pain remain unclear and have not been analyzed in suitable preclinical models. Here, we investigated whether acupuncture could potentially induce the reward of pain relief and orexin neuronal signaling in the lateral hypothalamus (LH) and exhibit a possible role in electroacupuncture (EA)-induced reward in spared nerve injury (SNI) rats. Therefore, by using conditioned place preference (CPP) paradigm, we noticed that EA induced the preference for cues associated with EA-induced pain relief in the early, but not late, phase of chronic pain. These observations were different from the immediate antihyperalgesic effects of EA. c-Fos/orexin double labeling revealed that EA stimulation on 14 days but not on 28 days after SNI modeling activated greater numbers of c-Fos positive orexin neurons in the LH after the CPP test. Moreover, the administration of an orexin-A antagonist in the LH significantly blocked the reward effects of pain relief induced by EA. Furthermore, by using cholera toxin b subunit combined with c-Fos detection, we found that the orexin circuit from the LH to the nucleus accumbens (NAc) shell was significantly activated after EA induced CPP. Microinjection of the orexin antagonist into the NAc shell substantially attenuated the CPP induced by EA. Intravenous injection of low-dose orexin-A together with EA resulted in significantly greater antihyperalgesia effects and CPP scores. Together, these findings clearly demonstrated that LH orexin signaling could potentially play a critical role in the reward effects of pain relief induced by acupuncture. The observations of the present study extended our understanding of orexin signaling in the LH and its role in EA-induced reward, providing new insights into the mechanisms of acupuncture analgesia.
Collapse
Affiliation(s)
| | | | | | | | | | - Sheng Liu
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
19
|
Ghalebandi S, Zareie F, Askari K, Yuzugulen J, Haghparast A. Intra-CA1 injection of orexin receptors antagonism attenuates the stress-induced analgesia in a rat acute pain model. Behav Brain Res 2022; 423:113785. [PMID: 35122794 DOI: 10.1016/j.bbr.2022.113785] [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: 09/30/2021] [Revised: 01/31/2022] [Accepted: 01/31/2022] [Indexed: 11/27/2022]
Abstract
Orexins or hypocretins are excitatory neuropeptides predominantly produced by neuronal clusters in the lateral hypothalamus. The orexinergic system's involvement in pain modulation makes it a candidate for pain control alternative to the opioid system. Moreover, orexin-1 and orexin -2 receptors (OX1r and OX2r, respectively) play a role in responsiveness to stressful stimuli. Some evidence indicates that the Cornu Ammonis 1 (CA1) region of the hippocampus potentially participates in the modulation of both pain and stress. In quest of better understanding the interaction between orexin receptors and stress-induced analgesia (SIA), The present study examined the involvement of OX1r and OX2r within the CA1 in response to acute pain after exposure to forced swim stress (FSS) for a 6-min period. Adult male Wistar rats received different doses of OX1r antagonist (SB334867; 1, 3, 10, and 30 nmol), OX2r antagonist (TCS OX2 29; 3, 10, 30 and 100 nmol), or vehicle (0.5 μl DMSO) through an implanted cannula. After that, animals individually experienced acute pain by performing the tail-flick test. Results indicated that FSS produces antinociceptive responses in the tail-flick test. Blockade of both orexin receptors within the CA1 region attenuated the analgesic effect of FSS. The antinociceptive effect of swim stress was prevented by lower doses of SB334867 than TCS OX2 29. These findings show that the orexinergic system might be partially involved in the SIA via the OX1 and OX2 receptors in the hippocampal CA1 region.
Collapse
Affiliation(s)
- Seyedehdelaram Ghalebandi
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, T.R. North Cyprus via Mersin 10, Turkey
| | - Fatemeh Zareie
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kobra Askari
- Department of Animal Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Jale Yuzugulen
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, T.R. North Cyprus via Mersin 10, Turkey
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Tachibana D, Nakamoto K, Tokuyama S. Changes in median eminence of fatty acid-binding protein 3 in a mouse model of pain. Neuropsychopharmacol Rep 2022; 42:52-58. [PMID: 35090101 PMCID: PMC8919114 DOI: 10.1002/npr2.12225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 11/09/2022] Open
Abstract
AIMS Fatty acid-binding protein (FABP) regulates polyunsaturated fatty acid (PUFA) intracellular trafficking and signal transduction. Our previous studies demonstrated that the alteration of PUFA in the hypothalamus is involved in pain process. However, how FABP subtypes change during pain remain unclear. Here, we examined the expression changes and localization in the hypothalamic FABP subtype in postoperative pain model mice. METHODS Paw incision-induced postoperative methods were adopted as a pain model in male ddY mice. Mechanical allodynia was examined using the von Frey test. The analysis of several FABPs mRNA was measured by real-time PCR, and cellular localization of its protein level was measured by immunofluorescent study. RESULTS Postoperative pain mouse elicited mechanical allodynia on Day 2 after paw incision, and mRNA expression of FABP3 increased significantly in the hypothalamus in the postoperative pain mouse model compared to that in control mice. FABP3 protein expressed in the median eminence and the arcuate nucleus, and colocalized with Iba-1, which is a microglial cell marker. Its protein level significantly increased in the median eminence on Day 2 after incision and returned to the control level on Day 4 after incision. CONCLUSIONS Our findings indicate that FABP3 in the median eminence may change in pain stimuli and may represent a molecular link controlling pain.
Collapse
Affiliation(s)
- Dan Tachibana
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Kazuo Nakamoto
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Shogo Tokuyama
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| |
Collapse
|
21
|
Madaan P, Behl T, Sehgal A, Singh S, Sharma N, Yadav S, Kaur S, Bhatia S, Al-Harrasi A, Abdellatif AAH, Ashraf GM, Abdel-Daim MM, Dailah HG, Anwer MK, Bungau S. Exploring the Therapeutic Potential of Targeting Purinergic and Orexinergic Receptors in Alcoholic Neuropathy. Neurotox Res 2022; 40:646-669. [DOI: 10.1007/s12640-022-00477-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/13/2022] [Accepted: 01/19/2022] [Indexed: 12/11/2022]
|
22
|
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: 0.5] [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
|
23
|
Azeez IA, Igado OO, Olopade JO. An overview of the orexinergic system in different animal species. Metab Brain Dis 2021; 36:1419-1444. [PMID: 34224065 DOI: 10.1007/s11011-021-00761-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 06/06/2021] [Indexed: 01/13/2023]
Abstract
Orexin (hypocretin), is a neuropeptide produced by a subset of neurons in the lateral hypothalamus. From the lateral hypothalamus, the orexin-containing neurons project their fibres extensively to other brain structures, and the spinal cord constituting the central orexinergic system. Generally, the term ''orexinergic system'' usually refers to the orexin peptides and their receptors, as well as to the orexin neurons and their projections to different parts of the central nervous system. The extensive networks of orexin axonal fibres and their terminals allow these neuropeptidergic neurons to exert great influence on their target regions. The hypothalamic neurons containing the orexin neuropeptides have been implicated in diverse functions, especially related to the control of a variety of homeostatic functions including feeding behaviour, arousal, wakefulness stability and energy expenditure. The broad range of functions regulated by the orexinergic system has led to its description as ''physiological integrator''. In the last two decades, the orexinergic system has been a topic of great interest to the scientific community with many reports in the public domain. From the documentations, variations exist in the neuroanatomical profile of the orexinergic neuron soma, fibres and their receptors from animal to animal. Hence, this review highlights the distinct variabilities in the morphophysiological aspects of the orexinergic system in the vertebrate animals, mammals and non-mammals, its presence in other brain-related structures, including its involvement in ageing and neurodegenerative diseases. The presence of the neuropeptide in the cerebrospinal fluid and peripheral tissues, as well as its alteration in different animal models and conditions are also reviewed.
Collapse
Affiliation(s)
- Idris A Azeez
- Department of Veterinary Anatomy, University of Jos, Jos, Nigeria
| | - Olumayowa O Igado
- Department of Veterinary Anatomy, University of Ibadan, Ibadan, Nigeria
| | - James O Olopade
- Department of Veterinary Anatomy, University of Ibadan, Ibadan, Nigeria.
| |
Collapse
|
24
|
Pourreza P, Haghparast A, Sadeghi M, Nazari-Serenjeh F, Askari K, Haghparast A. Orexin-2 receptor antagonism in the cornu ammonis 1 region of hippocampus prevented the antinociceptive responses induced by chemical stimulation of the lateral hypothalamus in the animal model of persistent pain. Behav Pharmacol 2021; 32:515-523. [PMID: 34320521 DOI: 10.1097/fbp.0000000000000646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Orexins are excitatory neuropeptides, mainly produced by neurons located in the lateral hypothalamus, which project to many brain areas. The orexinergic system plays a fundamental role in arousal, sleep/wakefulness, feeding, energy homeostasis, motivation, reward, stress and pain modulation. As a prominent part of the limbic system, the hippocampus has been involved in formalin-induced nociception modulation. Moreover, hippocampus regions express both orexin-1 (OX1) and orexin-2 (OX2) receptors. The present study investigated the role of OX2 receptors (OX2R) within the cornu ammonis 1 (CA1) region of the hippocampus in the mediation of lateral hypothalamus-induced antinociception. Fifty-three male Wistar rats were unilaterally implanted with two separate cannulae into the lateral hypothalamus and CA1. Animals were pretreated with intra-CA1 TCS OX2 29 as an OX2R antagonist before intra-lateral hypothalamus administration of carbachol (250 nM) as a muscarinic agonist for chemical stimulation of orexinergic neurons. Formalin test was used as an animal model of persistent pain, following intra-lateral hypothalamus carbachol microinjection. Results showed that the chemical stimulation of the lateral hypothalamus significantly attenuated formalin-evoked nociceptive behaviors during both phases of the formalin test, and administration of TCS OX2 29 into the CA1 blocked these antinociceptive responses in both phases, especially in the late phase. These findings suggest that OX2 receptors in the CA1 partially mediate the lateral hypothalamus-induced antinociceptive responses in persistent inflammatory pain.
Collapse
Affiliation(s)
- Pooya Pourreza
- Department of Basic Sciences, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad university
| | - Amir Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran
| | - Mehdi Sadeghi
- Department of Physiology, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr
| | | | - Kobra Askari
- Department of Animal Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran
| |
Collapse
|
25
|
Askari K, Oryan S, Eidi A, Zaringhalam J, Haghparast A. Modulatory role of the orexin system in stress-induced analgesia: Involvement of the ventral tegmental area. Eur J Pain 2021; 25:2266-2277. [PMID: 34288265 DOI: 10.1002/ejp.1840] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/17/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Exposure to stressful experiences is often accompanied by suppressing pain perception, referred to as stress-induced analgesia. The neuropeptides orexins are essential in regulating the mechanism that responds to stressful and painful stimuli. Meanwhile, the ventral tegmental area (VTA), as a part of descending pain inhibitory system, responds to noxious stimuli. This study aimed to investigate the role of intra-VTA administration of orexin receptor antagonists on stress-induced antinociceptive responses in the animal model of acute pain. METHOD Ninety-three adult Wistar rats weighing 230-250 g were unilaterally implanted by a cannulae above the VTA. Animals were pretreated with different doses (1, 3, 10 and 30 nM/0.3 μl) of SB334867 as the orexin-1 receptor antagonist and TCS OX2 29 as the orexin-2 receptor antagonist into the VTA, just 5 min before 6 min exposure to forced swim stress (FSS). Nociceptive threshold was measured using the tail-flick test as a model of acute pain. RESULTS The results showed that exposure to FSS could significantly increase analgesic responses. Moreover, intra-VTA administration of SB334768 and TCS OX2 29 blocked the antinociceptive effect of FSS in the tail-flick test. CONCLUSION The findings suggest that OX1 and OX2 receptors in the VTA might modulate the antinociceptive behaviours induced by FSS in part. SIGNIFICANCE Acute exposure to physical stress suppresses pain-related behaviors in the animal model of acute pain. Blockade of the OX1 and OX2 receptors in the VTA attenuates antinociceptive responses induced by FSS. The contribution of the OX2 receptors in the VTA is more predominant than OX1 receptors in stress-induced analgesia.
Collapse
Affiliation(s)
- Kobra Askari
- Department of Animal Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Shahrbanoo Oryan
- Department of Animal Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Akram Eidi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Jalal Zaringhalam
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Abstract
Background While understanding the pathophysiology of migraine has led to CGRP-based treatments, other potential targets have also been implicated in migraine. Objectives To catalog new promising targets for the treatment of migraine. Methods We completed a literature review focusing on 5HT1F, PACAP, melatonin, and orexins. Results The 5HT1F receptor agonist lasmiditan, following two positive randomized placebo-controlled trials, was FDA-approved for the acute treatment of migraine. PACAP-38 has shown analogous evidence to what was obtained for CGRP with its localization in key structures, provocation tests, and positive studies when antagonizing its receptor in animal models, although a PAC-1 receptor monoclonal antibody study was negative. Melatonin has undergone several randomized controlled trials showing a positive trend. Filorexant is the only dual orexin receptor antagonist, which was tested in humans with negative results. Conclusions Further and ongoing studies will determine the utility of these new therapies with lasmiditan and melatonin having demonstrated efficacy for the treatment of migraine.
Collapse
Affiliation(s)
- David Moreno-Ajona
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - María Dolores Villar-Martínez
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Peter James Goadsby
- NIHR-Wellcome Trust King's Clinical Research Facility/SLaM Biomedical Research Centre, King's College Hospital, London, United Kingdom; Department of Neurology, University of California, Los Angeles, Los Angeles CA USA
| |
Collapse
|
27
|
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: 24] [Impact Index Per Article: 6.0] [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
|
28
|
Orexinergic descending inhibitory pathway mediates linalool odor-induced analgesia in mice. Sci Rep 2021; 11:9224. [PMID: 33927235 PMCID: PMC8085205 DOI: 10.1038/s41598-021-88359-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/12/2021] [Indexed: 11/08/2022] Open
Abstract
Linalool odor exposure induces an analgesic effect in mice. This effect disappeared in the anosmic model mice, indicating that olfactory input evoked by linalool odor triggered this effect. Furthermore, hypothalamic orexinergic neurons play a pivotal role in this effect. However, the neuronal circuit mechanisms underlying this effect have not been fully addressed. In this study, we focused on the descending orexinergic projection to the spinal cord and examined whether this pathway contributes to the effect. We assessed the effect of intrathecal administration of orexin receptor antagonists on linalool odor-induced analgesia in the tail capsaicin test. We found that the selective orexin type 1 receptor antagonist, but not the selective orexin type 2 receptor antagonist, prevented the odor-induced analgesic effect. Furthermore, immunohistochemical analyses of c-Fos expression induced by the capsaicin test revealed that neuronal activity of spinal cord neurons was suppressed by linalool odor exposure, which was prevented by intrathecal administration of the orexin 1 receptor antagonist. These results indicate that linalool odor exposure drives the orexinergic descending pathway and suppresses nociceptive information flow at the spinal level.
Collapse
|
29
|
Regulatory Role of Orexin in the Antistress Effect of "Press Tack Needle" Acupuncture Treatment. Healthcare (Basel) 2021; 9:healthcare9050503. [PMID: 33925438 PMCID: PMC8146164 DOI: 10.3390/healthcare9050503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 11/18/2022] Open
Abstract
The aim of this research was to investigate the antistress effect of press tack needle (PTN) acupuncture treatment using rats with social isolation stress (SIS). Rats were divided into non-stress group (Grouped+sham), stress group (SIS+sham), and PTN-treated SIS group (SIS+PTN). Rats in the SIS+PTN and SIS+sham groups were housed alone for eight days. For the SIS+PTN group, a PTN (length, 0.3 or 1.2 mm) was fixed on the GV20 acupoint on day 7. We measured stress behavior based on the time the rats showed aggressive behavior and the levels of plasma corticosterone and orexin A on day 8. In addition, the orexin-1 receptor or orexin-2 receptor antagonist was administered to rats that were exposed to SIS. The duration of aggressive behavior was significantly prolonged in the SIS+sham group, and the prolonged duration was inhibited in the SIS+PTN (1.2 mm) group. The levels of plasma corticosterone and orexin A were significantly increased in the SIS+sham group; however, these increases were inhibited in the SIS+PTN group. The aggressive behavior was significantly reduced after the orexin-2 receptor antagonist was administered. These findings suggest that PTN treatment at GV20 may have an antistress effect, and the control of orexin is a mechanism underlying this phenomenon.
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Despite the development of several medications for the acute and preventive treatment of migraine, there are still many patients in whom lack of efficacy, tolerability, interactions or contraindications make other options necessary. CGRP-based drugs have opened the door to a new era of migraine-targeted treatments. Beyond CGRP, there are other promising targets covered here. RECENT FINDINGS For the acute treatment of migraine, 5-HT1F receptor agonists, ditans, are now available. Unlike triptans, 5-HT1B/1D receptor agonists, cardiovascular disease is not a contraindication for the use of ditans. The first study on a monoclonal antibody targeting PAC1 receptor was negative, although this may not be the end for the pituitary adenylate cyclase-activating polypeptide (PACAP) pathway as a target. SUMMARY Following positive phase-III clinical trials, lasmiditan is the first ditan to be FDA-approved. PACAP has experimental evidence suggesting a role in migraine pathophysiology. As for CGRP, the presence of PACAP in key migraine structures along with positive provocative tests for both PACAP-38 and PACAP-27 indicate this pathway may still be a pharmacological target. Glutamate-based targets have long been considered in migraine. Two clinical trials with memantine, an NMDA-R antagonist, for the preventive treatment of migraine have now been published. The hypothalamus has also been implicated in migraine pathophysiology: the potential role of orexins in migraine is discussed. Acid-sensing ion channels, as well as amylin-blocking drugs, may also become migraine treatments in the future: more research is warranted.
Collapse
|
31
|
Ahmadi-Soleimani SM, Azizi H, Abbasi-Mazar A. Intermittent REM sleep deprivation attenuates the development of morphine tolerance and dependence in male rats. Neurosci Lett 2021; 748:135735. [PMID: 33592307 DOI: 10.1016/j.neulet.2021.135735] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 02/04/2023]
Abstract
Opioid agonists are used in clinic for pain management, however this application is challenged by development of tolerance and dependence following prolonged exposure. Various approaches have been suggested to address this concern, however, there is still no consensus among the researchers. Neural processing of sleep and nociception are co-regulated through shared brain regions having bidirectional interplays. Thus, we aimed to investigate whether application of REM sleep deprivation (REM-SD) could affect morphine analgesic tolerance and dependence. To this end, adult male rats underwent sleep deprivation during light and dark phases (LSD and DSD, respectively) using the inverted flower pot method and then tolerance and dependence was induced by repeated injection of morphine for 7 days (10 mg/kg, daily, i.p.). Results indicated that REM-SD delays the development of tolerance to morphine during both phases; however this effect was more potent following LSD. Moreover, LSD decreased the baseline thermal threshold and total withdrawal score. One possible hypothesis for our observations is REM-SD-induced attenuation of orexin system which is still controversial among the researchers. Other stronger possibilities might be down-regulation of opioid receptors in response to sleep loss experience. Finally, it seems that modification of sleep periods may assist to decrease the severity of opioid tolerance and dependence.
Collapse
Affiliation(s)
- S Mohammad Ahmadi-Soleimani
- Deparment of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Alireza Abbasi-Mazar
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| |
Collapse
|
32
|
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: 1.8] [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]
|
33
|
Szczepanska-Sadowska E, Cudnoch-Jedrzejewska A, Sadowski B. Differential role of specific cardiovascular neuropeptides in pain regulation: Relevance to cardiovascular diseases. Neuropeptides 2020; 81:102046. [PMID: 32284215 DOI: 10.1016/j.npep.2020.102046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 12/11/2022]
Abstract
In many instances, the perception of pain is disproportionate to the strength of the algesic stimulus. Excessive or inadequate pain sensation is frequently observed in cardiovascular diseases, especially in coronary ischemia. The mechanisms responsible for individual differences in the perception of cardiovascular pain are not well recognized. Cardiovascular disorders may provoke pain in multiple ways engaging molecules released locally in the heart due to tissue ischemia, inflammation or cellular stress, and through neurogenic and endocrine mechanisms brought into action by hemodynamic disturbances. Cardiovascular neuropeptides, namely angiotensin II (Ang II), angiotensin-(1-7) [Ang-(1-7)], vasopressin, oxytocin, and orexins belong to this group. Although participation of these peptides in the regulation of circulation and pain has been firmly established, their mutual interaction in the regulation of pain in cardiovascular diseases has not been profoundly analyzed. In the present review we discuss the regulation of the release, and mechanisms of the central and systemic actions of these peptides on the cardiovascular system in the context of their central and peripheral nociceptive (Ang II) and antinociceptive [Ang-(1-7), vasopressin, oxytocin, orexins] properties. We also consider the possibility that they may play a significant role in the modulation of pain in cardiovascular diseases. The rationale for focusing attention on these very compounds was based on the following premises (1) cardiovascular disturbances influence the release of these peptides (2) they regulate vascular tone and cardiac function and can influence the intensity of ischemia - the factor initiating pain signals in the cardiovascular system, (3) they differentially modulate nociception through peripheral and central mechanisms, and their effect strongly depends on specific receptors and site of action. Accordingly, an altered release of these peptides and/or pharmacological blockade of their receptors may have a significant but different impact on individual sensation of pain and comfort of an individual patient.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, Warsaw, Poland.
| | - Agnieszka Cudnoch-Jedrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, Warsaw, Poland
| | - Bogdan Sadowski
- School of Engineering and Health, Bitwy Warszawskiej 1920 r. 18, Warsaw, Poland
| |
Collapse
|
34
|
Meylakh N, Marciszewski KK, Di Pietro F, Macefield VG, Macey PM, Henderson LA. Altered regional cerebral blood flow and hypothalamic connectivity immediately prior to a migraine headache. Cephalalgia 2020; 40:448-460. [PMID: 32164427 DOI: 10.1177/0333102420911623] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND There is evidence of altered resting hypothalamic activity patterns and connectivity prior to a migraine, however it remains unknown if these changes are driven by changes in overall hypothalamic activity levels. If they are, it would corroborate the idea that changes in hypothalamic function result in alteration in brainstem pain processing sensitivity, which either triggers a migraine headache itself or allows an external trigger to initiate a migraine headache. We hypothesise that hypothalamic activity increases immediately prior to a migraine headache and this is accompanied by altered functional connectivity to pain processing sites in the brainstem. METHODS In 34 migraineurs and 26 healthy controls, we collected a series comprising 108 pseudo-continuous arterial spin labelling images and 180 gradient-echo echo planar resting-state functional magnetic resonance volumes to measure resting regional cerebral blood flow and functional connectivity respectively. Images were pre-processed and analysed using custom SPM12 and Matlab software. RESULTS Our results reflect that immediately prior to a migraine headache, resting regional cerebral blood flow decreases in the lateral hypothalamus. In addition, resting functional connectivity strength decreased between the lateral hypothalamus and important regions of the pain processing pathway, such as the midbrain periaqueductal gray, dorsal pons, rostral ventromedial medulla and cingulate cortex, only during this critical period before a migraine headache. CONCLUSION These data suggest altered hypothalamic function and connectivity in the period immediately prior to a migraine headache and supports the hypothesis that the hypothalamus is involved in migraine initiation.
Collapse
Affiliation(s)
- Noemi Meylakh
- Department of Anatomy and Histology, University of Sydney, Sydney, NSW, Australia
| | - Kasia K Marciszewski
- Department of Anatomy and Histology, University of Sydney, Sydney, NSW, Australia
| | - Flavia Di Pietro
- Department of Anatomy and Histology, University of Sydney, Sydney, NSW, Australia
| | | | - Paul M Macey
- UCLA School of Nursing and Brain Research Institute, University of California, Los Angeles, CA, USA
| | - Luke A Henderson
- Department of Anatomy and Histology, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
35
|
Bai P, Bai S, Placzek MS, Lu X, Fiedler SA, Ntaganda B, Wey HY, Wang C. A New Positron Emission Tomography Probe for Orexin Receptors Neuroimaging. Molecules 2020; 25:molecules25051018. [PMID: 32106419 PMCID: PMC7179119 DOI: 10.3390/molecules25051018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 01/16/2023] Open
Abstract
The orexin receptor (OX) is critically involved in motivation and sleep−wake regulation and holds promising therapeutic potential in various mood disorders. To further investigate the role of orexin receptors (OXRs) in the living human brain and to evaluate the treatment potential of orexin-targeting therapeutics, we herein report a novel PET probe ([11C]CW24) for OXRs in the brain. CW24 has moderate binding affinity for OXRs (IC50 = 0.253 μM and 1.406 μM for OX1R and OX2R, respectively) and shows good selectivity to OXRs over 40 other central nervous system (CNS) targets. [11C]CW24 has high brain uptake in rodents and nonhuman primates, suitable metabolic stability, and appropriate distribution and pharmacokinetics for brain positron emission tomography (PET) imaging. [11C]CW24 warrants further evaluation as a PET imaging probe of OXRs in the brain.
Collapse
Affiliation(s)
- Ping Bai
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; (P.B.); (X.L.)
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; (S.B.); (S.A.F.); (B.N.); (H.-Y.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sha Bai
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; (S.B.); (S.A.F.); (B.N.); (H.-Y.W.)
| | - Michael S. Placzek
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; (S.B.); (S.A.F.); (B.N.); (H.-Y.W.)
| | - Xiaoxia Lu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; (P.B.); (X.L.)
| | - Stephanie A. Fiedler
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; (S.B.); (S.A.F.); (B.N.); (H.-Y.W.)
| | - Brenda Ntaganda
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; (S.B.); (S.A.F.); (B.N.); (H.-Y.W.)
| | - Hsiao-Ying Wey
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; (S.B.); (S.A.F.); (B.N.); (H.-Y.W.)
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; (S.B.); (S.A.F.); (B.N.); (H.-Y.W.)
- Correspondence:
| |
Collapse
|
36
|
Fakhoury M, Salman I, Najjar W, Merhej G, Lawand N. The Lateral Hypothalamus: An Uncharted Territory for Processing Peripheral Neurogenic Inflammation. Front Neurosci 2020; 14:101. [PMID: 32116534 PMCID: PMC7029733 DOI: 10.3389/fnins.2020.00101] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/24/2020] [Indexed: 12/20/2022] Open
Abstract
The roles of the hypothalamus and particularly the lateral hypothalamus (LH) in the regulation of inflammation and pain have been widely studied. The LH consists of a parasympathetic area that has connections with all the major parts of the brain. It controls the autonomic nervous system (ANS), regulates feeding behavior and wakeful cycles, and is a part of the reward system. In addition, it contains different types of neurons, most importantly the orexin neurons. These neurons, though few in number, perform critical functions such as inhibiting pain transmission and interfering with the reward system, feeding behavior and the hypothalamic pituitary axis (HPA). Recent evidence has identified a new role for orexin neurons in the modulation of pain transmission associated with several inflammatory diseases, including rheumatoid arthritis and ulcerative colitis. Here, we review recent findings on the various physiological functions of the LH with special emphasis on the orexin/receptor system and its role in mediating inflammatory pain.
Collapse
Affiliation(s)
- Marc Fakhoury
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Israa Salman
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Wassim Najjar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - George Merhej
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nada Lawand
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Neurology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
37
|
Richardson K, Sweatt N, Tran H, Apprey V, Uthayathas S, Taylor R, Gupta K. Significant Quantitative Differences in Orexin Neuronal Activation After Pain Assessments in an Animal Model of Sickle Cell Disease. Front Mol Biosci 2020; 7:5. [PMID: 32118032 PMCID: PMC7025496 DOI: 10.3389/fmolb.2020.00005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 01/10/2020] [Indexed: 12/23/2022] Open
Abstract
Sickle cell disease is a hemoglobinopathy that causes sickling of red blood cells, resulting in vessel blockage, stroke, anemia, inflammation, and extreme pain. The development and treatment of pain, in particular, neuropathic pain in sickle cell disease patients is poorly understood and impedes our progress toward the development of novel therapies to treat pain associated with sickle cell disease. The orexin/hypocretin system offers a novel approach to treat chronic pain and hyperalgesia. These neuropeptides are synthesized in three regions: perifornical area (PFA), lateral hypothalamus (LH), and dorsomedial hypothalamus (DMH). Data suggest that orexin-A neuropeptide has an analgesic effect on inflammatory pain and may affect mechanisms underlying the maintenance of neuropathic pain. The purpose of this study was to determine whether there are neuronal activation differences in the orexin system as a result of neuropathic pain testing in a mouse model of sickle cell disease. Female transgenic sickle mice that express exclusively (99%) human sickle hemoglobin (HbSS-BERK) and age-/gender-matched controls (HbAA-BERK mice; n = 10/group, 20-30 g) expressing normal human hemoglobin A were habituated to each test protocol and environment before collecting baseline measurements and testing. Four measures were used to assess pain-related behaviors: thermal/heat hyperalgesia, cold hyperalgesia, mechanical hyperalgesia, and deep-tissue hyperalgesia. Hypothalamic brain sections from HbAA-BERK and HbSS-BERK mice were processed to visualize orexin and c-Fos immunoreactivity and quantified. The percentage of double labeled neurons in the PFA was significantly higher than the percentage of double labeled neurons in the LH orexin field of HbAA-BERK mice (* p < 0.05). The percentages of double labeled neurons in PFA and DMH orexin fields are significantly higher than those neurons in the LH of HbSS-BERK mice (* p < 0.05). These data suggest that DMH orexin neurons were preferentially recruited during neuropathic pain testing and a more diverse distribution of orexin neurons may be required to produce analgesia in response to pain in the HbSS-BERK mice. Identifying specific orexin neuronal populations that are integral in neuropathic pain processing will allow us to elucidate mechanisms that provide a more selective, targeted approach in treating of neuropathic pain in sickle cell disease.
Collapse
Affiliation(s)
- Kimberlei Richardson
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| | - Nia Sweatt
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| | - Huy Tran
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Victor Apprey
- Department of Family Medicine, Howard University College of Medicine, Washington, DC, United States
| | - Subramaniam Uthayathas
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| | - Robert Taylor
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| | - Kalpna Gupta
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
- Division of Hematology/Oncology, Department of Medicine, University of California-Irvine School of Medicine, Irvine, CA, United States
| |
Collapse
|
38
|
Haghparast A, Matini T, Rezaee L, Rahban M, Tehranchi A, Haghparast A. Involvement of Orexinergic System Within the Nucleus Accumbens in Pain Modulatory Role of the Lateral Hypothalamus in Orofacial Pain Model. Neurochem Res 2020; 45:851-859. [DOI: 10.1007/s11064-020-02957-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/02/2020] [Accepted: 01/08/2020] [Indexed: 10/25/2022]
|
39
|
Hershner S, Dauvilliers Y, Chung F, Singh M, Wong J, Gali B, Kakkar R, Mignot E, Thorpy M, Auckley D. Knowledge Gaps in the Perioperative Management of Adults With Narcolepsy: A Call for Further Research. Anesth Analg 2020; 129:204-211. [PMID: 30882519 DOI: 10.1213/ane.0000000000004088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
There is increasing awareness that sleep disorders may be associated with increased perioperative risk. The Society of Anesthesia and Sleep Medicine created the Narcolepsy Perioperative Task Force: (1) to investigate the current state of knowledge of the perioperative risk for patients with narcolepsy, (2) to determine the viability of developing perioperative guidelines for the management of patients with narcolepsy, and (3) to delineate future research goals and clinically relevant outcomes. The Narcolepsy Perioperative Task Force established that there is evidence for increased perioperative risk in patients with narcolepsy; however, this evidence is sparse and based on case reviews, case series, and retrospective reviews. Mechanistically, there are a number of potential mechanisms by which patients with narcolepsy could be at increased risk for perioperative complications. These include aggravation of the disease itself, dysautonomia, narcolepsy-related medications, anesthesia interactions, and withdrawal of narcolepsy-related medications. At this time, there is inadequate research to develop an expert consensus or guidelines for the perioperative management of patients with narcolepsy. The paucity of available literature highlights the critical need to determine if patients with narcolepsy are at an increased perioperative risk and to establish appropriate research protocols and clearly delineated patient-centered outcomes. There is a real need for collaborative research among sleep medicine specialists, surgeons, anesthesiologists, and perioperative providers. This future research will become the foundation for the development of guidelines, or at a minimum, a better understanding how to optimize the perioperative care of patients with narcolepsy.
Collapse
Affiliation(s)
- Shelley Hershner
- From the Department of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Yves Dauvilliers
- Neurology and Physiology, Department of Neurology, Gui-de-Chauliac Hospital, Montpellier, France
| | - Frances Chung
- Department of Anesthesiology, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Mandeep Singh
- Department of Anesthesia and Pain Management, Women's College Hospital and Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada.,Toronto Sleep and Pulmonary Centre, Toronto, Canada
| | - Jean Wong
- Department of Anesthesiology, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Bhargavi Gali
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Rahul Kakkar
- Narcolepsy Network Inc, Lynnwood, WA.,Prana Health, Doral, Florida
| | - Emmanuel Mignot
- Department of Psychiatry and Behavioral Sciences, Stanford Center for Sleep Sciences and Medicine, Palo Alto, CA
| | - Michael Thorpy
- Saul R. Korey Department of Neurology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Dennis Auckley
- Division of Pulmonary, Critical Care and Sleep Medicine, Metro Health Medical Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
40
|
Yamaguchi M, Ishikawa M, Aono Y, Saigusa T. OX 2 receptors mediate the inhibitory effects of orexin-A on potassium chloride-induced increases in intracellular calcium ion levels in neurons derived from rat dorsal root ganglion in a chronic pain model. Neuropsychopharmacol Rep 2019; 40:30-38. [PMID: 31845549 PMCID: PMC7292216 DOI: 10.1002/npr2.12094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/06/2019] [Accepted: 09/10/2019] [Indexed: 02/06/2023] Open
Abstract
AIMS Orexin-A is known to induce anti-nociceptive effects in animal models of chronic pain. We have found that orexin-A inhibits KCl loading-induced increases in the intracellular calcium ion levels ([Ca2+ ]i ) in C-fiber-like neurons of rats showing inflammatory nociceptive behavior. Here, we examined the effects of orexin-A on the depolarization of C-fiber-like neurons derived from a rat model for another type of chronic pain, namely neuropathic pain. Thus, we analyzed the effects of orexin-A on KCl-induced increases in [Ca2+ ]i in C-fiber-like neurons of rats with sciatic nerve ligation. METHODS Paw withdrawal and threshold force in response to tactile stimuli were evaluated using von Frey filaments. Sham-operated rats served as controls. [Ca2+ ]i in neurons were visualized by calcium fluorescent probe. Changes in [Ca2+ ]i were assessed using relative fluorescence intensity. RESULTS Seven days after sciatic nerve ligation, paw withdrawal and threshold force for tactile stimuli were increased and reduced, respectively. KCl loading to neurons from either sciatic nerve-ligated or control rats increased relative fluorescence intensity. The KCl-induced increase in relative fluorescence intensity in sciatic nerve-ligated, but not that of control, rats was inhibited by orexin-A. The OX1 and OX2 receptor antagonist MK-4305 and OX2 receptor antagonist EMPA, but not the OX1 receptor antagonist SB 334867, each counteracted orexin-A-induced inhibition of KCl-provoked increases in relative fluorescence intensity. CONCLUSION The present findings constitute neuropharmacological evidence that OX2 but not OX1 receptors mediate the inhibitory effects of orexin-A on KCl-induced increases in [Ca2+ ]i in C-fiber-like neurons of rats showing hyperalgesia provoked by sciatic nerve ligation.
Collapse
Affiliation(s)
- Masami Yamaguchi
- Department of Anesthesiology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Manabu Ishikawa
- Department of Anesthesiology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Yuri Aono
- Department of Pharmacology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| | - Tadashi Saigusa
- Department of Pharmacology, Nihon University School of Dentistry at Matsudo, Matsudo, Japan
| |
Collapse
|
41
|
Silva C, McNaughton N. Are periaqueductal gray and dorsal raphe the foundation of appetitive and aversive control? A comprehensive review. Prog Neurobiol 2019; 177:33-72. [DOI: 10.1016/j.pneurobio.2019.02.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 01/19/2019] [Accepted: 02/08/2019] [Indexed: 12/28/2022]
|
42
|
Turku A, Leino TO, Karhu L, Yli-Kauhaluoma J, Kukkonen JP, Wallén EAA, Xhaard H. Structure-Activity Relationships of 1-Benzoylazulenes at the OX 1 and OX 2 Orexin Receptors. ChemMedChem 2019; 14:965-981. [PMID: 30892823 DOI: 10.1002/cmdc.201900074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/20/2019] [Indexed: 11/08/2022]
Abstract
We previously demonstrated the potential of di- or trisubstituted azulenes as ligands (potentiators, weak agonists, and antagonists) of the orexin receptors. In this study we investigated 27 1-benzoylazulene derivatives, uncovering seven potentiators of the orexin response on OX1 and two weak dual orexin receptor agonists. For potentiators, replacement of the azulene scaffold by indole retained the activity of four out of six compounds. The structure-activity relationships for agonism and potentiation can be summarized into a bicyclic aromatic ring system substituted with two hydrogen-bond acceptors (1-position, benzoyl; 6-position, carboxyl/ester) within 7-8 Å of each other; a third acceptor at the 3-position is also well tolerated. The same pharmacophoric signature is found in the preferred conformations of the orexin receptor agonist Nag26 from molecular dynamics simulations. Subtle changes switch the activity between weak agonism and potentiation, suggesting overlapping binding sites.
Collapse
Affiliation(s)
- Ainoleena Turku
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland.,Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 66, 00014, Helsinki, Finland
| | - Teppo O Leino
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Lasse Karhu
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Jyrki P Kukkonen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 66, 00014, Helsinki, Finland.,Department of Physiology, Faculty of Medicine, University of Helsinki, P.O. Box 63, 00014, Helsinki, Finland
| | - Erik A A Wallén
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Henri Xhaard
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| |
Collapse
|
43
|
Spinal Orexin-2 Receptors are Involved in Modulation of the Lateral Hypothalamic Stimulation-Induced Analgesia. Neurochem Res 2019; 44:1152-1158. [DOI: 10.1007/s11064-019-02749-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/01/2019] [Accepted: 02/01/2019] [Indexed: 12/16/2022]
|
44
|
Usui M, Kaneko K, Oi Y, Kobayashi M. Orexin facilitates GABAergic IPSCs via postsynaptic OX 1 receptors coupling to the intracellular PKC signalling cascade in the rat cerebral cortex. Neuropharmacology 2019; 149:97-112. [PMID: 30763655 DOI: 10.1016/j.neuropharm.2019.02.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/06/2019] [Accepted: 02/10/2019] [Indexed: 10/27/2022]
Abstract
Orexin has multiple physiological functions including wakefulness, appetite, nicotine intake, and nociception. The cerebral cortex receives abundant orexinergic projections and expresses both orexinergic receptor 1 (OX1R) and 2 (OX2R). However, little is known about orexinergic regulation of GABA-mediated inhibitory synaptic transmission. In the cerebral cortex, there are multiple GABAergic neural subtypes, each of which has its own morphological and physiological characteristics. Therefore, identification of presynaptic GABAergic neural subtypes is critical to understand orexinergic effects on GABAergic connections. We focused on inhibitory synapses at pyramidal neurons (PNs) from fast-spiking GABAergic neurons (FSNs) in the insular cortex by a paired whole-cell patch-clamp technique, and elucidated the mechanisms of orexin-induced IPSC regulation. We found that both orexin A and orexin B enhanced unitary IPSC (uIPSC) amplitude in FSN→PN connections without changing the paired-pulse ratio or failure rate. These effects were blocked by SB-334867, an OX1 receptor (OX1R) antagonist, but not by TCS-OX2-29, an OX2R antagonist. [Ala11, D-Leu15]-orexin B, a selective OX2R agonist, had little effect on uIPSCs. Variance-mean analysis demonstrated an increase in quantal content without a change in release probability or the number of readily releasable pools. Laser photolysis of caged GABA revealed that orexin A enhanced GABA-mediated currents in PNs. Downstream blockade of Gq/11 protein-coupled OX1Rs by IP3 receptor or protein kinase C (PKC) blockers and BAPTA injection into postsynaptic PNs diminished the orexin A-induced uIPSC enhancement. These results suggest that the orexinergic uIPSC enhancement is mediated via postsynaptic OX1Rs, which potentiate GABAA receptors through PKC activation.
Collapse
Affiliation(s)
- Midori Usui
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Department of Anaesthesiology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Keisuke Kaneko
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Department of Anaesthesiology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Yoshiyuki Oi
- Department of Anaesthesiology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Masayuki Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Dental Research Centre, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan; Molecular Dynamics Imaging Unit, RIKEN Centre for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
45
|
Niknia S, Kaeidi A, Hajizadeh MR, Mirzaei MR, Khoshdel A, Hajializadeh Z, Fahmidehkar MA, Mahmoodi M. Neuroprotective and antihyperalgesic effects of orexin-A in rats with painful diabetic neuropathy. Neuropeptides 2019; 73:34-40. [PMID: 30447858 DOI: 10.1016/j.npep.2018.11.001] [Citation(s) in RCA: 4] [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: 06/20/2018] [Revised: 11/06/2018] [Accepted: 11/06/2018] [Indexed: 01/19/2023]
Abstract
AIM OF STUDY Diabetes mellitus is related to the development of neuronal tissue injury in different peripheral and central nervous system regions. A common complication of diabetes is painful diabetic peripheral neuropathy (PDN). We have studied the neuroprotective and anti-nociceptive properties of neuropeptide orexin-A in an animal experimental model of diabetic neuropathy. METHODS All experiments were carried out on male Wistar rats (220-250 g). Diabetes was induced by a single intraperitoneal injection of 55 mg/kg (i.p.) streptozotocin (STZ). Orexin-A was chronically administrated into the implanted intrathecal catheter (0.6, 2.5 and 5 nM/L, daily, 4 weeks). The tail-flick and rotarod treadmill tests were used to evaluate the nociceptive threshold and motor coordination of these diabetic rats, respectively. Cleaved caspase-3, Bax, Bcl2 and the Bax/Bcl-2 ratio, as the biochemical indicators of apoptosis, were investigated in the dorsal half of the lumbar spinal cord tissue by western blotting method. RESULTS Treatment of the diabetic rats with orexin-A (5 nM/L) significantly attenuated the hyperalgesia and motor deficit in diabetic animals. Furthermore, orexin-A (5 nM/L) administration suppressed pro-apoptotic cleaved caspase-3 and Bax proteins. Also, orexin-A (5 nM/L) reduced the expression of Bax/Bcl-2 ratio in spinal cord dorsal half of rats with PDN. CONCLUSIONS Altogether our data suggest that the orexin-A has anti-hyperalgesic and neuroprotective effects in rats with PDN. Cellular mechanisms underlying the observed effects may, at least partially, be related to reducing the neuronal apoptosis.
Collapse
Affiliation(s)
- Seddigheh Niknia
- Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ayat Kaeidi
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Reza Hajizadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Reza Mirzaei
- Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Alireza Khoshdel
- Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Pistachio Safety Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Zahra Hajializadeh
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Ali Fahmidehkar
- Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Laboratory Science, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mehdi Mahmoodi
- Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Clinical Biochemistry, Afzalipoor Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
46
|
Lohman EB, Pacheco GR, Gharibvand L, Daher N, Devore K, Bains G, AlAmeri M, Berk LS. The immediate effects of cervical spine manipulation on pain and biochemical markers in females with acute non-specific mechanical neck pain: a randomized clinical trial. J Man Manip Ther 2018; 27:186-196. [PMID: 30935335 PMCID: PMC7025692 DOI: 10.1080/10669817.2018.1553696] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Study Design: Randomized clinical trial with pre-test, post-test control group design. Objectives: To examine the immediate effects of cervical spinal manipulation (CSM) on serum concentration of biochemical markers (oxytocin, neurotensin, orexin A, and cortisol). Background: Several studies have found an association between spinal manipulation (SM) and pain perception. However, the mechanism by which SM modulates pain remains undefined. Methods: Twenty-eight female subjects with non-specific mechanical neck pain were randomly assigned to one of two interventions (CSM versus sham CSM). Blood samples were drawn before and immediately after the respective interventions. Oxytocin, neurotensin, orexin A, and cortisol were measured from the blood and serum using the Milliplex Map Magnetic Bead Panel Immunoassay on the Luminex 200 Platform. Results: In the CSM group, there were significant increases in pre- versus post-manipulation mean oxytocin (154.5 ± 60.1 vs. 185.1 ± 75.6, p = .012); neurotensin (116.0 ± 26.5 vs.136.4 ± 34.1, p < . 001); orexin A (52.2 ± 31.1 vs. 73.8 ± 38.8, p < .01) serum concentration; but no significant differences in mean cortisol (p = .052) serum concentration. In the sham group, there were no significant differences in any of the biomarkers (p > .05). Conclusion: The results of the current study suggest that the mechanical stimuli provided through a CSM may modify neuropeptide expression by immediately increasing the serum concentration of nociception-related biomarkers (oxytocin, neurotensin, orexin A, but not cortisol) in the blood of female subjects with non-specific mechanical neck pain.
Collapse
Affiliation(s)
- E B Lohman
- a School of Allied Health Professions, Physical Therapy Department, Loma Linda University , Loma Linda , CA , USA
| | - G R Pacheco
- a School of Allied Health Professions, Physical Therapy Department, Loma Linda University , Loma Linda , CA , USA
| | - L Gharibvand
- b School of Allied Health Professions, Allied Health Studies Department, Loma Linda University , Loma Linda , CA , USA
| | - N Daher
- b School of Allied Health Professions, Allied Health Studies Department, Loma Linda University , Loma Linda , CA , USA
| | - K Devore
- b School of Allied Health Professions, Allied Health Studies Department, Loma Linda University , Loma Linda , CA , USA
| | - G Bains
- b School of Allied Health Professions, Allied Health Studies Department, Loma Linda University , Loma Linda , CA , USA
| | - M AlAmeri
- a School of Allied Health Professions, Physical Therapy Department, Loma Linda University , Loma Linda , CA , USA
| | - L S Berk
- b School of Allied Health Professions, Allied Health Studies Department, Loma Linda University , Loma Linda , CA , USA.,c School of Medicine, Department of Pathology and Human Anatomy, Loma Linda University , Loma Linda , CA , USA
| |
Collapse
|
47
|
Orexinergic Modulation of Spinal Motor Activity in the Neonatal Mouse Spinal Cord. eNeuro 2018; 5:eN-NWR-0226-18. [PMID: 30417080 PMCID: PMC6223113 DOI: 10.1523/eneuro.0226-18.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/15/2018] [Accepted: 10/20/2018] [Indexed: 11/21/2022] Open
Abstract
The role of orexin during development, and especially in terms of spinal cord function, is not well understood. It is for this reason that we focused on the network actions of orexin during the first week of development. We found that orexinergic fibers were present in the lumbar spinal cord of postnatal day 0 (P0) to P3 mice. The fibers were expressed mainly in the dorsal horn, but occasional fibers were observed in the ventral horn. Both orexin (OX) A and OXB increased the motoneurons (MNs) tonic neurogram discharge. However, only OXA was found to significantly increase spontaneous bursting activity and the frequency of fictive locomotor bursts. We show that OXA is able to act directly on MNs. To test the contribution of the recurrent MN collaterals, we blocked the nicotinic cholinergic drive and observed that OXA retained its ability to increase fictive locomotor activity. Additionally, we recorded neurograms from ventral lateral funiculi, where OXA had no effect on population discharge. These effects were also confirmed by recording from descending commissural interneurons via patch recordings. The loci of the effects of OXA were further investigated in a dorsal horn-removed preparation where OXA also shows an increase in the discharge from ventral root neurograms but no increase in the frequency of spontaneous or fictive locomotion burst activity. In summary, multiple lines of evidence from our work demonstrate the robust effects of orexins on spinal cord networks and MNs at the time of birth.
Collapse
|
48
|
Raoof M, Soofiabadi S, Abbasnejad M, Kooshki R, Esmaeili-Mahani S, Mansoori M. Activation of orexin-1 receptors in the ventrolateral periaqueductal grey matter (vlPAG) modulates pulpal nociception and the induction of substance P in vlPAG and trigeminal nucleus caudalis. Int Endod J 2018; 52:318-328. [PMID: 30152877 DOI: 10.1111/iej.13007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/24/2018] [Indexed: 12/24/2022]
Abstract
AIM To characterize the role of orexin-1 receptors (OX1Rs) in ventrolateral periaqueductal grey matter (vlPAG) on modulation of capsaicin-induced pulpal nociception in rats. METHODOLOGY Sixty-six adult male Wistar rats (2 months old) weighing between 230 and 260 g were used. The animals were cannulated for microinjection of drugs into the vlPAG matter. Pulpalgia was induced by intradental application of capsaicin solution (100 μg) into the incisor teeth of the rats. Ten min prior to capsaicin application, orexin-A (50, 100 and 150 pmol L-1 per rat) was administered. Orexin-A (150 pmol L-1 ) was also co-administrated with SB-334867 (40 nmol L-1 per rat), an OX1Rs antagonist; or bicuculline (1 μg per rat), a GABAA receptors antagonist. Moreover, treatment effects on the release of pro-nociceptive modulator substance P (SP) in vlPAG and trigeminal nucleus caudalis (Vc) of rats were explored using an immunofluorescence technique. One-way analysis of variance was used for the statistical analysis. RESULTS Orexin-A dose-dependently decreased capsaicin-induced nociceptive behaviour. However, SB-334867 (40 nmol L-1 per rat) pretreatment (P < 0.05), but not bicuculline (1 μg per rat), attenuated the analgesic effect of orexin-A (150 pmol L-1 ). The level of SP was significantly increased in Vc and decreased in vlPAG of capsaicin-treated rats (P < 0.05). Capsaicin-induced changes in SP levels, however, were prohibited by orexin-A treatment (150 pmol L-1 ) (P < 0.05). CONCLUSIONS Orexin-A administration into the vlPAG was associated with an inhibitory effect on capsaicin-induced pulpal nociception and bidirectional effects on the induction of SP in vlPAG and Vc of rats. Central activation of OX1Rs is a potential therapeutic tool for pulpalgia.
Collapse
Affiliation(s)
- M Raoof
- Endodontology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - S Soofiabadi
- Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - M Abbasnejad
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - R Kooshki
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - S Esmaeili-Mahani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - M Mansoori
- Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
49
|
Median nerve stimulation induces analgesia via orexin-initiated endocannabinoid disinhibition in the periaqueductal gray. Proc Natl Acad Sci U S A 2018; 115:E10720-E10729. [PMID: 30348772 DOI: 10.1073/pnas.1807991115] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Adequate pain management remains an unmet medical need. We previously revealed an opioid-independent analgesic mechanism mediated by orexin 1 receptor (OX1R)-initiated 2-arachidonoylglycerol (2-AG) signaling in the ventrolateral periaqueductal gray (vlPAG). Here, we found that low-frequency median nerve stimulation (MNS) through acupuncture needles at the PC6 (Neiguan) acupoint (MNS-PC6) induced an antinociceptive effect that engaged this mechanism. In mice, MNS-PC6 reduced acute thermal nociceptive responses and neuropathy-induced mechanical allodynia, increased the number of c-Fos-immunoreactive hypothalamic orexin neurons, and led to higher orexin A and lower GABA levels in the vlPAG. Such responses were not seen in mice with PC6 needle insertion only or electrical stimulation of the lateral deltoid, a nonmedian nerve-innervated location. Directly stimulating the surgically exposed median nerve also increased vlPAG orexin A levels. MNS-PC6-induced antinociception (MNS-PC6-IA) was prevented by proximal block of the median nerve with lidocaine as well as by systemic or intravlPAG injection of an antagonist of OX1Rs or cannabinoid 1 receptors (CB1Rs) but not by opioid receptor antagonists. Systemic blockade of OX1Rs or CB1Rs also restored vlPAG GABA levels after MNS-PC6. A cannabinoid (2-AG)-dependent mechanism was also implicated by the observations that MNS-PC6-IA was prevented by intravlPAG inhibition of 2-AG synthesis and was attenuated in Cnr1 -/- mice. These findings suggest that PC6-targeting low-frequency MNS activates hypothalamic orexin neurons, releasing orexins to induce analgesia through a CB1R-dependent cascade mediated by OX1R-initiated 2-AG retrograde disinhibition in the vlPAG. The opioid-independent characteristic of MNS-PC6-induced analgesia may provide a strategy for pain management in opioid-tolerant patients.
Collapse
|
50
|
Abstract
The neuropeptides orexins are important in regulating the neurobiological systems that respond to stressful stimuli. Furthermore, orexins are known to play a role many of the phenotypes associated with stress-related mental illness such as changes in cognition, sleep-wake states, and appetite. Interestingly, orexins are altered in stress-related psychiatric disorders such as Major Depressive Disorder and Anxiety Disorders. Thus, orexins may be a potential target for treatment of these disorders. In this review, we will focus on what is known about the role of orexins in acute and repeated stress, in stress-induced phenotypes relevant to psychiatric illness in preclinical models, and in stress-related psychiatric illness in humans. We will also briefly discuss how orexins may contribute to sex differences in the stress response and subsequent phenotypes relevant to mental health, as many stress-related psychiatric disorders are twice as prevalent in women.
Collapse
|