51
|
Role of orexin-2 receptors in the nucleus accumbens in antinociception induced by carbachol stimulation of the lateral hypothalamus in formalin test. Behav Pharmacol 2017; 27:431-8. [PMID: 26871404 DOI: 10.1097/fbp.0000000000000216] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Orexins, which are mainly produced by orexin-expressing neurons in the lateral hypothalamus (LH), play an important role in pain modulation. Previously, it has been established that the nucleus accumbens (NAc) is involved in the modulation of formalin-induced nociceptive responses, a model of tonic pain. In this study, the role of intra-accumbal orexin-2 receptors (OX2rs) in the mediation of formalin-induced pain was investigated. A volume of 0.5 μl of 10, 20, and 40 nmol/l solutions of TCS OX2 29, an OX2r antagonist, were unilaterally microinjected into the NAc 5 min before an intra-LH carbachol microinjection (0.5 μl of 250 nmol/l solution). After 5 min, animals received a subcutaneous injection of formalin 2.5% (50 μl) into the hind paw. Pain-related behaviors were assessed at 5 min intervals during a 60-min test period. The findings showed that TCS OX2 29 administration dose dependently blocked carbachol-induced antinociception during both phases of formalin-induced pain. The antianalgesic effect of TCS OX2 29 was greater during the late phase compared with the early phase. These observations suggest that the NAc, as a part of a descending pain-modulatory circuitry, partially mediates LH-induced analgesia in the formalin test through recruitment of OX2rs. This makes the orexinergic system a good potential therapeutic target in the control of persistent inflammatory pain.
Collapse
|
52
|
Shaw JK, Ferris MJ, Locke JL, Brodnik ZD, Jones SR, España RA. Hypocretin/orexin knock-out mice display disrupted behavioral and dopamine responses to cocaine. Addict Biol 2017; 22:1695-1705. [PMID: 27480648 DOI: 10.1111/adb.12432] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 05/18/2016] [Accepted: 06/26/2016] [Indexed: 02/03/2023]
Abstract
The hypocretin/orexin (HCRT) system is implicated in reward and reinforcement processes through actions on the mesolimbic dopamine (DA) system. Here we provide evidence for the relationship between HCRT and DA in vivo in anesthetized and freely moving mice. The ability of cocaine to elicit reward-related behaviors in mice lacking the HCRT prepro-peptide (HCRT knock-out; KO) and wild-type controls was determined using conditioned place preference. Using a combination of microdialysis and in vivo fast scan cyclic voltammetry in anesthetized and freely moving mice, we investigated the underlying role of HCRT in the regulation of DA release and uptake. We show that, unlike wild-type mice, HCRT KO mice fail to develop characteristic conditioned place preference for cocaine. These mice also demonstrated reduced DA release and uptake under baseline conditions in both anesthetized and freely moving experiments. Further, diminished DA signaling in HCRT KO mice persists following administration of cocaine. These findings indicate that HCRT is essential for the expression of behaviors associated with the rewarding effects of cocaine, and suggest that HCRT regulation of reward and reinforcement may be related to disruptions to DA neurotransmission.
Collapse
Affiliation(s)
- Jessica K. Shaw
- Department of Neurobiology and Anatomy; Drexel University College of Medicine; Philadelphia PA USA
| | - Mark J. Ferris
- Department of Physiology and Pharmacology; Wake Forest School of Medicine; Winston-Salem NC USA
| | - Jason L. Locke
- Department of Physiology and Pharmacology; Wake Forest School of Medicine; Winston-Salem NC USA
| | - Zachary D. Brodnik
- Department of Neurobiology and Anatomy; Drexel University College of Medicine; Philadelphia PA USA
| | - Sara R. Jones
- Department of Physiology and Pharmacology; Wake Forest School of Medicine; Winston-Salem NC USA
| | - Rodrigo A. España
- Department of Neurobiology and Anatomy; Drexel University College of Medicine; Philadelphia PA USA
| |
Collapse
|
53
|
The link between narcolepsy and autonomic cardiovascular dysfunction: a translational perspective. Clin Auton Res 2017; 28:545-555. [DOI: 10.1007/s10286-017-0473-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/25/2017] [Indexed: 01/09/2023]
|
54
|
Futamura A, Nozawa D, Araki Y, Tamura Y, Tokura S, Kawamoto H, Tokumaru Y, Kakihara S, Aoki T, Ohtake N. Identification of highly selective and potent orexin receptor 1 antagonists derived from a dual orexin receptor 1/2 antagonist based on the structural framework of pyrazoylethylbenzamide. Bioorg Med Chem 2017; 25:5203-5215. [DOI: 10.1016/j.bmc.2017.07.051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/15/2017] [Accepted: 07/18/2017] [Indexed: 11/28/2022]
|
55
|
Levy KA, Brodnik ZD, Shaw JK, Perrey DA, Zhang Y, España RA. Hypocretin receptor 1 blockade produces bimodal modulation of cocaine-associated mesolimbic dopamine signaling. Psychopharmacology (Berl) 2017; 234:2761-2776. [PMID: 28667509 PMCID: PMC5709206 DOI: 10.1007/s00213-017-4673-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/06/2017] [Indexed: 02/07/2023]
Abstract
RATIONALE Cocaine addiction is a chronic psychiatric disorder characterized by pathological motivation to obtain cocaine and behavioral and neurochemical hypersensitivity to cocaine-associated cues. These features of cocaine addiction are thought to be driven by aberrant phasic dopamine signaling. We previously demonstrated that blockade of the hypocretin receptor 1 (HCRTr1) attenuates cocaine self-administration and reduces cocaine-induced enhancement of dopamine signaling. Despite this evidence, the effects of HCRTr1 blockade on endogenous phasic dopamine release are unknown. OBJECTIVE In the current studies, we assessed whether blockade of HCRTr1 alters spontaneous and cue-evoked dopamine release in the nucleus accumbens core of freely moving rats. METHODS We first validated the behavioral and neurochemical effects of the novel, highly selective, HCRTr1 antagonist RTIOX-276 using cocaine self-administration and fast-scan cyclic voltammetry (FSCV) in anesthetized rats. We then used FSCV in freely moving rats to examine whether RTIOX-276 impacts spontaneous and cue-evoked dopamine release. Finally, we used ex vivo slice FSCV to determine whether the effects of RTIOX-276 on dopamine signaling involve dopamine terminal adaptations. RESULTS Doses of RTIOX-276 that attenuate the motivation for cocaine reduce spontaneous dopamine transient amplitude and cue-evoked dopamine release. Further, these doses attenuated cocaine-induced dopamine uptake inhibition at the level of dopamine terminals. CONCLUSION Our results provide support for the standing hypothesis that HCRTr1 blockade suppresses endogenous phasic dopamine signals, likely via actions at dopamine cell bodies. These results also elucidate a second process through which HCRTr1 blockade attenuates the effects of cocaine by reducing cocaine sensitivity at dopamine terminals.
Collapse
Affiliation(s)
- KA Levy
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| | - ZD Brodnik
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| | - JK Shaw
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| | - DA Perrey
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, U.S.A
| | - Y Zhang
- Research Triangle Institute, Research Triangle Park, North Carolina 27709, U.S.A
| | - RA España
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, U.S.A
| |
Collapse
|
56
|
Abstract
The discovery of the orexin system represents the single major progress in the sleep field of the last three to four decades. The two orexin peptides and their two receptors play a major role in arousal and sleep/wake cycles. Defects in the orexin system lead to narcolepsy with cataplexy in humans and dogs and can be experimentally reproduced in rodents. At least six orexin receptor antagonists have reached Phase II or Phase III clinical trials in insomnia, five of which are dual orexin receptor antagonists (DORAs) that target both OX1 and OX2 receptors (OX2Rs). All clinically tested DORAs induce and maintain sleep: suvorexant, recently registered in the USA and Japan for insomnia, represents the first hypnotic principle that acts in a completely different manner from the current standard medications. It is clear, however, that in the clinic, all DORAs promote sleep primarily by increasing rapid eye movement (REM) and are almost devoid of effects on slow-wave (SWS) sleep. At present, there is no consensus on whether the sole promotion of REM sleep has a negative impact in patients suffering from insomnia. However, sleep onset REM (SOREM), which has been documented with DORAs, is clearly an undesirable effect, especially for narcoleptic patients and also in fragile populations (e.g. elderly patients) where REM-associated loss of muscle tone may promote an elevated risk of falls. Debate thus remains as to the ideal orexin agent to achieve a balanced increase in REM and non-rapid eye movement (NREM) sleep. Here, we review the evidence that an OX2R antagonist should be at least equivalent, or perhaps superior, to a DORA for the treatment of insomnia. An OX2R antagonist may produce more balanced sleep than a DORA. Rodent sleep experiments show that the OX2R is the primary target of orexin receptor antagonists in sleep modulation. Furthermore, an OX2R antagonist should, in theory, have a lower narcoleptic/cataplexic potential. In the clinic, the situation remains equivocal, since OX2R antagonists are in early stages: MK-1064 has completed Phase I, and MIN202 is currently in clinical Phase II/III trials. However, data from insomnia patients have not yet been released. Promotional material suggests that balanced sleep is indeed induced by MIN-202, whereas in volunteers MK-1064 has been reported to act similarly to DORAs.
Collapse
Affiliation(s)
- Laura H Jacobson
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Sui Chen
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Sanjida Mir
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Daniel Hoyer
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia.
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia.
- Department of Chemical Physiology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
57
|
Structure and Function of Peptide-Binding G Protein-Coupled Receptors. J Mol Biol 2017; 429:2726-2745. [PMID: 28705763 DOI: 10.1016/j.jmb.2017.06.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors and are important human drug targets. Of the 826 human GPCRs, 118 of them recognize endogenous peptide or protein ligands, and 30 of the 118 are targeted by approved drug molecules, including the very high-profile class B glucagon-like peptide 1 receptor. In this review, we analyze the 21 experimentally determined three-dimensional structures of the known peptide-binding GPCRs in relation to the endogenous peptides and drug molecules that modulate their cell signaling processes. Our integrated analyses reveal that half of the marketed drugs and most of the drugs in clinical trials that interact with peptide GPCRs are small molecules with a wide range of binding modes distinct from those of large peptide ligands. As we continue to collect additional data on these receptors from orthogonal approaches, including nuclear magnetic resonance and electron microscopy, we are beginning to understand how these receptors interact with their ligands at the molecular level and how improving the pharmacology of GPCR signal transduction requires us to study these receptors using multiple biophysical techniques.
Collapse
|
58
|
Affiliation(s)
- Christoph Boss
- Drug Discovery and Preclinical Research, Actelion Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Catherine Roch
- Drug Discovery and Preclinical Research, Actelion Pharmaceuticals Ltd, Allschwil, Switzerland
| |
Collapse
|
59
|
Connor KM, Ceesay P, Hutzelmann J, Snavely D, Krystal AD, Trivedi MH, Thase M, Lines C, Herring WJ, Michelson D. Phase II Proof-of-Concept Trial of the Orexin Receptor Antagonist Filorexant (MK-6096) in Patients with Major Depressive Disorder. Int J Neuropsychopharmacol 2017; 20:613-618. [PMID: 28582570 PMCID: PMC5570043 DOI: 10.1093/ijnp/pyx033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 06/01/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We evaluated the orexin receptor antagonist filorexant (MK-6096) for treatment augmentation in patients with major depressive disorder. METHODS We conducted a 6-week, double-blind, placebo-controlled, parallel-group, Phase II, proof-of-concept study. Patients with major depressive disorder (partial responders to ongoing antidepressant therapy) were randomized 1:1 to once-daily oral filorexant 10 mg or matching placebo. RESULTS Due to enrollment challenges, the study was terminated early, resulting in insufficient statistical power to detect a prespecified treatment difference; of 326 patients planned, 129 (40%) were randomized and 128 took treatment. There was no statistically significant difference in the primary endpoint of change from baseline to week 6 in Montgomery Asberg Depression Rating Scale total score; the estimated treatment difference for filorexant-placebo was -0.7 (with negative values favoring filorexant) (P=.679). The most common adverse events were somnolence and suicidal ideation. CONCLUSIONS The interpretation of the results is limited by the enrollment, which was less than originally planned, but the available data do not suggest efficacy of orexin receptor antagonism with filorexant for the treatment of depression. (Clinical Trial Registry: clinicaltrials.gov: NCT01554176).
Collapse
Affiliation(s)
- Kathryn M Connor
- Merck & Co., Inc., Kenilworth, New Jersey (Dr. Connor, Dr. Ceesay, Ms. Hutzelmann, Mr. Snavely, Dr. Lines, Dr. Herring, and Dr. Michelson); Duke University Hospital, Durham, North Carolina (Dr. Krystal); University of Texas Southwestern Medical Center, Dallas, Texas (Dr. Trivedi); University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania (Dr. Thase)
| | - Paulette Ceesay
- Merck & Co., Inc., Kenilworth, New Jersey (Dr. Connor, Dr. Ceesay, Ms. Hutzelmann, Mr. Snavely, Dr. Lines, Dr. Herring, and Dr. Michelson); Duke University Hospital, Durham, North Carolina (Dr. Krystal); University of Texas Southwestern Medical Center, Dallas, Texas (Dr. Trivedi); University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania (Dr. Thase)
| | - Jill Hutzelmann
- Merck & Co., Inc., Kenilworth, New Jersey (Dr. Connor, Dr. Ceesay, Ms. Hutzelmann, Mr. Snavely, Dr. Lines, Dr. Herring, and Dr. Michelson); Duke University Hospital, Durham, North Carolina (Dr. Krystal); University of Texas Southwestern Medical Center, Dallas, Texas (Dr. Trivedi); University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania (Dr. Thase)
| | - Duane Snavely
- Merck & Co., Inc., Kenilworth, New Jersey (Dr. Connor, Dr. Ceesay, Ms. Hutzelmann, Mr. Snavely, Dr. Lines, Dr. Herring, and Dr. Michelson); Duke University Hospital, Durham, North Carolina (Dr. Krystal); University of Texas Southwestern Medical Center, Dallas, Texas (Dr. Trivedi); University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania (Dr. Thase)
| | - Andrew D Krystal
- Merck & Co., Inc., Kenilworth, New Jersey (Dr. Connor, Dr. Ceesay, Ms. Hutzelmann, Mr. Snavely, Dr. Lines, Dr. Herring, and Dr. Michelson); Duke University Hospital, Durham, North Carolina (Dr. Krystal); University of Texas Southwestern Medical Center, Dallas, Texas (Dr. Trivedi); University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania (Dr. Thase)
| | - Madhukar H Trivedi
- Merck & Co., Inc., Kenilworth, New Jersey (Dr. Connor, Dr. Ceesay, Ms. Hutzelmann, Mr. Snavely, Dr. Lines, Dr. Herring, and Dr. Michelson); Duke University Hospital, Durham, North Carolina (Dr. Krystal); University of Texas Southwestern Medical Center, Dallas, Texas (Dr. Trivedi); University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania (Dr. Thase)
| | - Michael Thase
- Merck & Co., Inc., Kenilworth, New Jersey (Dr. Connor, Dr. Ceesay, Ms. Hutzelmann, Mr. Snavely, Dr. Lines, Dr. Herring, and Dr. Michelson); Duke University Hospital, Durham, North Carolina (Dr. Krystal); University of Texas Southwestern Medical Center, Dallas, Texas (Dr. Trivedi); University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania (Dr. Thase)
| | - Christopher Lines
- Merck & Co., Inc., Kenilworth, New Jersey (Dr. Connor, Dr. Ceesay, Ms. Hutzelmann, Mr. Snavely, Dr. Lines, Dr. Herring, and Dr. Michelson); Duke University Hospital, Durham, North Carolina (Dr. Krystal); University of Texas Southwestern Medical Center, Dallas, Texas (Dr. Trivedi); University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania (Dr. Thase)
| | - W Joseph Herring
- Merck & Co., Inc., Kenilworth, New Jersey (Dr. Connor, Dr. Ceesay, Ms. Hutzelmann, Mr. Snavely, Dr. Lines, Dr. Herring, and Dr. Michelson); Duke University Hospital, Durham, North Carolina (Dr. Krystal); University of Texas Southwestern Medical Center, Dallas, Texas (Dr. Trivedi); University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania (Dr. Thase)
| | - David Michelson
- Merck & Co., Inc., Kenilworth, New Jersey (Dr. Connor, Dr. Ceesay, Ms. Hutzelmann, Mr. Snavely, Dr. Lines, Dr. Herring, and Dr. Michelson); Duke University Hospital, Durham, North Carolina (Dr. Krystal); University of Texas Southwestern Medical Center, Dallas, Texas (Dr. Trivedi); University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania (Dr. Thase)
| |
Collapse
|
60
|
Born S, Gauvin DV, Mukherjee S, Briscoe R. Preclinical assessment of the abuse potential of the orexin receptor antagonist, suvorexant. Regul Toxicol Pharmacol 2017; 86:181-192. [PMID: 28279667 DOI: 10.1016/j.yrtph.2017.03.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 12/22/2022]
Abstract
Suvorexant (Belsomra®) is a dual orexin receptor antagonist approved for the treatment of insomnia. Because of its pharmacology within the central nervous system, intended therapeutic indication, and first-in-class status, an assessment of suvorexant abuse liability potential was required prior to marketing approval. The nonclinical abuse liability potential studies for suvorexant included: 1) rat drug-dependence model to assess physical dependence following abrupt cessation; 2) rat drug-discrimination model to examine the potential similarity of the interoceptive or subjective effects of suvorexant to those elicited by zolpidem and morphine; 3) self-administration model to assess the relative reinforcing efficacy of suvorexant in rhesus monkeys conditioned to self-administer methohexital. No significant signs of spontaneous drug withdrawal or 'discontinuation syndrome' were observed in rats following abrupt discontinuation of suvorexant. Suvorexant did not elicit complete cross-generalization to either a zolpidem or morphine training/reference stimuli in rats, and suvorexant was devoid of behavioral evidence of positive reinforcing efficacy in monkeys. These nonclinical findings suggested that suvorexant will have low abuse potential in humans. In the final regulatory risk assessment, suvorexant was placed into Schedule IV, likely due to its first-in-class status, its sedative properties, and the outcome of the clinical abuse potential assessment.
Collapse
|
61
|
Skudlarek JW, DiMarco CN, Babaoglu K, Roecker AJ, Bruno JG, Pausch MA, O'Brien JA, Cabalu TD, Stevens J, Brunner J, Tannenbaum PL, Wuelfing WP, Garson SL, Fox SV, Savitz AT, Harrell CM, Gotter AL, Winrow CJ, Renger JJ, Kuduk SD, Coleman PJ. Investigation of orexin-2 selective receptor antagonists: Structural modifications resulting in dual orexin receptor antagonists. Bioorg Med Chem Lett 2017; 27:1364-1370. [DOI: 10.1016/j.bmcl.2017.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/03/2017] [Accepted: 02/05/2017] [Indexed: 01/07/2023]
|
62
|
Orexin 2 receptor regulation of the hypothalamic-pituitary-adrenal (HPA) response to acute and repeated stress. Neuroscience 2017; 348:313-323. [PMID: 28257896 DOI: 10.1016/j.neuroscience.2017.02.038] [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] [Received: 09/12/2016] [Revised: 02/16/2017] [Accepted: 02/17/2017] [Indexed: 12/11/2022]
Abstract
Orexins are hypothalamic neuropeptides that have a documented role in mediating the acute stress response. However, their role in habituation to repeated stress, and the role of orexin receptors (OX1R and OX2R) in the stress response, has yet to be defined. Orexin neuronal activation and levels in the cerebrospinal fluid (CSF) were found to be stimulated with acute restraint, but were significantly reduced by day five of repeated restraint. As certain disease states such as panic disorder are associated with increased central orexin levels and failure to habituate to repeated stress, the effect of activating orexin signaling via Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) on the hypothalamic-pituitary-adrenal (HPA) response was evaluated after repeated restraint. While vehicle-treated rats displayed habituation of Adrenocorticotropic Hormone (ACTH) from day 1 to day 5 of restraint, stimulating orexins did not further increase ACTH beyond vehicle levels for either acute or repeated restraint. We delineated the roles of orexin receptors in acute and repeated stress using a selective OX2R antagonist (MK-1064). Pretreatment with MK-1064 reduced day 1 ACTH levels, but did not allow further habituation on day 5 compared with vehicle-treated rats, indicating that endogenous OX2R activity plays a role in acute stress, but not in habituation to repeated stress. However, in restrained rats with further stimulated orexins by DREADDs, MK-1064 decreased ACTH levels on day 5. Collectively, these results indicate that the OX2R plays a role in acute stress, and can prevent habituation to repeated stress under conditions of high orexin release.
Collapse
|
63
|
Carrive P, Kuwaki T. Orexin and Central Modulation of Cardiovascular and Respiratory Function. Curr Top Behav Neurosci 2017; 33:157-196. [PMID: 27909989 DOI: 10.1007/7854_2016_46] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Orexin makes an important contribution to the regulation of cardiorespiratory function. When injected centrally under anesthesia, orexin increases blood pressure, heart rate, sympathetic nerve activity, and the amplitude and frequency of respiration. This is consistent with the location of orexin neurons in the hypothalamus and the distribution of orexin terminals at all levels of the central autonomic and respiratory network. These cardiorespiratory responses are components of arousal and are necessary to allow the expression of motivated behaviors. Thus, orexin contributes to the cardiorespiratory response to acute stressors, especially those of a psychogenic nature. Consequently, upregulation of orexin signaling, whether it is spontaneous or environmentally induced, can increase blood pressure and lead to hypertension, as is the case for the spontaneously hypertensive rat and the hypertensive BPH/2J Schlager mouse. Blockade of orexin receptors will reduce blood pressure in these animals, which could be a new pharmacological approach for the treatment of some forms of hypertension. Orexin can also magnify the respiratory reflex to hypercapnia in order to maintain respiratory homeostasis, and this may be in part why it is upregulated during obstructive sleep apnea. In this pathological condition, blockade of orexin receptors would make the apnea worse. To summarize, orexin is an important modulator of cardiorespiratory function. Acting on orexin signaling may help in the treatment of some cardiovascular and respiratory disorders.
Collapse
Affiliation(s)
- Pascal Carrive
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Tomoyuki Kuwaki
- Department of Physiology, Graduate School of Medical & Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
64
|
Stump CA, Cooke AJ, Bruno J, Cabalu TD, Gotter AL, Harell CM, Kuduk SD, McDonald TP, O’Brien J, Renger JJ, Williams PD, Winrow CJ, Coleman PJ. Discovery of highly potent and selective orexin 1 receptor antagonists (1-SORAs) suitable for in vivo interrogation of orexin 1 receptor pharmacology. Bioorg Med Chem Lett 2016; 26:5809-5814. [DOI: 10.1016/j.bmcl.2016.10.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/07/2016] [Accepted: 10/08/2016] [Indexed: 12/26/2022]
|
65
|
Kordi Jaz E, Moghimi A, Fereidoni M, Asadi S, Shamsizadeh A, Roohbakhsh A. SB-334867, an orexin receptor 1 antagonist, decreased seizure and anxiety in pentylenetetrazol-kindled rats. Fundam Clin Pharmacol 2016; 31:201-207. [DOI: 10.1111/fcp.12249] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 09/30/2016] [Accepted: 10/11/2016] [Indexed: 01/18/2023]
Affiliation(s)
- Elham Kordi Jaz
- Department of Biology; Faculty of Sciences; Ferdowsi University of Mashhad; Azadi Sq. Mashhad Iran
| | - Ali Moghimi
- Department of Biology; Faculty of Sciences; Ferdowsi University of Mashhad; Azadi Sq. Mashhad Iran
| | - Masoud Fereidoni
- Department of Biology; Faculty of Sciences; Ferdowsi University of Mashhad; Azadi Sq. Mashhad Iran
| | - Saeedeh Asadi
- Department of Biology; Faculty of Sciences; Ferdowsi University of Mashhad; Azadi Sq. Mashhad Iran
| | - Ali Shamsizadeh
- Physiology-Pharmacology Research Center and Department of Physiology and Pharmacology; School of Medicine; Rafsanjan University of Medical Sciences; Pistachio Co Street Rafsanjan Iran
| | - Ali Roohbakhsh
- Pharmaceutical Research Center; Mashhad University of Medical Sciences; Azadi Square Mashhad Iran
- School of Pharmacy; Mashhad University of Medical Sciences; Azadi Square Mashhad Iran
| |
Collapse
|
66
|
Elevated levels of alpha-synuclein blunt cellular signal transduction downstream of Gq protein-coupled receptors. Cell Signal 2016; 30:82-91. [PMID: 27871937 DOI: 10.1016/j.cellsig.2016.11.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 10/21/2016] [Accepted: 11/17/2016] [Indexed: 01/06/2023]
Abstract
Alpha-synuclein is central to Parkinson's disease pathogenesis and pathology, however its precise functions are still unclear. It has been shown to bind both PLCβ1 and MAPKs, but how this property influences the downstream signaling of Gq protein-coupled receptors has not been elucidated. Here we show that recombinant expression of alpha-synuclein in human neuroblastoma cells enhances cellular levels of PLCβ1 but blunts its signaling pathway, preventing the agonist-dependent rise of cytoplasmic Ca2+. In addition, overexpressing alpha-synuclein abolishes the activation of ERK1/2 upon agonist stimulation, indicating an upstream action in the signal transduction pathway. This data demonstrates that alpha-synuclein, when recombinantly expressed, interferes with the normal signaling of Gq-protein coupled receptors, which are then dysfunctional. Since many neurotransmitter systems utilize these receptor signaling pathways to mediate different abilities affected in Parkinson's disease, we argue this novel perspective might be helpful in designing treatment strategies for some of the non-motor symptoms in Parkinson's disease and synucleinopathies.
Collapse
|
67
|
Emam AH, Hajesfandiari N, Shahidi S, Komaki A, Ganji M, Sarihi A. Modulation of nociception by medial pre-optic area orexin a receptors and its relation with morphine in male rats. Brain Res Bull 2016; 127:141-147. [PMID: 27641968 DOI: 10.1016/j.brainresbull.2016.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 09/13/2016] [Accepted: 09/14/2016] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Recent studies have shown that medial pre-optic area (MPOA) of hypothalamus are involved in nociception. Orexin A (hypocretin 1) has been found to have numerous applications including pain modulation. However, the role of orexin A receptors in the MPOA on the nociception has not been yet studied. Therefore, the aim of the present study is to investigate the effect of orexin A microinjection on MPOA on the nociception transmission and morphine induced analgesia in adult male rats. METHODS Using stereotaxic surgery, a cannula was implanted at a site 1mm above the MPOA in the anesthetized rats. After the recovery period, tail-flick (TF) latency was measured as 0, 15, 30, 45 and 60min following the onset of two experimental protocols. Two experiments were carried out. Experiment 1: The male rats received intra-MPOA of 25, 100, 1000, 10000pmol/0.5μl orexin A or 0.5μl of aCSF (control, just 5min before the TF assay. Experiment 2: The aim of this experiment was to examine the effect of orexin microinjection into MPOA on morphine analgesia (3mg/kg,s.c). Morphine was administered 30min before orexin A intra-MPOA microinjection (four doses similar to experiment 1) or aCSF, then TF latency was measured. RESULTS The results indicated that microinjection of orexin A into the MPOA showed anti-nociceptive effect in a time-dependent manner. Dose response curve results also revealed that the maximum effective dose of orexin A injection into MPOA for pain inhibition is 1000pmol/0.5μl. Co-administration of systemic morphine and orexin into the MPOA has additive analgesia with different time course compared morphine or orexin alone. CONCLUSION It can be concluded that MPOA OrexinA receptors play an important role in the modulation of pain in normal and morphine treated male rats.
Collapse
Affiliation(s)
- Amir Hossein Emam
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Naeimeh Hajesfandiari
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maziar Ganji
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abdolrahman Sarihi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
68
|
Messina A, De Fusco C, Monda V, Esposito M, Moscatelli F, Valenzano A, Carotenuto M, Viggiano E, Chieffi S, De Luca V, Cibelli G, Monda M, Messina G. Role of the Orexin System on the Hypothalamus-Pituitary-Thyroid Axis. Front Neural Circuits 2016; 10:66. [PMID: 27610076 PMCID: PMC4997012 DOI: 10.3389/fncir.2016.00066] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 08/08/2016] [Indexed: 12/25/2022] Open
Abstract
Hypocretin/orexin (ORX) are two hypothalamic neuropeptides discovered in 1998. Since their discovery, they have been one of the most studied neuropeptide systems because of their projecting fields innervating various brain areas. The orexinergic system is tied to sleep-wakefulness cycle, and narcolepsy is a consequence of their system hypofunction. Orexinergic system is also involved in many other autonomic functions such as feeding, thermoregulation, cardiovascular and neuroendocrine regulation. The main aim of this mini review article is to investigate the relationship between ORX and thyroid system regulation. Although knowledge about the ORX system is evolving, its putative effects on hypothalamic-pituitary-thyroid (HPT) axis still appear unclear. We analyzed some studies about ORX control of HPT axis to know better the relationship between them. The studies that were analyzed suggest Hypocretin/ORX to modulate the thyroid regulation, but the nature (excitatory or inhibitory) of this possible interaction remains actually unclear and needs to be confirmed.
Collapse
Affiliation(s)
- Antonietta Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Second University of Naples Naples, Italy
| | - Carolina De Fusco
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Second University of Naples Naples, Italy
| | - Vincenzo Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Second University of Naples Naples, Italy
| | - Maria Esposito
- Neapolitan Brain Group (NBG), Clinic of Child and Adolescent Neuropsychiatry, Department of Mental, Physical Health and Preventive Medicine, Second University of Naples Naples, Italy
| | - Fiorenzo Moscatelli
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Marco Carotenuto
- Neapolitan Brain Group (NBG), Clinic of Child and Adolescent Neuropsychiatry, Department of Mental, Physical Health and Preventive Medicine, Second University of Naples Naples, Italy
| | - Emanuela Viggiano
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Second University of Naples Naples, Italy
| | - Sergio Chieffi
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Second University of Naples Naples, Italy
| | - Vincenzo De Luca
- Department of Psychiatry, University of Toronto Toronto, ON, Canada
| | - Giuseppe Cibelli
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Second University of Naples Naples, Italy
| | - Giovanni Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetic and Sport Medicine, Second University of NaplesNaples, Italy; Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| |
Collapse
|
69
|
Martin EJ, Hernandez ME, Hayward LF. Blockade of orexin receptors attenuates the cardiovascular response to air-jet stress in spontaneously hypertensive rats. Auton Neurosci 2016; 201:8-16. [PMID: 27591948 DOI: 10.1016/j.autneu.2016.08.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/15/2016] [Accepted: 08/19/2016] [Indexed: 12/28/2022]
Abstract
This study tested the hypothesis that orexin plays a role in the elevated pressor response to acute stress in the spontaneously hypertensive rat (SHR). The pressor response to air jet stress (AJS) (n=11/group) was 2.5 times greater in vehicle treated SHR versus Wistar (WIS) rats. Systemic delivery of 30mg/kg of the dual orexin receptor antagonist almorexant did not significantly change resting mean arterial pressure (MAP) but did attenuate the pressor response elicited by AJS to a greater extent in the SHR compared to the Wistar rats (~65% versus ~33% reduction respectively; n=6/group). Alternatively 100mg/kg almorexant reduced resting MAP in the SHR (~25mm Hg drop) and attenuated both the heart rate (HR; ~50% reduction) and MAP (~62% reduction) response to AJS in both strains (n=6/group). Systemic application of SB-334867 (3mg/kg), an orexin receptor type 1 antagonist (n=5/group), selectively reduced resting MAP and attenuated the HR response to AJS in the SHR but had no effect on the pressor response in either strain. The potential role of endogenous orexin release in cardiovascular control in the SHR was linked to a significant increase in brain-derived neurotrophic factor mRNA expression in the hypothalamus and elevated orexin receptor expression (type 2 only) in the dorsal pons when compared to WIS (n=4/group). These results demonstrate that the exaggerated pressor response in the SHR to stress is linked to increased orexin receptor activation and possibly altered orexin receptor expression in the dorsal pons and BDNF expression in the hypothalamus.
Collapse
Affiliation(s)
- Eric J Martin
- University of Florida, College of Veterinary Medicine, Dept. of Physiological Sciences, Gainesville, FL 32610, United States
| | - Morgan E Hernandez
- University of Florida, College of Veterinary Medicine, Dept. of Physiological Sciences, Gainesville, FL 32610, United States
| | - Linda F Hayward
- University of Florida, College of Veterinary Medicine, Dept. of Physiological Sciences, Gainesville, FL 32610, United States.
| |
Collapse
|
70
|
Connor KM, Mahoney E, Jackson S, Hutzelmann J, Zhao X, Jia N, Snyder E, Snavely D, Michelson D, Roth T, Herring WJ. A Phase II Dose-Ranging Study Evaluating the Efficacy and Safety of the Orexin Receptor Antagonist Filorexant (MK-6096) in Patients with Primary Insomnia. Int J Neuropsychopharmacol 2016; 19:pyw022. [PMID: 26979830 PMCID: PMC5006195 DOI: 10.1093/ijnp/pyw022] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 03/02/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Filorexant (MK-6096) is an orexin receptor antagonist; here, we evaluate the efficacy of filorexant in the treatment of insomnia in adults. METHODS A double-blind, placebo-controlled, randomized, two 4-week-period, adaptive crossover polysomnography study was conducted at 51 sites worldwide. Patients (18 to <65 years) with insomnia received 1 of 4 doses of oral filorexant (2.5, 5, 10, 20mg) once daily at bedtime during one period and matching placebo in the other period in 1 of 8 possible treatment sequences. Polysomnography was performed on night 1 and end of week 4 of each period. The primary endpoint was sleep efficiency at night 1 and end of week 4. Secondary endpoints included wakefulness after persistent sleep onset and latency to onset of persistent sleep. RESULTS A total of 324 patients received study treatment, 315 received ≥1 dose of placebo, and 318 ≥1 dose of filorexant (2.5mg, n=79; 5mg, n=78; 10mg, n=80; 20mg, n=81). All filorexant doses (2.5/5/10/20mg) were significantly superior to placebo in improving sleep among patients with insomnia as measured by sleep efficiency and wakefulness after persistent sleep onset on night 1 and end of week 4. The 2 higher filorexant doses (10/20mg) were also significantly more effective than placebo in improving sleep onset as measured by latency to onset of persistent sleep at night 1 and end of week 4. Filorexant was generally well tolerated. CONCLUSIONS Orexin receptor antagonism by filorexant significantly improved sleep efficiency in nonelderly patients with insomnia. Dose-related improvements in sleep onset and maintenance outcomes were also observed with filorexant.
Collapse
Affiliation(s)
- Kathryn M Connor
- Merck & Co., Inc., Kenilworth, NJ (Dr Connor, Ms Mahoney, Ms Jackson, Ms Hutzelmann, Dr Zhao, Dr Jia, Dr Snyder, Mr Snavely, Dr Michelson, and Dr Herring); Henry Ford Hospital, Detroit, MI (Dr Roth).Current affiliations: Biogen, Cambridge, MA (E.M.). Janssen Pharmaceutical Research and Development, Raritan, NJ (X.Z.). Eli Lilly and Company, Indianapolis, IN (N.J.)
| | - Erin Mahoney
- Merck & Co., Inc., Kenilworth, NJ (Dr Connor, Ms Mahoney, Ms Jackson, Ms Hutzelmann, Dr Zhao, Dr Jia, Dr Snyder, Mr Snavely, Dr Michelson, and Dr Herring); Henry Ford Hospital, Detroit, MI (Dr Roth).Current affiliations: Biogen, Cambridge, MA (E.M.). Janssen Pharmaceutical Research and Development, Raritan, NJ (X.Z.). Eli Lilly and Company, Indianapolis, IN (N.J.)
| | - Saheeda Jackson
- Merck & Co., Inc., Kenilworth, NJ (Dr Connor, Ms Mahoney, Ms Jackson, Ms Hutzelmann, Dr Zhao, Dr Jia, Dr Snyder, Mr Snavely, Dr Michelson, and Dr Herring); Henry Ford Hospital, Detroit, MI (Dr Roth).Current affiliations: Biogen, Cambridge, MA (E.M.). Janssen Pharmaceutical Research and Development, Raritan, NJ (X.Z.). Eli Lilly and Company, Indianapolis, IN (N.J.)
| | - Jill Hutzelmann
- Merck & Co., Inc., Kenilworth, NJ (Dr Connor, Ms Mahoney, Ms Jackson, Ms Hutzelmann, Dr Zhao, Dr Jia, Dr Snyder, Mr Snavely, Dr Michelson, and Dr Herring); Henry Ford Hospital, Detroit, MI (Dr Roth).Current affiliations: Biogen, Cambridge, MA (E.M.). Janssen Pharmaceutical Research and Development, Raritan, NJ (X.Z.). Eli Lilly and Company, Indianapolis, IN (N.J.)
| | - Xin Zhao
- Merck & Co., Inc., Kenilworth, NJ (Dr Connor, Ms Mahoney, Ms Jackson, Ms Hutzelmann, Dr Zhao, Dr Jia, Dr Snyder, Mr Snavely, Dr Michelson, and Dr Herring); Henry Ford Hospital, Detroit, MI (Dr Roth).Current affiliations: Biogen, Cambridge, MA (E.M.). Janssen Pharmaceutical Research and Development, Raritan, NJ (X.Z.). Eli Lilly and Company, Indianapolis, IN (N.J.)
| | - Nan Jia
- Merck & Co., Inc., Kenilworth, NJ (Dr Connor, Ms Mahoney, Ms Jackson, Ms Hutzelmann, Dr Zhao, Dr Jia, Dr Snyder, Mr Snavely, Dr Michelson, and Dr Herring); Henry Ford Hospital, Detroit, MI (Dr Roth).Current affiliations: Biogen, Cambridge, MA (E.M.). Janssen Pharmaceutical Research and Development, Raritan, NJ (X.Z.). Eli Lilly and Company, Indianapolis, IN (N.J.)
| | - Ellen Snyder
- Merck & Co., Inc., Kenilworth, NJ (Dr Connor, Ms Mahoney, Ms Jackson, Ms Hutzelmann, Dr Zhao, Dr Jia, Dr Snyder, Mr Snavely, Dr Michelson, and Dr Herring); Henry Ford Hospital, Detroit, MI (Dr Roth).Current affiliations: Biogen, Cambridge, MA (E.M.). Janssen Pharmaceutical Research and Development, Raritan, NJ (X.Z.). Eli Lilly and Company, Indianapolis, IN (N.J.)
| | - Duane Snavely
- Merck & Co., Inc., Kenilworth, NJ (Dr Connor, Ms Mahoney, Ms Jackson, Ms Hutzelmann, Dr Zhao, Dr Jia, Dr Snyder, Mr Snavely, Dr Michelson, and Dr Herring); Henry Ford Hospital, Detroit, MI (Dr Roth).Current affiliations: Biogen, Cambridge, MA (E.M.). Janssen Pharmaceutical Research and Development, Raritan, NJ (X.Z.). Eli Lilly and Company, Indianapolis, IN (N.J.)
| | - David Michelson
- Merck & Co., Inc., Kenilworth, NJ (Dr Connor, Ms Mahoney, Ms Jackson, Ms Hutzelmann, Dr Zhao, Dr Jia, Dr Snyder, Mr Snavely, Dr Michelson, and Dr Herring); Henry Ford Hospital, Detroit, MI (Dr Roth).Current affiliations: Biogen, Cambridge, MA (E.M.). Janssen Pharmaceutical Research and Development, Raritan, NJ (X.Z.). Eli Lilly and Company, Indianapolis, IN (N.J.)
| | - Thomas Roth
- Merck & Co., Inc., Kenilworth, NJ (Dr Connor, Ms Mahoney, Ms Jackson, Ms Hutzelmann, Dr Zhao, Dr Jia, Dr Snyder, Mr Snavely, Dr Michelson, and Dr Herring); Henry Ford Hospital, Detroit, MI (Dr Roth).Current affiliations: Biogen, Cambridge, MA (E.M.). Janssen Pharmaceutical Research and Development, Raritan, NJ (X.Z.). Eli Lilly and Company, Indianapolis, IN (N.J.)
| | - W Joseph Herring
- Merck & Co., Inc., Kenilworth, NJ (Dr Connor, Ms Mahoney, Ms Jackson, Ms Hutzelmann, Dr Zhao, Dr Jia, Dr Snyder, Mr Snavely, Dr Michelson, and Dr Herring); Henry Ford Hospital, Detroit, MI (Dr Roth).Current affiliations: Biogen, Cambridge, MA (E.M.). Janssen Pharmaceutical Research and Development, Raritan, NJ (X.Z.). Eli Lilly and Company, Indianapolis, IN (N.J.)
| |
Collapse
|
71
|
Orexin, Stress and Central Cardiovascular Control. A Link with Hypertension? Neurosci Biobehav Rev 2016; 74:376-392. [PMID: 27477446 DOI: 10.1016/j.neubiorev.2016.06.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 05/28/2016] [Accepted: 06/13/2016] [Indexed: 01/28/2023]
Abstract
Orexin, the arousal peptide, originates from neurons located in an area of the dorsal hypothalamus well known for integrating defense responses and their cardiovascular component. Orexin neurons, which are driven in large part by the limbic forebrain, send projections to many regions in the brain, including regions involved in cardiovascular control, as far down as sympathetic preganglionic neurons in the spinal cord. Central injections of orexin evoke sympathetically mediated cardiovascular responses. Conversely, blockade of orexin receptors reduce the cardiovascular responses to acute stressors, preferentially of a psychological nature. More importantly, lasting upregulation of orexin signaling can lead to a hypertensive state. This can be observed in rats exposed to chronic stress as well as in strains known to display spontaneous hypertension such as the spontaneously hypertensive rat (SHR) or the hypertensive BPH/2J Schlager mouse. Thus, there is a link between orexin, stress and hypertension, and orexin upregulation could be a factor in the development of essential hypertension. Orexin receptor antagonists have anti-hypertensive effects that could be of clinical use.
Collapse
|
72
|
Tanaka Y, Aoki I, Ishine T, Renger JJ, Winrow CJ, Hisada S. [Preclinical and clinical results of dual orexin receptor antagonist, suvorexant (BELSOMRA(®)), a novel therapeutic agent for insomnia]. Nihon Yakurigaku Zasshi 2016; 148:46-56. [PMID: 27430679 DOI: 10.1254/fpj.148.46] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
|
73
|
McDonald T, Liang HA, Sanoja R, Gotter AL, Kuduk SD, Coleman PJ, Smith KM, Winrow CJ, Renger JJ. Pharmacological evaluation of orexin receptor antagonists in preclinical animal models of pain. J Neurogenet 2016; 30:32-41. [DOI: 10.3109/01677063.2016.1171862] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
74
|
Yardley DA. Pharmacologic management of bone-related complications and bone metastases in postmenopausal women with hormone receptor-positive breast cancer. BREAST CANCER-TARGETS AND THERAPY 2016; 8:73-82. [PMID: 27217795 PMCID: PMC4861000 DOI: 10.2147/bctt.s97963] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
There is a high risk for bone loss and skeletal-related events, including bone metastases, in postmenopausal women with hormone receptor-positive breast cancer. Both the disease itself and its therapeutic treatments can negatively impact bone, resulting in decreases in bone mineral density and increases in bone loss. These negative effects on the bone can significantly impact morbidity and mortality. Effective management and minimization of bone-related complications in postmenopausal women with hormone receptor-positive breast cancer remain essential. This review discusses the current understanding of molecular and biological mechanisms involved in bone turnover and metastases, increased risk for bone-related complications from breast cancer and breast cancer therapy, and current and emerging treatment strategies for managing bone metastases and bone turnover in postmenopausal women with hormone receptor-positive breast cancer.
Collapse
Affiliation(s)
- Denise A Yardley
- Sarah Cannon Research Institute, Nashville, TN, USA; Tennessee Oncology, Nashville, TN, USA
| |
Collapse
|
75
|
Kumar A, Chanana P, Choudhary S. Emerging role of orexin antagonists in insomnia therapeutics: An update on SORAs and DORAs. Pharmacol Rep 2016; 68:231-42. [DOI: 10.1016/j.pharep.2015.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 12/28/2022]
|
76
|
Trofimova I, Robbins TW. Temperament and arousal systems: A new synthesis of differential psychology and functional neurochemistry. Neurosci Biobehav Rev 2016; 64:382-402. [PMID: 26969100 DOI: 10.1016/j.neubiorev.2016.03.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 11/15/2015] [Accepted: 03/05/2016] [Indexed: 10/22/2022]
Abstract
This paper critically reviews the unidimensional construct of General Arousal as utilised by models of temperament in differential psychology for example, to underlie 'Extraversion'. Evidence suggests that specialization within monoamine neurotransmitter systems contrasts with the attribution of a "general arousal" of the Ascending Reticular Activating System. Experimental findings show specialized roles of noradrenaline, dopamine, and serotonin systems in hypothetically mediating three complementary forms of arousal that are similar to three functional blocks described in classical models of behaviour within kinesiology, clinical neuropsychology, psychophysiology and temperament research. In spite of functional diversity of monoamine receptors, we suggest that their functionality can be classified using three universal aspects of actions related to expansion, to selection-integration and to maintenance of chosen behavioural alternatives. Monoamine systems also differentially regulate analytic vs. routine aspects of activities at cortical and striatal neural levels. A convergence between main temperament models in terms of traits related to described functional aspects of behavioural arousal also supports the idea of differentiation between these aspects analysed here in a functional perspective.
Collapse
Affiliation(s)
- Irina Trofimova
- CILab, Department of Psychiatry and Behavioral Neurosciences, McMaster University, 92 Bowman St., Hamilton L8S2T6, Canada.
| | - Trevor W Robbins
- Department of Psychology and the Behavioural and Clinical Neuroscience Institute, Downing St., Cambridge CB23EB, UK.
| |
Collapse
|
77
|
Ezzatpanah S, Babapour V, Haghparast A. Differential contribution of orexin receptors within the ventral tegmental area to modulation of persistent inflammatory pain. Eur J Pain 2016; 20:1090-101. [DOI: 10.1002/ejp.833] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2015] [Indexed: 11/06/2022]
Affiliation(s)
- S. Ezzatpanah
- Department of Basic Sciences; Faculty of Veterinary Medicine; University of Tehran; Iran
| | - V. Babapour
- Department of Basic Sciences; Faculty of Veterinary Medicine; University of Tehran; Iran
| | - A. Haghparast
- Neuroscience Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
| |
Collapse
|
78
|
Lopez MF, Moorman DE, Aston-Jones G, Becker HC. The highly selective orexin/hypocretin 1 receptor antagonist GSK1059865 potently reduces ethanol drinking in ethanol dependent mice. Brain Res 2016; 1636:74-80. [PMID: 26851547 DOI: 10.1016/j.brainres.2016.01.049] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/20/2016] [Accepted: 01/27/2016] [Indexed: 01/01/2023]
Abstract
The orexin/hypocretin (ORX) system plays a major role in motivation for natural and drug rewards. In particular, a number of studies have shown that ORX signaling through the orexin 1 receptor (OX1R) regulates alcohol seeking and consumption. Despite the association between ORX signaling and motivation for alcohol, no study to date has investigated what role the ORX system plays in alcohol dependence, an understanding of which would have significant clinical relevance. This study was designed to evaluate the effect of the highly selective OX1R antagonist GSK1059865 on voluntary ethanol intake in ethanol-dependent and control non-dependent mice. Mice were subjected to a protocol in which they were evaluated for baseline ethanol intake and then exposed to intermittent ethanol or air exposure in inhalation chambers. Each cycle of chronic intermittent ethanol (CIE), or air, exposure was followed by a test of ethanol intake. Once the expected effect of increased voluntary ethanol intake was obtained in ethanol dependent mice, mice were tested for the effect of GSK1059865 on ethanol and sucrose intake. Treatment with GSK1059865 significantly decreased ethanol drinking in a dose-dependent manner in CIE-exposed mice. In contrast GSK1059865 decreased drinking in air-exposed mice only at the highest dose used. There was no effect of GSK1059865 on sucrose intake. Thus, ORX signaling through the OX1R, using a highly-selective antagonist, has a profound influence on high levels of alcohol drinking induced in a dependence paradigm, but limited or no influence on moderate alcohol drinking or sucrose drinking. These results indicate that the ORX system may be an important target system for treating disorders of compulsive reward seeking such as alcoholism and other addictions in which motivation is strongly elevated.
Collapse
Affiliation(s)
- Marcelo F Lopez
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, United States; Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, United States.
| | - David E Moorman
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, United States.
| | - Gary Aston-Jones
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Howard C Becker
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, United States; Charleston Alcohol Research Center, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC 29425, United States; Department of Veterans Affairs, Ralph H. Johnson Medical Center, Charleston, SC 29425, United States
| |
Collapse
|
79
|
Du MK, Hunt NJ, Waters KA, Machaalani R. Cumulative effects of repetitive intermittent hypercapnic hypoxia on orexin in the developing piglet hypothalamus. Int J Dev Neurosci 2015; 48:1-8. [PMID: 26548856 DOI: 10.1016/j.ijdevneu.2015.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 10/30/2015] [Accepted: 10/30/2015] [Indexed: 01/17/2023] Open
Abstract
Orexin neuropeptides (OxA and OxB) and their receptors (OX1R and OX2R) are involved in maintenance of sleep and wakefulness, and are regulated by various environmental stimuli. We studied piglets, in the early neonatal period, exposed to 48-min of intermittent hypercapnic hypoxia (IHH; 7% O2/8% CO2) alternating with air. Three groups of 13-14 day-old piglets with IHH exposure of 1-day (1D-IHH) (n=7), 2-days (2D-IHH) (n=7) and 4-days (4D-IHH) (n=8) were compared to controls (exposed only to air, n=8). Immunoreactivity of OxA and OxB was studied in the piglet hypothalamic regions of the dorsomedial hypothalamus (DMH), perifornical area (PeF) and lateral hypothalamic area (LH). Results showed that after 1D- and 2D-IHH, total OxA and OxB expression decreased by 20% (p ≤ 0.005) and 40% (p<0.001), respectively. After 4D-IHH, the decrease in OxA and OxB was 50% (p<0.001). These findings indicate that a chronic IHH exposure induces greater changes in orexin neuropeptide expression than an acute 1-day exposure in the hypothalamus. This may be causally related to the dysregulation of sleep.
Collapse
Affiliation(s)
- Man K Du
- Department of Pathology, University of Sydney, NSW 2006, Australia; The BOSCH Institute, University of Sydney, NSW 2006, Australia
| | - Nicholas J Hunt
- The BOSCH Institute, University of Sydney, NSW 2006, Australia; Department of Medicine, Blackburn Building, D06, University of Sydney, NSW 2006, Australia
| | - Karen A Waters
- Department of Medicine, Blackburn Building, D06, University of Sydney, NSW 2006, Australia; The Children's Hospital, Westmead Sydney, NSW 2145, Australia
| | - Rita Machaalani
- The BOSCH Institute, University of Sydney, NSW 2006, Australia; Department of Medicine, Blackburn Building, D06, University of Sydney, NSW 2006, Australia; The Children's Hospital, Westmead Sydney, NSW 2145, Australia.
| |
Collapse
|
80
|
Sun H, Yee KL, Gill S, Liu W, Li X, Panebianco D, Mangin E, Morrison D, McCrea J, Wagner JA, Troyer MD. Psychomotor effects, pharmacokinetics and safety of the orexin receptor antagonist suvorexant administered in combination with alcohol in healthy subjects. J Psychopharmacol 2015; 29:1159-69. [PMID: 26464455 DOI: 10.1177/0269881115609015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A double-blind crossover study investigated psychomotor effects, pharmacokinetics, and safety of the orexin receptor antagonist suvorexant with and without alcohol. Healthy adults (n=31) were randomized to receive placebo or suvorexant (40 mg) plus placebo solution or alcohol (0.7 g/kg) in each of four treatments (single doses; morning administration). The US Food and Drug Administration approved suvorexant dose is 10 mg (up to 20 mg) daily. Pharmacodynamic effects were assessed using tests of digit vigilance (DVT; primary endpoint), choice reaction time, digit symbol substitution, numeric working memory, immediate/delayed word recall, body sway and subjective alertness. Suvorexant alone did not significantly affect DVT reaction time, but did impact some pharmacodynamic tests. Suvorexant with alcohol increased reaction time versus either alone (mean difference at 2 h: 44 ms versus suvorexant, p<0.001; 24 ms, versus alcohol, p<0.05) and had additive negative effects on tests of vigilance, working/episodic memory, postural stability and alertness. No effects of suvorexant alone or with alcohol were observed by 9 h. No important changes in pharmacokinetic parameters were observed upon co-administration. All treatments were generally well tolerated without serious adverse events. In conclusion, co-administration of 40 mg suvorexant and 0.7 g/kg alcohol had additive negative psychomotor effects. Patients are advised not to consume alcohol with suvorexant.
Collapse
Affiliation(s)
- Hong Sun
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - Sean Gill
- Merck & Co., Inc., Kenilworth, NJ, USA
| | - Wen Liu
- Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Roecker AJ, Mercer SP, Bergman JM, Gilbert KF, Kuduk SD, Harrell CM, Garson SL, Fox SV, Gotter AL, Tannenbaum PL, Prueksaritanont T, Cabalu TD, Cui D, Lemaire W, Winrow CJ, Renger JJ, Coleman PJ. Discovery of diazepane amide DORAs and 2-SORAs enabled by exploration of isosteric quinazoline replacements. Bioorg Med Chem Lett 2015; 25:4992-4999. [DOI: 10.1016/j.bmcl.2014.12.081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 12/22/2014] [Accepted: 12/24/2014] [Indexed: 01/25/2023]
|
82
|
Nicole P, Couvineau P, Jamin N, Voisin T, Couvineau A. Crucial role of the orexin-B C-terminus in the induction of OX1 receptor-mediated apoptosis: analysis by alanine scanning, molecular modelling and site-directed mutagenesis. Br J Pharmacol 2015; 172:5211-23. [PMID: 26282891 PMCID: PMC4687804 DOI: 10.1111/bph.13287] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 06/12/2015] [Accepted: 08/11/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Orexins (A and B) are hypothalamic peptides that interact with OX1 and OX2 receptors and are involved in the sleep/wake cycle. We previously demonstrated that OX1 receptors are highly expressed in colon cancer tumours and colonic cancer cell lines where orexins induce apoptosis and inhibit tumour growth in preclinical animal models. The present study explored the structure-function relationships of orexin-B and OX1 receptors. EXPERIMENTAL APPROACH The contribution of all orexin-B residues in orexin-B-induced apoptosis was investigated by alanine scanning. To determine which OX1 receptor domains are involved in orexin-B binding and apoptosis, a 3D model of OX1 receptor docked to the orexin-B C-terminus (AA-20-28) was developed. Substitution of residues present in OX1 receptor transmembrane (TM) domains by site-directed mutagenesis was performed. KEY RESULTS Alanine substitution of orexin-B residues, L(11) , L(15) , A(22) , G(24) , I(25) , L(26) and M(28) , altered orexin-B's binding affinity. Substitution of these residues and of the Q(16) , A(17) , S(18) , N(20) and T(27) residues inhibited apoptosis in CHO-S-OX1 receptor cells. The K(120) , P(123) , Y(124) , N(318) , K(321) , F(340) , T(341) , H(344) and W(345) residues localized in TM2, TM3, TM6 and TM7 of OX1 receptors were shown to play a role in orexin-B recognition and orexin-B/OX1 receptor-induced apoptosis. CONCLUSIONS AND IMPLICATIONS The C-terminus of orexin-B (i) plays an important role in its pro-apoptotic effect; and (ii) interacts with some residues localized in the OX1 receptor TM. This study defines the structure-function relationship for orexin-B recognition by human OX1 receptors and orexin-B/OX1 receptor-induced apoptosis, an important step for the future development of new agonist molecules.
Collapse
Affiliation(s)
- Pascal Nicole
- Faculté de Médecine Site Bichat, INSERM U1149/Inflammation Research Center (CRI), Paris‐Diderot UniversityDHU UNITY16, rue H. Huchard75018ParisFrance
| | - Pierre Couvineau
- Faculté de Médecine Site Bichat, INSERM U1149/Inflammation Research Center (CRI), Paris‐Diderot UniversityDHU UNITY16, rue H. Huchard75018ParisFrance
| | - Nadege Jamin
- Laboratoire des Protéines et Systèmes MembranairesCEA, iBiTecS, I2BCF‐91191Gif‐sur‐Yvette CedexFrance
| | - Thierry Voisin
- Faculté de Médecine Site Bichat, INSERM U1149/Inflammation Research Center (CRI), Paris‐Diderot UniversityDHU UNITY16, rue H. Huchard75018ParisFrance
| | - Alain Couvineau
- Faculté de Médecine Site Bichat, INSERM U1149/Inflammation Research Center (CRI), Paris‐Diderot UniversityDHU UNITY16, rue H. Huchard75018ParisFrance
| |
Collapse
|
83
|
Wardach J, Wagner M, Jeong Y, Holden JE. Lateral Hypothalamic Stimulation Reduces Hyperalgesia Through Spinally Descending Orexin-A Neurons in Neuropathic Pain. West J Nurs Res 2015; 38:292-307. [PMID: 26475681 DOI: 10.1177/0193945915610083] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
No evidence to date shows that lateral hypothalamic (LH) stimulation produces orexin-A-mediated antinociception in the spinal cord dorsal horn (SCDH) in a model of neuropathic pain. We conducted experiments to examine the effect of orexin-A-mediated LH stimulation in female rats with chronic constriction injury (CCI) on thermal hyperalgesia. Rats receiving carbachol into the LH demonstrated antinociception on both the left CCI and right nonligated paws (p < .05). Rats were given carbachol in the LH followed by intrathecal injection of the orexin-1 (OX1) receptor antagonist SB-334867, which blocked LH-induced antinociception compared with control groups (p < .05) in the left paw, but not in the right paw. These findings support the hypothesis that LH stimulation produces antinociception in rats with thermal hyperalgesia from neuropathic pain via an orexin-A connection between the LH and the SCDH. Identification of this pathway may lead to studies using orexins to manage clinical pain.
Collapse
|
84
|
Roecker AJ, Cox CD, Coleman PJ. Orexin Receptor Antagonists: New Therapeutic Agents for the Treatment of Insomnia. J Med Chem 2015; 59:504-30. [PMID: 26317591 DOI: 10.1021/acs.jmedchem.5b00832] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Since its discovery in 1998, the orexin system, composed of two G-protein coupled receptors, orexins 1 and 2, and two neuropeptide agonists, orexins A and B, has captured the attention of the scientific community as a potential therapeutic target for the treatment of obesity, anxiety, and sleep/wake disorders. Genetic evidence in rodents, dogs, and humans was revealed between 1999 and 2000, demonstrating a causal link between dysfunction or deletion of the orexin system and narcolepsy, a disorder characterized by hypersomnolence during normal wakefulness. These findings encouraged efforts to discover agonists to treat narcolepsy and, alternatively, antagonists to treat insomnia. This perspective will focus on the discovery and development of structurally diverse orexin antagonists suitable for preclinical pharmacology studies and human clinical trials. The work described herein culminated in the 2014 FDA approval of suvorexant as a first-in-class dual orexin receptor antagonist for the treatment of insomnia.
Collapse
Affiliation(s)
- Anthony J Roecker
- Department of Medicinal Chemistry, Merck Research Laboratories , P.O. Box 4, 770 Sumneytown Pike, WP14-2, West Point, Pennsylvania 19486, United States
| | - Christopher D Cox
- Department of Medicinal Chemistry, Merck Research Laboratories , P.O. Box 4, 770 Sumneytown Pike, WP14-2, West Point, Pennsylvania 19486, United States
| | - Paul J Coleman
- Department of Medicinal Chemistry, Merck Research Laboratories , P.O. Box 4, 770 Sumneytown Pike, WP14-2, West Point, Pennsylvania 19486, United States
| |
Collapse
|
85
|
Beig MI, Horiuchi J, Dampney RAL, Carrive P. Both Ox1R and Ox2R orexin receptors contribute to the cardiorespiratory response evoked from the perifornical hypothalamus. Clin Exp Pharmacol Physiol 2015; 42:1059-67. [DOI: 10.1111/1440-1681.12461] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/23/2015] [Accepted: 07/09/2015] [Indexed: 11/30/2022]
Affiliation(s)
- Mirza I Beig
- School of Medical Sciences; University of New South Wales; Sydney NSW Australia
| | - Jouji Horiuchi
- Department of Biomedical Engineering; Toyo University; Kawagoe Saitama Japan
| | - Roger AL Dampney
- School of Medical Sciences; University of New South Wales; Sydney NSW Australia
- School of Medical Sciences and Bosch Institute for Biomedical Research; University of Sydney; Sydney NSW Australia
| | - Pascal Carrive
- School of Medical Sciences; University of New South Wales; Sydney NSW Australia
| |
Collapse
|
86
|
Johnson PL, Federici LM, Fitz SD, Renger JJ, Shireman B, Winrow CJ, Bonaventure P, Shekhar A. OREXIN 1 AND 2 RECEPTOR INVOLVEMENT IN CO2 -INDUCED PANIC-ASSOCIATED BEHAVIOR AND AUTONOMIC RESPONSES. Depress Anxiety 2015; 32:671-83. [PMID: 26332431 PMCID: PMC4729192 DOI: 10.1002/da.22403] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 07/07/2015] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The neuropeptides orexin A and B play a role in reward and feeding and are critical for arousal. However, it was not initially appreciated that most prepro-orexin synthesizing neurons are almost exclusively concentrated in the perifornical hypothalamus, which when stimulated elicits panic-associated behavior and cardiovascular responses in rodents and self-reported "panic attacks" and "fear of dying" in humans. More recent studies support a role for the orexin system in coordinating an integrative stress response. For instance, orexin neurons are highly reactive to anxiogenic stimuli, are hyperactive in anxiety pathology, and have strong projections to anxiety and panic-associated circuitry. Although the two cognate orexin receptors are colocalized in many brain regions, the orexin 2 receptor (OX2R) most robustly maps to the histaminergic wake-promoting region, while the orexin 1 receptor (OX1R) distribution is more exclusive and dense in anxiety and panic circuitry regions, such as the locus ceruleus. Overall, this suggests that OX1Rs play a critical role in mobilizing anxiety and panic responses. METHODS Here, we used a CO2 -panic provocation model to screen a dual OX1/2R antagonist (DORA-12) to globally inhibit orexin activity, then a highly selective OX1R antagonist (SORA1, Compound 56) or OX2R antagonist (SORA2, JnJ10397049) to assess OX1R and OX2R involvement. RESULTS All compounds except the SORA2 attenuated CO2 -induced anxiety-like behaviors, and all but the SORA2 and DORA attenuated CO2 -induced cardiovascular responses. CONCLUSIONS SORA1s may represent a novel method of treating anxiety disorders, with no apparent sedative effects that were present with a benzodiazepine.
Collapse
Affiliation(s)
- Philip L Johnson
- Departments of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana.,Departments of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana.,Departments of Medical Neuroscience Program, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Lauren M Federici
- Departments of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana.,Departments of Medical Neuroscience Program, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Stephanie D Fitz
- Departments of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Brock Shireman
- Janssen Research and Development LLC, San Diego, California
| | | | | | - Anantha Shekhar
- Departments of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana.,Departments of Medical Neuroscience Program, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana.,Indiana Clinical and Translational Sciences Institute, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
87
|
Mohammad Ahmadi Soleimani S, Azizi H, Mirnajafi-Zadeh J, Semnanian S. Orexin type 1 receptor antagonism in rat locus coeruleus prevents the analgesic effect of intra-LC met-enkephalin microinjection. Pharmacol Biochem Behav 2015. [DOI: 10.1016/j.pbb.2015.07.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
88
|
Hypothalamic dopaminergic neurons in an animal model of seasonal affective disorder. Neurosci Lett 2015; 602:17-21. [PMID: 26116821 DOI: 10.1016/j.neulet.2015.06.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 05/26/2015] [Accepted: 06/17/2015] [Indexed: 11/20/2022]
Abstract
Light has profound effects on mood regulation as exemplified in seasonal affective disorder (SAD) and the therapeutic benefits of light therapy. However, the underlying neural pathways through which light regulates mood are not well understood. Our previous work has developed the diurnal grass rat, Arvicanthis niloticus, as an animal model of SAD. Following housing conditions of either 12:12 h dim light:dark (DLD) or 8:16 h short photoperiod (SP), which mimic the lower light intensity or short day-length of winter, respectively, grass rats exhibit an increase in depression-like behavior compared to those housed in a 12:12 h bright light:dark (BLD) condition. Furthermore, we have shown that the orexinergic system is involved in mediating the effects of light on mood and anxiety. To explore other potential neural substrates involved in the depressive phenotype, the present study examined hypothalamic dopaminergic (DA) and somatostatin (SST) neurons in the brains of grass rats housed in DLD, SP and BLD. Using immunostaining for tyrosine hydroxylase (TH) and SST, we found that the number of TH- and SST-ir cells in the hypothalamus was significantly lower in the DLD and SP groups compared to the BLD group. We also found that treating BLD animals with a selective orexin receptor 1 (OX1R) antagonist SB-334867 significantly reduced the number of hypothalamic TH-ir cells. The present study suggests that the hypothalamic DA neurons are sensitive to daytime light deficiency and are regulated by an orexinergic pathway. The results support the hypothesis that the orexinergic pathways mediate the effects of light on other neuronal systems that collectively contribute to light-dependent changes in the affective state.
Collapse
|
89
|
Abstract
Suvorexant (Belsorma(®)) is the first orexin receptor antagonist approved by the US FDA (August 2014) for insomnia treatment. Following comprehensive Phase II/III studies, with up to 12 months of treatment in adult and elderly patients, there is little doubt that suvorexant induces and maintains sleep. However, the FDA and sponsor disagreed about effective versus safe doses (November 2012). The FDA considered that 5-15 mg were efficient and probably safe, whereas the sponsors had proposed 15-40 mg. The final approved doses are 5, 10, 15 and 20 mg. The major issues are next-morning somnolence and safety as seen in driving tests, with possible signs of muscle weakness, weird dreams, sleep walking, other nighttime behaviors and suicidal ideation. Despite its limitations, suvorexant's market entry offers a truly novel treatment for insomnia, paving the way for follow-up compounds and opening therapeutic avenues in other disorders for orexin receptor modulating compounds.
Collapse
Affiliation(s)
- Laura H Jacobson
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | | | | |
Collapse
|
90
|
Brodnik ZD, Bernstein DL, Prince CD, España RA. Hypocretin receptor 1 blockade preferentially reduces high effort responding for cocaine without promoting sleep. Behav Brain Res 2015; 291:377-384. [PMID: 26049058 DOI: 10.1016/j.bbr.2015.05.051] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 05/25/2015] [Accepted: 05/30/2015] [Indexed: 11/25/2022]
Abstract
Recent evidence suggests that blockade of the hypocretin receptor 1 may act as a useful pharmacotherapy for cocaine abuse. Here we investigated the extent to which various doses of a hypocretin receptor 1 antagonist, SB-334867, affect cocaine self-administration at varying doses of cocaine and across a range of effort requirements, and tested if these SB-334867 doses produce sedative effects. First, we trained animals to self-administer one of three doses of cocaine on a progressive ratio schedule, and then tested the effects of three doses of SB-334867. Responding for cocaine was then analyzed to segregate features of relatively high and low effort requirements across the progressive ratio session. In another set of experiments, we tested potential sleep-promoting effects of the same doses of SB-334867. Our data indicate that blockade of hypocretin receptor 1 preferentially reduces high effort responding for cocaine at levels that do not promote sedation.
Collapse
Affiliation(s)
- Zachary D Brodnik
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - David L Bernstein
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Courtney D Prince
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Rodrigo A España
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
91
|
Sifferlen T, Boller A, Chardonneau A, Cottreel E, Gatfield J, Treiber A, Roch C, Jenck F, Aissaoui H, Williams JT, Brotschi C, Heidmann B, Siegrist R, Boss C. Substituted pyrrolidin-2-ones: Centrally acting orexin receptor antagonists promoting sleep. Part 2. Bioorg Med Chem Lett 2015; 25:1884-91. [DOI: 10.1016/j.bmcl.2015.03.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 03/12/2015] [Accepted: 03/16/2015] [Indexed: 01/08/2023]
|
92
|
Identification of MK-8133: An orexin-2 selective receptor antagonist with favorable development properties. Bioorg Med Chem Lett 2015; 25:2488-92. [PMID: 25981685 DOI: 10.1016/j.bmcl.2015.04.066] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/17/2015] [Accepted: 04/20/2015] [Indexed: 12/31/2022]
Abstract
Antagonism of orexin receptors has shown clinical efficacy as a novel paradigm for the treatment of insomnia and related disorders. Herein, molecules related to the dual orexin receptor antagonist filorexant were transformed into compounds that were selective for the OX2R subtype. Judicious selection of the substituents on the pyridine ring and benzamide groups led to 6b; which was highly potent, OX2R selective, and exhibited excellent development properties.
Collapse
|
93
|
Doghramji PP. Integrating Modern Concepts of Insomnia and its Contemporary Treatment into Primary Care. Postgrad Med 2015; 126:82-101. [PMID: 25295652 DOI: 10.3810/pgm.2014.09.2802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
94
|
Song J, Kim E, Kim CH, Song HT, Lee JE. The role of orexin in post-stroke inflammation, cognitive decline, and depression. Mol Brain 2015; 8:16. [PMID: 25884812 PMCID: PMC4357085 DOI: 10.1186/s13041-015-0106-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 02/23/2015] [Indexed: 01/02/2023] Open
Abstract
Ischemic stroke results in diverse pathophysiologies, including cerebral inflammation, neuronal loss, cognitive dysfunction, and depression. Studies aimed at identifying therapeutic solutions to alleviate these outcomes are important due to the increase in the number of stroke patients annually. Recently, many studies have reported that orexin, commonly known as a neuropeptide regulator of sleep/wakefulness and appetite, is associated with neuronal cell apoptosis, memory function, and depressive symptoms. Here, we briefly summarize recent studies regarding the role and future perspectives of orexin in post-ischemic stroke. This review advances our understanding of the role of orexin in post-stroke pathologies, focusing on its possible function as a therapeutic regulator in the post-ischemic brain. Ultimately, we suggest the clinical potential of orexin to regulate post-stroke pathologies.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Yonsei University College of Medicine, Seoul, 120-752, South Korea.
| | - Eosu Kim
- Department of Pharmacology, Yonsei University College of Medicine, 120-752, Seoul, South Korea.
| | - Chul-Hoon Kim
- Department of Psychiatry, Yonsei University College of Medicine, 120-752, Seoul, South Korea.
| | - Ho-Taek Song
- Department of Diagnostic Radiology, Yonsei University College of Medicine, 120-752, Seoul, South Korea.
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, 120-752, South Korea. .,BK21 Plus Project for Medical Sciences, and Brain Research Institute, Yonsei University, College of Medicine, Seoul, 120-752, South Korea.
| |
Collapse
|
95
|
Discovery of piperidine ethers as selective orexin receptor antagonists (SORAs) inspired by filorexant. Bioorg Med Chem Lett 2015; 25:444-50. [DOI: 10.1016/j.bmcl.2014.12.056] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/15/2014] [Accepted: 12/16/2014] [Indexed: 01/28/2023]
|
96
|
Hoffmann J, Supronsinchai W, Akerman S, Andreou AP, Winrow CJ, Renger J, Hargreaves R, Goadsby PJ. Evidence for orexinergic mechanisms in migraine. Neurobiol Dis 2015; 74:137-43. [DOI: 10.1016/j.nbd.2014.10.022] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 10/08/2014] [Accepted: 10/29/2014] [Indexed: 11/29/2022] Open
|
97
|
Affiliation(s)
- Peter J Goadsby
- NIHR-Wellcome Trust King’s Clinical Research Facility, King’s College London, UK
- Department of Neurology, University of California, San Francisco, USA
| |
Collapse
|
98
|
Gotter AL, Garson SL, Stevens J, Munden RL, Fox SV, Tannenbaum PL, Yao L, Kuduk SD, McDonald T, Uslaner JM, Tye SJ, Coleman PJ, Winrow CJ, Renger JJ. Differential sleep-promoting effects of dual orexin receptor antagonists and GABAA receptor modulators. BMC Neurosci 2014; 15:109. [PMID: 25242351 PMCID: PMC4261741 DOI: 10.1186/1471-2202-15-109] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/17/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The current standard of care for insomnia includes gamma-aminobutyric acid receptor A (GABAA) activators, which promote sleep as well as general central nervous system depression. Dual orexin receptor antagonists (DORAs) represent an alternative mechanism for insomnia treatment that induces somnolence by blocking the wake-promoting effects of orexin neuropeptides. The current study compares the role and interdependence of these two mechanisms on their ability to influence sleep architecture and quantitative electroencephalography (qEEG) spectral profiles across preclinical species. RESULTS Active-phase dosing of DORA-22 induced consistent effects on sleep architecture in mice, rats, dogs, and rhesus monkeys; attenuation of active wake was accompanied by increases in both non-rapid eye movement (NREM) and rapid eye movement (REM) sleep. Eszopiclone, a representative GABAA receptor modulator, promoted sleep in rats and rhesus monkeys that was marked by REM sleep suppression, but had inconsistent effects in mice and paradoxically promoted wakefulness in dogs. Active-phase treatment of rats with DORA-12 similarly promoted NREM and REM sleep to magnitudes nearly identical to those seen during normal resting-phase sleep following vehicle treatment, whereas eszopiclone suppressed REM even to levels below those seen during the active phase. The qEEG changes induced by DORA-12 in rats also resembled normal resting-phase patterns, whereas eszopiclone induced changes distinct from normal active- or inactive-phase spectra. Co-dosing experiments, as well as studies in transgenic rats lacking orexin neurons, indicated partial overlap in the mechanism of sleep promotion by orexin and GABA modulation with the exception of the REM suppression exclusive to GABAA receptor modulation. Following REM deprivation in mice, eszopiclone further suppressed REM sleep while DORA-22 facilitated recovery including increased REM sleep. CONCLUSION DORAs promote NREM and importantly REM sleep that is similar in proportion and magnitude to that seen during the normal resting phase across mammalian animal models. While limited overlap exists between therapeutic mechanisms, orexin signaling does not appear involved in the REM suppression exhibited by GABAA receptor modulators. The ability of DORAs to promote proportional NREM and REM sleep following sleep deprivation suggests that this mechanism may be effective in alleviating recovery from sleep disturbance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - John J Renger
- Department of Neuroscience, Merck Research Laboratories, 770 Sumneytown Pike, PO Box 4, West Point, PA 19486-0004, USA.
| |
Collapse
|
99
|
Both Ox1r and Ox2r orexin receptors contribute to the cardiovascular and locomotor components of the novelty stress response in the rat. Neuropharmacology 2014; 89:146-56. [PMID: 25239810 DOI: 10.1016/j.neuropharm.2014.09.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 09/06/2014] [Accepted: 09/08/2014] [Indexed: 11/22/2022]
Abstract
Orexin contributes to the expression of the cardiovascular and behavioural response of some forms of stress, including novelty stress. Thus, Almorexant, a dual receptor antagonist that blocks the two known orexin receptors, Ox1R and Ox2R, reduces these responses. However, it is not known if the reduction results from blockade of one receptor only or both. To answer this question, the selective Ox1R antagonist ACT335827 and the selective Ox2R antagonist EMPA were injected intragastrically (300 mg/kg) or intraperitoneally (30 and 100 mg/kg) either alone or as a cocktail and compared to Almorexant in rats exposed to novelty stress. Cardiovascular and locomotor responses were recorded by radio-telemetry. Triple immunolabelling was also conducted to establish the distribution of Ox1R and Ox2R in sympathetic preganglionic neurons and orexin neurons. Intraperitoneal injections of ACT335827 (100 mg/kg) reduced the pressor and tachycardic but not the locomotor response of novelty (by 32% and 48%, respectively). Intraperitoneal injections of EMPA (100 mg/kg) only reduced the pressor response (42%). However when given together, ACT335827 and EMPA reduced all 3 components (65%, 60% and 57% of the tachycardic, pressor and locomotor responses, respectively) as Almorexant (100 mg/kg) did (69%, 87% and 72%, respectively). Triple immunolabelling revealed that sympathetic preganglionic neurons were mainly Ox1R positive only while orexin neurons were both Ox1R and Ox2R positive. This study shows that orexin's contribution to the cardiovascular and locomotor components of the novelty stress response is not mediated by one receptor alone, but by both receptors and at different levels of the neuraxis.
Collapse
|
100
|
Winrow CJ, Renger JJ. Discovery and development of orexin receptor antagonists as therapeutics for insomnia. Br J Pharmacol 2014; 171:283-93. [PMID: 23731216 DOI: 10.1111/bph.12261] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/13/2013] [Accepted: 05/16/2013] [Indexed: 01/23/2023] Open
Abstract
Insomnia persistently affects the quality and quantity of sleep. Currently approved treatments for insomnia primarily target γ-aminobutyric acid-A (GABA-A) receptor signalling and include benzodiazepines and GABA-A receptor modulators. These drugs are used to address this sleep disorder, but have the potential for side effects such as tolerance and dependence, making them less attractive as maintenance therapy. Forward and reverse genetic approaches in animals have implicated orexin signalling (also referred to as hypocretin signalling) in the control of vigilance and sleep/wake states. Screening for orexin receptor antagonists using in vitro and in vivo methods in animals has identified compounds that block one or other of the orexin receptors (single or dual orexin receptor antagonists [SORAs and DORAs], respectively) in animals and humans. SORAs have primarily been used as probes to further elucidate the roles of the individual orexin receptors, while a number of DORAs have progressed to clinical development as pharmaceutical candidates for insomnia. The DORA almorexant demonstrated significant improvements in a number of clinically relevant sleep parameters in animal models and in patients with insomnia but its development was halted. SB-649868 and suvorexant have demonstrated efficacy and tolerability in Phase II and III trials respectively. Furthermore, suvorexant is currently under review by the Food and Drug Administration for the treatment of insomnia. Based on the publication of recent non-clinical and clinical data, orexin receptor antagonists potentially represent a targeted, effective and well-tolerated new class of medications for insomnia.
Collapse
Affiliation(s)
- C J Winrow
- Department of Neuroscience, Merck Research Laboratories, West Point, PA, USA
| | | |
Collapse
|