1
|
Luo PX, Serna Godoy A, Zakharenkov HC, Vang N, Wright EC, Balantac TA, Archdeacon SC, Black AM, Lake AA, Ramirez AV, Lozier LE, Perez MD, Bhangal I, Desta NM, Trainor BC. Hypocretin in the nucleus accumbens shell modulates social approach in female but not male California mice. Neuropsychopharmacology 2024; 49:2000-2010. [PMID: 39117901 PMCID: PMC11480414 DOI: 10.1038/s41386-024-01937-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024]
Abstract
The hypocretin (Hcrt) system modulates arousal and anxiety-related behaviors and has been considered as a novel treatment target for stress-related affective disorders. We examined the effects of Hcrt acting in the nucleus accumbens shell (NAcSh) and anterodorsal bed nucleus of the stria terminalis (adBNST) on social behavior in male and female California mice (Peromyscus californicus). In female but not male California mice, infusion of Hcrt1 into NAcSh decreased social approach. Weak effects of Hcrt1 on social vigilance were observed in both females and males. No behavioral effects of Hcrt1 infused into the adBNST were observed. Analyses of sequencing data from California mice and Mus musculus NAc showed that Hcrtr2 was more abundant than Hcrtr1, so we infused the selective Hcrt receptor 2 antagonist into the NAcSh, which increased social approach in females previously exposed to social defeat. A calcium imaging study in the NAcSh of females before and after stress exposure showed that neural activity increased immediately following the expression of social avoidance but not during freezing behavior. This observation is consistent with previous studies that identified populations of neurons in the NAc that drive avoidance. Intriguingly, calcium transients were not affected by stress. These data suggest that hypocretin acting in the NAcSh plays a key role in modulating stress-induced social avoidance.
Collapse
Affiliation(s)
- Pei X Luo
- Department of Psychology, University of California, Davis, CA, USA
| | | | | | - Nou Vang
- Department of Psychology, University of California, Davis, CA, USA
| | - Emily C Wright
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | | | | | - Alexis M Black
- Department of Psychology, University of California, Davis, CA, USA
| | - Alyssa A Lake
- Department of Psychology, University of California, Davis, CA, USA
| | - Alison V Ramirez
- Department of Psychology, University of California, Davis, CA, USA
| | - Lauren E Lozier
- Department of Psychology, University of California, Davis, CA, USA
| | - Melvin D Perez
- Department of Psychology, University of California, Davis, CA, USA
| | - Irvin Bhangal
- Department of Psychology, University of California, Davis, CA, USA
| | - Nile M Desta
- Department of Psychology, University of California, Davis, CA, USA
| | - Brian C Trainor
- Department of Psychology, University of California, Davis, CA, USA.
| |
Collapse
|
2
|
Bjorness TE, Greene RW. Orexin-mediated motivated arousal and reward seeking. Peptides 2024; 180:171280. [PMID: 39159833 DOI: 10.1016/j.peptides.2024.171280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024]
Abstract
The neuromodulator orexin has been identified as a key factor for motivated arousal including recent evidence that sleep deprivation-induced enhancement of reward behavior is modulated by orexin. While orexin is not necessary for either reward or arousal behavior, orexin neurons' broad projections, ability to sense the internal state of the animal, and high plasticity of signaling in response to natural rewards and drugs of abuse may underlie heightened drug seeking, particularly in a subset of highly motivated reward seekers. As such, orexin receptor antagonists have gained deserved attention for putative use in addiction treatments. Ongoing and future clinical trials are expected to identify individuals most likely to benefit from orexin receptor antagonist treatment to promote abstinence, such as those with concurrent sleep disorders or high craving, while attention to methodological considerations will aid interpretation of the numerous preclinical studies investigating disparate aspects of the role of orexin in reward and arousal.
Collapse
Affiliation(s)
- Theresa E Bjorness
- Research Service, VA North Texas Health Care System, Dallas, TX 75126, USA; Departments of Psychiatry University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Robert W Greene
- Departments of Psychiatry University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba 305-8577, Japan
| |
Collapse
|
3
|
Atzil S, Satpute AB, Zhang J, Parrish MH, Shablack H, MacCormack JK, Leshin J, Goel S, Brooks JA, Kang J, Xu Y, Cohen M, Lindquist KA. The impact of sociality and affective valence on brain activation: A meta-analysis. Neuroimage 2023; 268:119879. [PMID: 36642154 DOI: 10.1016/j.neuroimage.2023.119879] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 01/07/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Thirty years of neuroimaging reveal the set of brain regions consistently associated with pleasant and unpleasant affect in humans-or the neural reference space for valence. Yet some of humans' most potent affective states occur in the context of other humans. Prior work has yet to differentiate how the neural reference space for valence varies as a product of the sociality of affective stimuli. To address this question, we meta-analyzed across 614 social and non-social affective neuroimaging contrasts, summarizing the brain regions that are consistently activated for social and non-social affective information. We demonstrate that across the literature, social and non-social affective stimuli yield overlapping activations within regions associated with visceromotor control, including the amygdala, hypothalamus, anterior cingulate cortex and insula. However, we find that social processing differs from non-social affective processing in that it involves additional cortical activations in the medial prefrontal and posterior cingulum that have been associated with mentalizing and prediction. A Bayesian classifier was able to differentiate unpleasant from pleasant affect, but not social from non-social affective states. Moreover, it was not able to classify unpleasantness from pleasantness at the highest levels of sociality. These findings suggest that highly social scenarios may be equally salient to humans, regardless of their valence.
Collapse
Affiliation(s)
- Shir Atzil
- The Hebrew University of Jerusalem, Jerusalem, Israel.
| | | | - Jiahe Zhang
- Northeastern University, Boston, MA, United States
| | | | - Holly Shablack
- Washington and Lee University, Lexington, VA, United States
| | | | - Joseph Leshin
- University of North Carolina, Chapel Hill, NC, United States
| | | | - Jeffrey A Brooks
- Hume AI, New York, NY, United States; University of California, Berkeley, CA, United States
| | - Jian Kang
- University of Michigan, Ann Arbor, MI, United States
| | - Yuliang Xu
- University of Michigan, Ann Arbor, MI, United States
| | - Matan Cohen
- The Hebrew University of Jerusalem, Jerusalem, Israel
| | | |
Collapse
|
4
|
Sharma A, Tripathi V, Kumar V. Hypothalamic molecular correlates of photoperiod-induced spring migration in intact and castrated male redheaded buntings. Mol Cell Endocrinol 2023; 561:111829. [PMID: 36526025 DOI: 10.1016/j.mce.2022.111829] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
This study investigated the molecular changes associated with neural plasticity in photoperiodic induction of spring migration in intact and castrated redheaded bunting, Emberiza bruniceps. We measured the hypothalamic mRNA expression of genes in birds that were photostimulated into winter non-migratory and spring (vernal) migratory phenotypes under short and long photoperiods, respectively. These included genes associated with the appetitive phase of reproduction (spring migration drive, th and ddc genes encoding for tyrosine hydroxylase and dopamine decarboxylase enzymes, respectively), sleep/awake state (pmch gene encoding for pro-melanin concentrating hormone; hcrt and hcrtr2 encoding for the hypocretin/orexin and its receptor, respectively) and neurogenesis (dcx and neuN coding for doublecortin and neuronal nuclear proteins, respectively). Higher th mRNA levels suggested an upregulated dopamine synthesis in the hypothalamus of spring migrants. Similarly, elevated hcrt and hcrtr2 mRNA levels suggested an increased wakefulness, and those of dcx and neuN genes suggested an enhanced neurogenesis during the spring migration state. Further, compared to intact birds, the lower th and pmch, and higher hcrtr2 and neuN mRNA levels in castrates suggested a role of testicular steroids in modulation of the appetitive phase of reproduction, sleep and awake states, and neurogenesis during spring migration period. These results provide insights into molecular changes linked with important hypothalamic molecular pathways and steroidal influence in the photoperiodic induction of spring migration in obligate migratory songbirds.
Collapse
Affiliation(s)
- Aakansha Sharma
- IndoUS Center in Chronobiology, Department of Zoology, University of Delhi, Delhi, 110007, India.
| | - Vatsala Tripathi
- Department of Zoology, Dyal Singh College, University of Delhi, Delhi, 110003, India.
| | - Vinod Kumar
- IndoUS Center in Chronobiology, Department of Zoology, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
5
|
Tyree SM, Jennings KJ, Gonzalez OC, Li SB, Nicholson JR, von Heimendahl M, de Lecea L. Optogenetic and pharmacological interventions link hypocretin neurons to impulsivity in mice. Commun Biol 2023; 6:74. [PMID: 36658362 PMCID: PMC9852239 DOI: 10.1038/s42003-023-04409-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 01/03/2023] [Indexed: 01/20/2023] Open
Abstract
Neurons in the lateral hypothalamus expressing the neuropeptide Hypocretin, also known as orexin, are known critical modulators of arousal stability. However, their role in the different components of the arousal construct such as attention and decision making is poorly understood. Here we study Hypocretin neuronal circuit dynamics during stop action impulsivity in a Go/NoGo task in mice. We show that Hypocretin neuronal activity correlates with anticipation of reward. We then assessed the causal role of Hypocretin neuronal activity using optogenetics in a Go/NoGo task. We show that stimulation of Hypocretin neurons during the cue period dramatically increases the number of premature responses. These effects are mimicked by amphetamine, reduced by atomoxetine, a norepinephrine uptake inhibitor, and blocked by a Hypocretin receptor 1 selective antagonist. We conclude that Hypocretin neurons have a key role in the integration of salient stimuli during wakefulness to produce appropriate and timely responses to rewarding and aversive cues.
Collapse
Affiliation(s)
- Susan M. Tyree
- grid.168010.e0000000419368956Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford, CA USA ,Present Address: Atlantia Clinical Trials, Cork, Ireland
| | - Kimberly J. Jennings
- grid.168010.e0000000419368956Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford, CA USA ,grid.55460.320000000121548364Present Address: University of Texas, Austin, TX USA
| | - Oscar C. Gonzalez
- grid.168010.e0000000419368956Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford, CA USA
| | - Shi-bin Li
- grid.168010.e0000000419368956Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford, CA USA
| | - Janet R. Nicholson
- grid.420061.10000 0001 2171 7500Boehringer Ingelheim Pharma GmbH & Co.KG, Biberach, Germany
| | - Moritz von Heimendahl
- grid.420061.10000 0001 2171 7500Boehringer Ingelheim Pharma GmbH & Co.KG, Biberach, Germany
| | - Luis de Lecea
- grid.168010.e0000000419368956Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford, CA USA
| |
Collapse
|
6
|
López JM, Carballeira P, Pozo J, León-Espinosa G, Muñoz A. Hypothalamic orexinergic neuron changes during the hibernation of the Syrian hamster. Front Neuroanat 2022; 16:993421. [PMID: 36157325 PMCID: PMC9501701 DOI: 10.3389/fnana.2022.993421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
Hibernation in small mammals is a highly regulated process with periods of torpor involving drops in body temperature and metabolic rate, as well as a general decrease in neural activity, all of which proceed alongside complex brain adaptive changes that appear to protect the brain from extreme hypoxia and low temperatures. All these changes are rapidly reversed, with no apparent brain damage occurring, during the short periods of arousal, interspersed during torpor—characterized by transitory and partial rewarming and activity, including sleep activation, and feeding in some species. The orexins are neuropeptides synthesized in hypothalamic neurons that project to multiple brain regions and are known to participate in the regulation of a variety of processes including feeding behavior, the sleep-wake cycle, and autonomic functions such as brown adipose tissue thermogenesis. Using multiple immunohistochemical techniques and quantitative analysis, we have characterized the orexinergic system in the brain of the Syrian hamster—a facultative hibernator. Our results revealed that orexinergic neurons in this species consisted of a neuronal population restricted to the lateral hypothalamic area, whereas orexinergic fibers distribute throughout the rostrocaudal extent of the brain, particularly innervating catecholaminergic and serotonergic neuronal populations. We characterized the changes of orexinergic cells in the different phases of hibernation based on the intensity of immunostaining for the neuronal activity marker C-Fos and orexin A (OXA). During torpor, we found an increase in C-Fos immunostaining intensity in orexinergic neurons, accompanied by a decrease in OXA immunostaining. These changes were accompanied by a volume reduction and a fragmentation of the Golgi apparatus (GA) as well as a decrease in the colocalization of OXA and the GA marker GM-130. Importantly, during arousal, C-Fos and OXA expression in orexinergic neurons was highest and the structural appearance and the volume of the GA along with the colocalization of OXA/GM-130 reverted to euthermic levels. We discuss the involvement of orexinergic cells in the regulation of mammalian hibernation and, in particular, the possibility that the high activation of orexinergic cells during the arousal stage guides the rewarming as well as the feeding and sleep behaviors characteristic of this phase.
Collapse
Affiliation(s)
- Jesús M. López
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
| | - Paula Carballeira
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
| | - Javier Pozo
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
| | - Gonzalo León-Espinosa
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-Centro de Estudios Universitarios (CEU), Madrid, Spain
| | - Alberto Muñoz
- Departamento de Biología Celular, Universidad Complutense, Madrid, Spain
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica (CTB), Universidad Politécnica de Madrid, Madrid, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- *Correspondence: Alberto Muñoz,
| |
Collapse
|
7
|
Eghtesad M, Elahdadi Salmani M, Lashkarbolouki T, Goudarzi I. Lateral Hypothalamus Corticotropin-releasing Hormone Receptor-1 Inhibition and Modulating Stress-induced Anxiety Behavior. Basic Clin Neurosci 2022; 13:373-384. [PMID: 36457881 PMCID: PMC9706292 DOI: 10.32598/bcn.2021.445.3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/15/2020] [Accepted: 10/12/2020] [Indexed: 10/28/2024] Open
Abstract
INTRODUCTION Stress is a reaction to unwanted events disturbing body homeostasis and its pathways and target areas. Stress affects the brain through the lateral hypothalamic area (LHA), the orexinergic system that mediates the effect of corticotropin-releasing hormone (CRH) through CRH Receptor Type 1 (CRHr1). Therefore, this study explores the outcome of stress exposure on anxiety development and the involvement of the LHA through LHA-CRHr1. METHODS Male Wistar rats (220-250 g) implanted with a cannula on either side of the LHA received acute or chronic stress. Subsequently, exploratory behavior was examined using the Open Field (OF), and anxiety was tested by Elevated Plus Maze (EPM). Before sacrifice, the cerebrospinal fluid (CSF) and the blood were sampled. Nissl stain was performed on fixed brain tissues. RESULTS Acute stress reduced exploration in of and increased anxiety in EPM. LHA-CRHr1 inhibition reversed the variables to increase the exploration and decrease anxiety. In contrast, chronic stress did not show any effect on anxiety-related behaviors. Chronic stress decreased the cell population in the LHA, which was prevented by the CRHr1 inhibition. However, the CRHr1 inhibition could not reverse the chronic stress-induced increase in the CSF orexin level. Furthermore, plasma corticosterone levels increased through acute or chronic stress, impeded by the inhibition of CRHr1. CONCLUSION Our results recognize LHA-CRHr1 as a capable candidate that modulates acute stress-induced anxiety development and chronic stress-induced changes in the cellular population of the region. HIGHLIGHTS Acute stress, increased immobility of the rat in open field and elevated plus maze.Chronic stress, increased orexin production while decreasing neuronal survival.The anxiety and immobility were not developed in presence of CRHr1.CRHr1 blocking reversed the chronic stress changes in corticosterone and orexin. PLAIN LANGUAGE SUMMARY Lateral Hypothalamus (LH) is a region involved in sleep and appetite regulation and recently known to play role in stress pathophysiology. The stress mediating function of the LH is performed through Corticotropin Releasing Hormone Receptor type-1 (CRHr1). This study explored the role of LH- CRHr1 in anxiety development and orexin production. Acute and chronic stress affected the behavior and molecular changes, differently. The acute stress increased the anxiety condition, while the chronic stress could only change the molecular criteria. Although we assumed that the inability of the chronic stress to develop anxiety may be attributable to habituation, the chronic stress could increase the plasma corticosterone and orexin level. All of the stress mal-changes in behavior and molecular level prevented by antagonising CRHr1 in the LH, indicating a gating function of LH-CRHr1 for stress development.
Collapse
Affiliation(s)
- Masoumeh Eghtesad
- Department of Animal Biology, School of Biology, Damghan University, Damghan, Iran
| | | | - Taghi Lashkarbolouki
- Department of Animal Biology, School of Biology, Damghan University, Damghan, Iran
| | - Iran Goudarzi
- Department of Animal Biology, School of Biology, Damghan University, Damghan, Iran
| |
Collapse
|
8
|
Dingemanse J, Charef P, Black J, Gouws C. Investigation of the effect of the dual orexin receptor antagonist almorexant on ophthalmological, spermatogenic, and hormonal variables in healthy male subjects. Biomed Pharmacother 2020; 133:110955. [PMID: 33190032 DOI: 10.1016/j.biopha.2020.110955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND/AIMS The aim of this single-center, double-blind study was to investigate the effect of a 4-week once daily administration of 200 mg almorexant on tear film break-up time, spermatogenesis, hormone levels, and pancreatic elastase in stool in healthy male subjects. METHODS Almorexant 200 mg or matching placebo was administered in the evening for 4 weeks once daily to 56 healthy male subjects. Changes in ophthalmological variables, sperm composition, hormone levels, and pancreatic elastase levels in stool were evaluated periodically up to 8 weeks after discontinuation of drug administration. Blood samples for pharmacokinetic measurements were taken after 4 weeks to confirm compliance to study drug intake. RESULTS The results of this study revealed no treatment effects of almorexant, neither on tear film break-up time nor on other ophthalmological variables investigated during this study. Furthermore, spermatogenesis, hormones of the hypothalamic-pituitary-adrenal and -gonadal axes, and endocrine pancreatic secretion were shown to be not affected by a 4-week once daily administration of almorexant. CONCLUSION Almorexant was well tolerated and had no effect on the spectrum of pharmacodynamic variables assessed. Ophthalmology and testicular findings detected in preclinical studies were not observed in this clinical study. Therefore, these preclinical findings appear not to be relevant for humans and do not prevent from conducting larger clinical trials with either healthy subjects or patients.
Collapse
Affiliation(s)
- Jasper Dingemanse
- Idorsia Pharmaceuticals Ltd, Clinical Pharmacology, Allschwil Switzerland.
| | - Pascal Charef
- Idorsia Pharmaceuticals Ltd, Clinical Science, Allschwil, Switzerland
| | - Jed Black
- Stanford Center for Sleep Science and Medicine, Palo Alto, California, United States; Neuropharma, Inc., Park City, Utah, United States
| | - Chris Gouws
- Pasteur Medical Centre, Bloemfontein, South Africa
| |
Collapse
|
9
|
Salehabadi S, Abrari K, Elahdadi Salmani M, Nasiri M, Lashkarbolouki T. Investigating the role of the amygdala orexin receptor 1 in memory acquisition and extinction in a rat model of PTSD. Behav Brain Res 2020; 384:112455. [DOI: 10.1016/j.bbr.2019.112455] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 11/15/2022]
|
10
|
Wilson MA, Liberzon I, Lindsey ML, Lokshina Y, Risbrough VB, Sah R, Wood SK, Williamson JB, Spinale FG. Common pathways and communication between the brain and heart: connecting post-traumatic stress disorder and heart failure. Stress 2019; 22:530-547. [PMID: 31161843 PMCID: PMC6690762 DOI: 10.1080/10253890.2019.1621283] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Psychiatric illnesses and cardiovascular disease (CVD) contribute to significant overall morbidity, mortality, and health care costs, and are predicted to reach epidemic proportions with the aging population. Within the Veterans Administration (VA) health care system, psychiatric illnesses such as post-traumatic stress disorder (PTSD) and CVD such as heart failure (HF), are leading causes of hospital admissions, prolonged hospital stays, and resource utilization. Numerous studies have demonstrated associations between PTSD symptoms and CVD endpoints, particularly in the Veteran population. Not only does PTSD increase the risk of HF, but this relationship is bi-directional. Accordingly, a VA-sponsored conference entitled "Cardiovascular Comorbidities in PTSD: The Brain-Heart Consortium" was convened to explore potential relationships and common biological pathways between PTSD and HF. The conference was framed around the hypothesis that specific common systems are dysregulated in both PTSD and HF, resulting in a synergistic acceleration and amplification of both disease processes. The conference was not intended to identify all independent pathways that give rise to PTSD and HF, but rather identify shared systems, pathways, and biological mediators that would be modifiable in both disease processes. The results from this conference identified specific endocrine, autonomic, immune, structural, genetic, and physiological changes that may contribute to shared PTSD-CVD pathophysiology and could represent unique opportunities to develop therapies for both PTSD and HF. Some recommendations from the group for future research opportunities are provided.
Collapse
Affiliation(s)
- Marlene A. Wilson
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine and Research Service, Columbia VA Health Care System, Columbia SC
- Corresponding author information: Marlene A. Wilson, Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia SC 29208, Research Service, Columbia VA Health Care System, Columbia SC 29209, ; 803-216-3507
| | - Israel Liberzon
- Department of Psychiatry, Texas A&M College of Medicine, Bryan, TX
| | - Merry L. Lindsey
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, and Research Service, Omaha VA Medical Center, Omaha NE
| | - Yana Lokshina
- Department of Psychiatry, Texas A&M College of Medicine, Bryan, TX
| | - Victoria B. Risbrough
- VA Center of Excellence for Stress and Mental Health, La Jolla CA, Dept. of Psychiatry, University of California San Diego
| | - Renu Sah
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Susan K. Wood
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine and Research Service, Columbia VA Health Care System, Columbia SC
| | - John B. Williamson
- Department of Neurology, University of Florida College of Medicine, Gainesville FL
| | - Francis G. Spinale
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine and Research Service, Columbia VA Health Care System., Columbia SC
| |
Collapse
|
11
|
Yakupov EZ, Artemieva DV, Troshina YV. [Multimodality of clinical effects of orexin]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 118:114-119. [PMID: 30698570 DOI: 10.17116/jnevro2018118121114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The article describes the main biological properties of orexin as a wakefulness hormone and its possible points of application in practical medicine. An example of a drug, which blocks orexin receptors and can be used as a hypnotic drug to treat sleep disorders, is presented.
Collapse
Affiliation(s)
- E Z Yakupov
- Kazan State Medical University, Kazan, Russia
| | | | | |
Collapse
|
12
|
Bahramzadeh Zoeram S, Elahdadi Salmani M, Lashkarbolouki T, Goudarzi I. Hippocampal orexin receptor blocking prevented the stress induced social learning and memory deficits. Neurobiol Learn Mem 2018; 157:12-23. [PMID: 30458283 DOI: 10.1016/j.nlm.2018.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 10/14/2018] [Accepted: 11/16/2018] [Indexed: 01/09/2023]
Abstract
Stress as a homeostatic challenge leads to the malfunction of learning and memory processes, namely social learning and memory. The orexin system is involved in stress responses through connections to the hypothalamic-pituitary axis (HPA). In addition, the hippocampus, a structure vulnerable to stress-induced changes, expresses orexin receptors 1 and 2 (OXr1 and OXr2) in various sub-regions. The present study is aimed at assessing the effects of hippocampal orexin receptor blockade on social learning and memory impairments and anxiety development following stress. Male Wistar rats (220-250 g) underwent cannula implantation in the hippocampus. Acute (two mild electric shocks, 5.5 mA) and chronic stresses (ten days of restraint, 6 h daily) were applied with or without injection of orexin receptor antagonists (SB-334867 or TCS OX 29). Sociability and social novelty in animals were assessed in a three-chamber social maze at the end of stress application. Anxiety and exploratory behavior of animals were then examined, with 20 min intervals, using the open field (OF) and elevated plus maze (EPM) tests, respectively. Cisterna Magna cerebro-spinal fluid (CSF) was drained, before sacrifice, for orexin (OX) assay and trunk blood was collected to measure the plasma corticosterone (CRT). Neither the acute nor the chronic stress could affect the sociability. The acute but not chronic stress prevented the animal from sniffing the familiar caged rat in the novelty session, a response which was reversed following the blockade of both OXRs. Furthermore, acute but not chronic stress, led to increased anxiety and immobility behavior which were both impeded by blocking the orexin receptor (OXR). Conversely, OX content in CSF increased due to chronic restraint stress, an effect that was reversed by orexin blockade. Finally, elevated plasma CRT was recorded in response to both acute and chronic stresses. The observed increase in plasma CRT in chronically-stressed rats was abolished following inhibition of OXRs, however a similar effect was not seen in the acute-stress group. Our results identify hippocampal OXRs as potential candidates capable of preventing acute stress-induced impairments of social novelty and anxiety behavior, and chronic stress-induced plasma CRT and CSF orexin, changes. OXR manipulation may improve adaptation to stress pathophysiology.
Collapse
Affiliation(s)
| | | | | | - Iran Goudarzi
- School of Biology, Damghan University, Damghan, Iran
| |
Collapse
|
13
|
Iyer M, Essner RA, Klingenberg B, Carter ME. Identification of discrete, intermingled hypocretin neuronal populations. J Comp Neurol 2018; 526:2937-2954. [PMID: 30019757 DOI: 10.1002/cne.24490] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 01/04/2023]
Abstract
Neurons in the lateral hypothalamic area that express hypocretin (Hcrt) neuropeptides help regulate many behaviors including wakefulness and reward seeking. These neurons project throughout the brain, including to neural populations that regulate wakefulness, such as the locus coeruleus (LC) and tuberomammilary nucleus (TMN), as well as to populations that regulate reward, such as the nucleus accumbens (NAc) and ventral tegmental area (VTA). To address the roles of Hcrt neurons in seemingly disparate behaviors, it has been proposed that Hcrt neurons can be anatomically subdivided into at least two distinct subpopulations: a "medial group" that projects to the LC and TMN, and a "lateral group" that projects to the NAc and VTA. Here, we use a dual retrograde tracer strategy to test the hypotheses that Hcrt neurons can be classified based on their downstream projections and medial/lateral location within the hypothalamus. We found that individual Hcrt neurons were significantly more likely to project to both the LC and TMN or to both the VTA and NAc than would be predicted by chance. In contrast, we found that Hcrt neurons that projected to the LC or TMN were mostly distinct from Hcrt neurons that projected to the VTA or NAc. Interestingly, these two populations of Hcrt neurons are intermingled within the hypothalamus and cannot be classified into medial or lateral groups. These results suggest that Hcrt neurons can be distinguished based on their downstream projections but are intermingled within the hypothalamus.
Collapse
Affiliation(s)
- Manasi Iyer
- Department of Biology, Williams College, Williamstown, Massachusetts.,Program in Neuroscience, Williams College, Williamstown, Massachusetts
| | - Rachel A Essner
- Department of Biology, Williams College, Williamstown, Massachusetts.,Program in Neuroscience, Williams College, Williamstown, Massachusetts
| | - Bernhard Klingenberg
- Department of Mathematics and Statistics, Williams College, Williamstown, Massachusetts
| | - Matthew E Carter
- Department of Biology, Williams College, Williamstown, Massachusetts.,Program in Neuroscience, Williams College, Williamstown, Massachusetts
| |
Collapse
|
14
|
da Silva EN, Horta-Júnior JDAC, Gargaglioni LH, Dias MB. ATP in the lateral hypothalamus/perifornical area enhances the CO 2 chemoreflex control of breathing. Exp Physiol 2018; 103:1679-1691. [PMID: 30242927 DOI: 10.1113/ep087182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/20/2018] [Indexed: 12/24/2022]
Abstract
NEW FINDINGS What is the central question of this study? ATP is known to modulate the chemosensitivity of some brain areas. However, whether the ATP contributes specifically to the mechanism of chemoreception in the lateral hypothalamus/perifornical area (LH/PFA) remains to be determined. What is the main finding and its importance? ATP, acting on the LH/PFA, enhances the hypercapnic ventilatory response in rats during wakefulness, in the dark period. Our results highlight the importance of ATP as a modulator of central chemoreception and provide new insight regarding the mechanisms involved in LH/PFA chemosensitivity and the sleep-wake differences in the CO2 /H+ -dependent drive to breathe. ABSTRACT The lateral hypothalamus/perifornical area (LH/PFA) is a central chemoreceptor site, which acts in an arousal state-dependent manner. It has been shown that purinergic signalling through ATP influences the CO2 /H+ responsiveness of other chemosensitive regions, but it is unknown whether ATP is also involved in the mechanisms that underlie LH/PFA chemoreception. Here, we studied the effects of microdialysis of a P2X-receptor agonist [α,β-methylene ATP (α,β-meATP), 10 mm] and a non-selective P2-receptor antagonist [pyridoxal-phosphate-6-azophenyl-2',4'-disulfonate (PPADS), 1 mm] into the LH/PFA of conscious rats on ventilation in room air and in 7% CO2 . In the dark (active) phase, but not in the light, microdialysis of α,β-meATP caused an augmented hypercapnic ventilatory response during wakefulness, but not during non-REM sleep (P < 0.001). PPADS caused no change in CO2 ventilatory responses in either the dark period or the light period. Our data suggest that ATP in LH/PFA contributes to the hypercapnic ventilatory response in conscious rats during wakefulness in the dark phase of the diurnal cycle.
Collapse
Affiliation(s)
- Eliandra N da Silva
- Department of Physiology, Institute of Bioscience, Sao Paulo State University-UNESP, Botucatu, SP, Brazil
| | | | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University-FCAV, Jaboticabal, SP, Brazil
| | - Mirela B Dias
- Department of Physiology, Institute of Bioscience, Sao Paulo State University-UNESP, Botucatu, SP, Brazil
| |
Collapse
|
15
|
Maggi S, Balzani E, Lassi G, Garcia-Garcia C, Plano A, Espinoza S, Mus L, Tinarelli F, Nolan PM, Gainetdinov RR, Balci F, Nieus T, Tucci V. The after-hours circadian mutant has reduced phenotypic plasticity in behaviors at multiple timescales and in sleep homeostasis. Sci Rep 2017; 7:17765. [PMID: 29259298 PMCID: PMC5736711 DOI: 10.1038/s41598-017-18130-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 12/06/2017] [Indexed: 12/01/2022] Open
Abstract
Circadian clock is known to adapt to environmental changes and can significantly influence cognitive and physiological functions. In this work, we report specific behavioral, cognitive, and sleep homeostatic defects in the after hours (Afh) circadian mouse mutant, which is characterized by lengthened circadian period. We found that the circadian timing irregularities in Afh mice resulted in higher interval timing uncertainty and suboptimal decisions due to incapability of processing probabilities. Our phenotypic observations further suggested that Afh mutants failed to exhibit the necessary phenotypic plasticity for adapting to temporal changes at multiple time scales (seconds-to-minutes to circadian). These behavioral effects of Afh mutation were complemented by the specific disruption of the Per/Cry circadian regulatory complex in brain regions that govern food anticipatory behaviors, sleep, and timing. We derive statistical predictions, which indicate that circadian clock and sleep are complementary processes in controlling behavioral/cognitive performance during 24 hrs. The results of this study have pivotal implications for understanding how the circadian clock modulates sleep and behavior.
Collapse
Affiliation(s)
- Silvia Maggi
- Department of Neuroscience and Brain Technologies - Genetics and Epigenetics of Behaviour - Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy.,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Edoardo Balzani
- Department of Neuroscience and Brain Technologies - Genetics and Epigenetics of Behaviour - Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Glenda Lassi
- Department of Neuroscience and Brain Technologies - Genetics and Epigenetics of Behaviour - Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Celina Garcia-Garcia
- Department of Neuroscience and Brain Technologies - Genetics and Epigenetics of Behaviour - Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Andrea Plano
- Department of Neuroscience and Brain Technologies - Genetics and Epigenetics of Behaviour - Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Stefano Espinoza
- Department of Neuroscience and Brain Technologies - Genetics and Epigenetics of Behaviour - Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Liudmila Mus
- Department of Neuroscience and Brain Technologies - Genetics and Epigenetics of Behaviour - Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Federico Tinarelli
- Department of Neuroscience and Brain Technologies - Genetics and Epigenetics of Behaviour - Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Patrick M Nolan
- MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, UK
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.,Skolkovo Institute of Science & Technology, Skolkovo, Moscow, Russia
| | - Fuat Balci
- Koç University, Department of Psychology, Istanbul, Turkey
| | - Thierry Nieus
- Department of Neuroscience and Brain Technologies - Genetics and Epigenetics of Behaviour - Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy
| | - Valter Tucci
- Department of Neuroscience and Brain Technologies - Genetics and Epigenetics of Behaviour - Istituto Italiano di Tecnologia, via Morego, 30, 16163, Genova, Italy.
| |
Collapse
|
16
|
Dingemanse J, Krause A. Impact of pharmacokinetic-pharmacodynamic modelling in early clinical drug development. Eur J Pharm Sci 2017; 109S:S53-S58. [PMID: 28535992 DOI: 10.1016/j.ejps.2017.05.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 12/27/2022]
Abstract
Early clinical pharmacology studies in healthy subjects are often dissociated from patient studies. In this review we encourage the use of modelling and simulation techniques to generate valuable information at an early stage of clinical development. We illustrate these principles by presenting 5 different case studies from a spectrum of therapeutic drug classes. Their application leads to a better understanding of drug characteristics early on, thereby facilitating the design of dose-finding studies in the target patient population and saving resources.
Collapse
Affiliation(s)
- Jasper Dingemanse
- Department of Clinical Pharmacology, Actelion Pharmaceuticals Ltd, Gewerbestrasse 16, 4123 Allschwil, Switzerland.
| | - Andreas Krause
- Department of Clinical Pharmacology, Actelion Pharmaceuticals Ltd, Gewerbestrasse 16, 4123 Allschwil, Switzerland
| |
Collapse
|
17
|
|
18
|
Sharko AC, Fadel JR, Kaigler KF, Wilson MA. Activation of orexin/hypocretin neurons is associated with individual differences in cued fear extinction. Physiol Behav 2017; 178:93-102. [PMID: 27746261 PMCID: PMC5391308 DOI: 10.1016/j.physbeh.2016.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/23/2016] [Accepted: 10/11/2016] [Indexed: 01/15/2023]
Abstract
Identifying the neurobiological mechanisms that underlie differential sensitivity to stress is critical for understanding the development and expression of stress-induced disorders, such as post-traumatic stress disorder (PTSD). Preclinical studies have suggested that rodents display different phenotypes associated with extinction of Pavlovian conditioned fear responses, with some rodent populations being resistant to extinction. An emerging literature also suggests a role for orexins in the consolidation processes associated with fear learning and extinction. To examine the possibility that the orexin system might be involved in individual differences in fear extinction, we used a Pavlovian conditioning paradigm in outbred Long-Evans rats. Rats showed significant variability in the extinction of cue-conditioned freezing and extinction recall, and animals were divided into groups based on their extinction profiles based on a median split of percent freezing behavior during repeated exposure to the conditioned cue. Animals resistant to extinction (high freezers) showed more freezing during repeated cue presentations during the within trial and between trial extinction sessions compared with the group showing significant extinction (low freezers), although there were no differences between these groups in freezing upon return to the conditioned context or during the conditioning session. Following the extinction recall session, activation of orexin neurons was determined using dual label immunohistochemistry for cFos in orexin positive neurons in the hypothalamus. Individual differences in the extinction of cue conditioned fear were associated with differential activation of hypothalamic orexin neurons. Animals showing poor extinction of cue-induced freezing (high freezers) had significantly greater percentage of orexin neurons with Fos in the medial hypothalamus than animals displaying significant extinction and good extinction recall (low freezers). Further, the freezing during extinction learning was positively correlated with the percentage of activated orexin neurons in both the lateral and medial hypothalamic regions. No differences in the overall density of orexin neurons or Fos activation were seen between extinction phenotypes. Although correlative, our results support other studies implicating a role of the orexinergic system in regulating extinction of conditioned responses to threat.
Collapse
Affiliation(s)
- Amanda C Sharko
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina, School of Medicine, Columbia, SC, USA; WJB Dorn Veterans Affairs Medical Center, Columbia, SC, USA
| | - Jim R Fadel
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina, School of Medicine, Columbia, SC, USA; WJB Dorn Veterans Affairs Medical Center, Columbia, SC, USA
| | - Kris F Kaigler
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina, School of Medicine, Columbia, SC, USA; WJB Dorn Veterans Affairs Medical Center, Columbia, SC, USA
| | - Marlene A Wilson
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina, School of Medicine, Columbia, SC, USA; WJB Dorn Veterans Affairs Medical Center, Columbia, SC, USA.
| |
Collapse
|
19
|
Treiber A, de Kanter R, Roch C, Gatfield J, Boss C, von Raumer M, Schindelholz B, Muehlan C, van Gerven J, Jenck F. The Use of Physiology-Based Pharmacokinetic and Pharmacodynamic Modeling in the Discovery of the Dual Orexin Receptor Antagonist ACT-541468. J Pharmacol Exp Ther 2017; 362:489-503. [DOI: 10.1124/jpet.117.241596] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/20/2017] [Indexed: 01/01/2023] Open
|
20
|
Eban-Rothschild A, de Lecea L. Neuronal substrates for initiation, maintenance, and structural organization of sleep/wake states. F1000Res 2017; 6:212. [PMID: 28357049 PMCID: PMC5345773 DOI: 10.12688/f1000research.9677.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2017] [Indexed: 11/20/2022] Open
Abstract
Animals continuously alternate between sleep and wake states throughout their life. The daily organization of sleep and wakefulness is orchestrated by circadian, homeostatic, and motivational processes. Over the last decades, much progress has been made toward determining the neuronal populations involved in sleep/wake regulation. Here, we will discuss how the application of advanced
in vivo tools for cell type–specific manipulations now permits the functional interrogation of different features of sleep/wake state regulation: initiation, maintenance, and structural organization. We will specifically focus on recent studies examining the roles of wake-promoting neuronal populations.
Collapse
Affiliation(s)
- Ada Eban-Rothschild
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
21
|
Hypocretin/Orexin Peptides Alter Spike Encoding by Serotonergic Dorsal Raphe Neurons through Two Distinct Mechanisms That Increase the Late Afterhyperpolarization. J Neurosci 2016; 36:10097-115. [PMID: 27683906 DOI: 10.1523/jneurosci.0635-16.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 08/11/2016] [Indexed: 01/02/2023] Open
Abstract
UNLABELLED Orexins (hypocretins) are neuropeptides that regulate multiple homeostatic processes, including reward and arousal, in part by exciting serotonergic dorsal raphe neurons, the major source of forebrain serotonin. Here, using mouse brain slices, we found that, instead of simply depolarizing these neurons, orexin-A altered the spike encoding process by increasing the postspike afterhyperpolarization (AHP) via two distinct mechanisms. This orexin-enhanced AHP (oeAHP) was mediated by both OX1 and OX2 receptors, required Ca(2+) influx, reversed near EK, and decayed with two components, the faster of which resulted from enhanced SK channel activation, whereas the slower component decayed like a slow AHP (sAHP), but was not blocked by UCL2077, an antagonist of sAHPs in some neurons. Intracellular phospholipase C inhibition (U73122) blocked the entire oeAHP, but neither component was sensitive to PKC inhibition or altered PKA signaling, unlike classical sAHPs. The enhanced SK current did not depend on IP3-mediated Ca(2+) release but resulted from A-current inhibition and the resultant spike broadening, which increased Ca(2+) influx and Ca(2+)-induced-Ca(2+) release, whereas the slower component was insensitive to these factors. Functionally, the oeAHP slowed and stabilized orexin-induced firing compared with firing produced by a virtual orexin conductance lacking the oeAHP. The oeAHP also reduced steady-state firing rate and firing fidelity in response to stimulation, without affecting the initial rate or fidelity. Collectively, these findings reveal a new orexin action in serotonergic raphe neurons and suggest that, when orexin is released during arousal and reward, it enhances the spike encoding of phasic over tonic inputs, such as those related to sensory, motor, and reward events. SIGNIFICANCE STATEMENT Orexin peptides are known to excite neurons via slow postsynaptic depolarizations. Here we elucidate a significant new orexin action that increases and prolongs the postspike afterhyperpolarization (AHP) in 5-HT dorsal raphe neurons and other arousal-system neurons. Our mechanistic studies establish involvement of two distinct Ca(2+)-dependent AHP currents dependent on phospholipase C signaling but independent of IP3 or PKC. Our functional studies establish that this action preserves responsiveness to phasic inputs while attenuating responsiveness to tonic inputs. Thus, our findings bring new insight into the actions of an important neuropeptide and indicate that, in addition to producing excitation, orexins can tune postsynaptic excitability to better encode the phasic sensory, motor, and reward signals expected during aroused states.
Collapse
|
22
|
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]
|
23
|
Graebner AK, Iyer M, Carter ME. Understanding how discrete populations of hypothalamic neurons orchestrate complicated behavioral states. Front Syst Neurosci 2015; 9:111. [PMID: 26300745 PMCID: PMC4523943 DOI: 10.3389/fnsys.2015.00111] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 07/16/2015] [Indexed: 01/01/2023] Open
Abstract
A major question in systems neuroscience is how a single population of neurons can interact with the rest of the brain to orchestrate complex behavioral states. The hypothalamus contains many such discrete neuronal populations that individually regulate arousal, feeding, and drinking. For example, hypothalamic neurons that express hypocretin (Hcrt) neuropeptides can sense homeostatic and metabolic factors affecting wakefulness and orchestrate organismal arousal. Neurons that express agouti-related protein (AgRP) can sense the metabolic needs of the body and orchestrate a state of hunger. The organum vasculosum of the lamina terminalis (OVLT) can detect the hypertonicity of blood and orchestrate a state of thirst. Each hypothalamic population is sufficient to generate complicated behavioral states through the combined efforts of distinct efferent projections. The principal challenge to understanding these brain systems is therefore to determine the individual roles of each downstream projection for each behavioral state. In recent years, the development and application of temporally precise, genetically encoded tools has greatly improved our understanding of the structure and function of these neural systems. This review will survey recent advances in our understanding of how these individual hypothalamic populations can orchestrate complicated behavioral states due to the combined efforts of individual downstream projections.
Collapse
Affiliation(s)
- Allison K Graebner
- Program in Neuroscience, Department of Biology, Williams College Williamstown, MA, USA
| | - Manasi Iyer
- Program in Neuroscience, Department of Biology, Williams College Williamstown, MA, USA
| | - Matthew E Carter
- Program in Neuroscience, Department of Biology, Williams College Williamstown, MA, USA
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Wang Z, Liu S, Kakizaki M, Hirose Y, Ishikawa Y, Funato H, Yanagisawa M, Yu Y, Liu Q. Orexin/hypocretin activates mTOR complex 1 (mTORC1) via an Erk/Akt-independent and calcium-stimulated lysosome v-ATPase pathway. J Biol Chem 2014; 289:31950-31959. [PMID: 25278019 PMCID: PMC4231673 DOI: 10.1074/jbc.m114.600015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/30/2014] [Indexed: 02/05/2023] Open
Abstract
The lack of the neuropeptide orexin, also known as hypocretin, results in narcolepsy, a chronic sleep disorder characterized by frequent sleep/cataplexy attacks and rapid eye movement sleep abnormalities. However, the downstream pathways of orexin signaling are not clearly understood. Here, we show that orexin activates the mTOR pathway, a central regulator of cell growth and metabolism, in the mouse brain and multiple recombinant cell lines that express the G protein-coupled receptors (GPCRs), orexin 1 receptor (OX1R) or orexin 2 receptor (OX2R). This orexin/GPCR-stimulated mTOR activation is sensitive to rapamycin, an inhibitor of mTOR complex 1 (mTORC1) but is independent of two well known mTORC1 activators, Erk and Akt. Rather, our studies indicate that orexin activates mTORC1 via extracellular calcium influx and the lysosome pathway involving v-ATPase and Rag GTPases. Moreover, a cytoplasmic calcium transient is sufficient to mimic orexin/GPCR signaling to mTORC1 activation in a v-ATPase-dependent manner. Together, our studies suggest that the mTORC1 pathway functions downstream of orexin/GPCR signaling, which plays a crucial role in many physiological and metabolic processes.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Shimeng Liu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390,; College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Miyo Kakizaki
- International Institute of Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan, and
| | - Yuuki Hirose
- International Institute of Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan, and
| | - Yukiko Ishikawa
- International Institute of Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan, and
| | - Hiromasa Funato
- International Institute of Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan, and; Department of Anatomy, Toho University School of Medicine, Tokyo 143-8540, Japan
| | - Masashi Yanagisawa
- International Institute of Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan, and
| | - Yonghao Yu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390,.
| | - Qinghua Liu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390,; International Institute of Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan, and.
| |
Collapse
|
27
|
Li J, Hu Z, de Lecea L. The hypocretins/orexins: integrators of multiple physiological functions. Br J Pharmacol 2014; 171:332-50. [PMID: 24102345 DOI: 10.1111/bph.12415] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 07/16/2013] [Accepted: 08/02/2013] [Indexed: 12/28/2022] Open
Abstract
The hypocretins (Hcrts), also known as orexins, are two peptides derived from a single precursor produced in the posterior lateral hypothalamus. Over the past decade, the orexin system has been associated with numerous physiological functions, including sleep/arousal, energy homeostasis, endocrine, visceral functions and pathological states, such as narcolepsy and drug abuse. Here, we review the discovery of Hcrt/orexins and their receptors and propose a hypothesis as to how the orexin system orchestrates these multifaceted physiological functions.
Collapse
Affiliation(s)
- Jingcheng Li
- Department of Physiology, Third Military Medical University, Chongqing, China
| | | | | |
Collapse
|
28
|
Roecker AJ, Reger TS, Mattern MC, Mercer SP, Bergman JM, Schreier JD, Cube RV, Cox CD, Li D, Lemaire W, Bruno JG, Harrell CM, Garson SL, Gotter AL, Fox SV, Stevens J, Tannenbaum PL, Prueksaritanont T, Cabalu TD, Cui D, Stellabott J, Hartman GD, Young SD, Winrow CJ, Renger JJ, Coleman PJ. Discovery of MK-3697: a selective orexin 2 receptor antagonist (2-SORA) for the treatment of insomnia. Bioorg Med Chem Lett 2014; 24:4884-90. [PMID: 25248679 DOI: 10.1016/j.bmcl.2014.08.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 08/15/2014] [Accepted: 08/19/2014] [Indexed: 01/12/2023]
Abstract
Orexin receptor antagonists have demonstrated clinical utility for the treatment of insomnia. The majority of clinical efforts to date have focused on the development of dual orexin receptor antagonists (DORAs), small molecules that antagonize both the orexin 1 and orexin 2 receptors. Our group has recently disclosed medicinal chemistry efforts to identify highly potent, orally bioavailable selective orexin 2 receptor antagonists (2-SORAs) that possess acceptable profiles for clinical development. Herein we report additional SAR studies within the 'triaryl' amide 2-SORA series focused on improvements in compound stability in acidic media and time-dependent inhibition of CYP3A4. These studies resulted in the discovery of 2,5-disubstituted isonicotinamide 2-SORAs such as compound 24 that demonstrated improved stability and TDI profiles as well as excellent sleep efficacy across species.
Collapse
Affiliation(s)
- Anthony J Roecker
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States.
| | - Thomas S Reger
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States.
| | - M Christa Mattern
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Swati P Mercer
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Jeffrey M Bergman
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - John D Schreier
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Rowena V Cube
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Christopher D Cox
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Dansu Li
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Wei Lemaire
- Department of In Vitro Pharmacology, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Joseph G Bruno
- Department of In Vitro Pharmacology, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - C Meacham Harrell
- Department of Neuroscience, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Susan L Garson
- Department of Neuroscience, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Anthony L Gotter
- Department of Neuroscience, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Steven V Fox
- Department of In Vivo Pharmacology, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Joanne Stevens
- Department of In Vivo Pharmacology, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Pamela L Tannenbaum
- Department of In Vivo Pharmacology, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Thomayant Prueksaritanont
- Department of Drug Metabolism, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Tamara D Cabalu
- Department of Drug Metabolism, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Donghui Cui
- Department of Drug Metabolism, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Joyce Stellabott
- Department of Basic Pharmaceutical Sciences, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - George D Hartman
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Steven D Young
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Christopher J Winrow
- Department of Neuroscience, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - John J Renger
- Department of Neuroscience, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Paul J Coleman
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 4, 770 Sumneytown Pike, West Point, PA 19486, United States
| |
Collapse
|
29
|
Giardino WJ, de Lecea L. Hypocretin (orexin) neuromodulation of stress and reward pathways. Curr Opin Neurobiol 2014; 29:103-8. [PMID: 25050887 DOI: 10.1016/j.conb.2014.07.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 06/11/2014] [Accepted: 07/02/2014] [Indexed: 01/08/2023]
Abstract
Hypocretin (also known as orexin) is a peptide neuromodulator that is expressed exclusively in the lateral hypothalamic area and plays a fundamental role in wakefulness and arousal. Chronic stress and compulsive drug-seeking are two examples of dysregulated states of hyperarousal that are influenced by hypocretin transmission throughout hypothalamic, extended amygdala, brainstem, and mesolimbic pathways. Here, we review current advances in the understanding of hypocretin's modulatory actions underlying conditions of negative and positive emotional valence, focusing particularly on mechanisms that facilitate adaptive (and maladaptive) responses to stressful or rewarding environmental stimuli. We conclude by discussing progress toward integrated theories for hypocretin modulation of divergent behavioral domains.
Collapse
Affiliation(s)
- William J Giardino
- Department of Psychiatry and Behavioral Sciences, Stanford University, 1201 Welch Road, P154, Stanford, CA 94305, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, 1201 Welch Road, P154, Stanford, CA 94305, USA.
| |
Collapse
|
30
|
Roecker AJ, Mercer SP, Harrell CM, Garson SL, Fox SV, Gotter AL, Prueksaritanont T, Cabalu TD, Cui D, Lemaire W, Winrow CJ, Renger JJ, Coleman PJ. Discovery of dual orexin receptor antagonists with rat sleep efficacy enabled by expansion of the acetonitrile-assisted/diphosgene-mediated 2,4-dichloropyrimidine synthesis. Bioorg Med Chem Lett 2014; 24:2079-85. [DOI: 10.1016/j.bmcl.2014.03.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 03/14/2014] [Accepted: 03/17/2014] [Indexed: 10/25/2022]
|
31
|
Neuroscience-driven discovery and development of sleep therapeutics. Pharmacol Ther 2014; 141:300-34. [DOI: 10.1016/j.pharmthera.2013.10.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 10/25/2013] [Indexed: 01/18/2023]
|
32
|
Chen X, Li S, Kirouac GJ. Blocking of corticotrophin releasing factor receptor-1 during footshock attenuates context fear but not the upregulation of prepro-orexin mRNA in rats. Pharmacol Biochem Behav 2014; 120:1-6. [PMID: 24491435 DOI: 10.1016/j.pbb.2014.01.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 01/21/2014] [Accepted: 01/26/2014] [Indexed: 10/25/2022]
Abstract
Hypothalamic neuropeptides called orexins (hypocretins) are well known for their roles in promoting arousal. Orexins have also been shown to play a role in fear and anxiety produced by the exposure of rats to an acute episode of moderately intense footshocks. Recent evidence indicates that stress activates orexin neurons through a corticotropin releasing factor (CRF) mechanism. In this study, we examined the effect of a CRF receptor-1 (CRF-R1) antagonist antalarmin (20mg/kg, i.p.) given before shock exposure on subsequent expression of contextual fear and the levels of prepro-orexin (ppOX) mRNA in the hypothalamus. Antalarmin decreased fear and ultrasonic vocalization expression to the shock context at 2 and 10 days after shock exposure. However, antalarmin did not prevent the increases in ppOX mRNA produced by the shock experience. This study provides evidence that blocking of CRF-R1 at the time of footshocks attenuates contextual fear. While an increase in the activity of the orexin system may contribute to fear, this activation does not appear to be sufficient for fear expression.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Department of Oral Biology, Faculty of Dentistry, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sa Li
- Department of Oral Biology, Faculty of Dentistry, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gilbert J Kirouac
- Department of Oral Biology, Faculty of Dentistry, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Psychiatry, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
33
|
Abstract
In 1998, our group discovered a cDNA that encoded the precursor of two putative neuropeptides that we called hypocretins for their hypothalamic expression and their similarity to the secretin family of neuropeptides. In the last 16 years, numerous studies have placed the hypocretin system as an integrator of homeostatic functions with a crucial, non-redundant function as arousal stabilizer. We recently applied optogenetic methods to interrogate the role of individual neuronal circuits in sleep-to-wake transitions. The neuronal connections between the hypocretin system and the locus coeruleus (LC) seem to be crucial in establishing the appropriate dynamic of spontaneous awakenings.
Collapse
|
34
|
Leonard CS, Kukkonen JP. Orexin/hypocretin receptor signalling: a functional perspective. Br J Pharmacol 2014; 171:294-313. [PMID: 23848055 PMCID: PMC3904253 DOI: 10.1111/bph.12296] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 06/17/2013] [Accepted: 07/03/2013] [Indexed: 10/26/2022] Open
Abstract
Multiple homeostatic systems are regulated by orexin (hypocretin) peptides and their two known GPCRs. Activation of orexin receptors promotes waking and is essential for expression of normal sleep and waking behaviour, with the sleep disorder narcolepsy resulting from the absence of orexin signalling. Orexin receptors also influence systems regulating appetite/metabolism, stress and reward, and are found in several peripheral tissues. Nevertheless, much remains unknown about the signalling pathways and targets engaged by native receptors. In this review, we integrate knowledge about the orexin receptor signalling capabilities obtained from studies in expression systems and various native cell types (as presented in Kukkonen and Leonard, this issue of British Journal of Pharmacology) with knowledge of orexin signalling in different tissues. The tissues reviewed include the CNS, the gastrointestinal tract, the pituitary gland, pancreas, adrenal gland, adipose tissue and the male reproductive system. We also summarize the findings in different native and recombinant cell lines, especially focusing on the different cascades in CHO cells, which is the most investigated cell line. This reveals that while a substantial gap exists between what is known about orexin receptor signalling and effectors in recombinant systems and native systems, mounting evidence suggests that orexin receptor signalling is more diverse than originally thought. Moreover, rather than being restricted to orexin receptor 'overexpressing' cells, this signalling diversity may be utilized by native receptors in a site-specific manner.
Collapse
Affiliation(s)
- C S Leonard
- Department of Physiology, New York Medical College, Valhalla, NY, USA
| | | |
Collapse
|
35
|
Roecker AJ, Mercer SP, Schreier JD, Cox CD, Fraley ME, Steen JT, Lemaire W, Bruno JG, Harrell CM, Garson SL, Gotter AL, Fox SV, Stevens J, Tannenbaum PL, Prueksaritanont T, Cabalu TD, Cui D, Stellabott J, Hartman GD, Young SD, Winrow CJ, Renger JJ, Coleman PJ. Discovery of 5''-chloro-N-[(5,6-dimethoxypyridin-2-yl)methyl]-2,2':5',3''-terpyridine-3'-carboxamide (MK-1064): a selective orexin 2 receptor antagonist (2-SORA) for the treatment of insomnia. ChemMedChem 2013; 9:311-22. [PMID: 24376006 DOI: 10.1002/cmdc.201300447] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Indexed: 01/12/2023]
Abstract
The field of small-molecule orexin antagonist research has evolved rapidly in the last 15 years from the discovery of the orexin peptides to clinical proof-of-concept for the treatment of insomnia. Clinical programs have focused on the development of antagonists that reversibly block the action of endogenous peptides at both the orexin 1 and orexin 2 receptors (OX1 R and OX2 R), termed dual orexin receptor antagonists (DORAs), affording late-stage development candidates including Merck's suvorexant (new drug application filed 2012). Full characterization of the pharmacology associated with antagonism of either OX1 R or OX2 R alone has been hampered by the dearth of suitable subtype-selective, orally bioavailable ligands. Herein, we report the development of a selective orexin 2 antagonist (2-SORA) series to afford a potent, orally bioavailable 2-SORA ligand. Several challenging medicinal chemistry issues were identified and overcome during the development of these 2,5-disubstituted nicotinamides, including reversible CYP inhibition, physiochemical properties, P-glycoprotein efflux and bioactivation. This article highlights structural modifications the team utilized to drive compound design, as well as in vivo characterization of our 2-SORA clinical candidate, 5''-chloro-N-[(5,6-dimethoxypyridin-2-yl)methyl]-2,2':5',3''-terpyridine-3'-carboxamide (MK-1064), in mouse, rat, dog, and rhesus sleep models.
Collapse
Affiliation(s)
- Anthony J Roecker
- Department of Medicinal Chemistry, Merck Research Laboratories, P.O. Box 4, Sumneytown Pike, West Point, PA 19486 (USA).
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Wong AYC, Borduas JF, Clarke S, Lee KFH, Béïque JC, Bergeron R. Calcium influx through N-type channels and activation of SK and TRP-like channels regulates tonic firing of neurons in rat paraventricular thalamus. J Neurophysiol 2013; 110:2450-64. [DOI: 10.1152/jn.00363.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The thalamus is a major relay and integration station in the central nervous system. While there is a large body of information on the firing and network properties of neurons contained within sensory thalamic nuclei, less is known about the neurons located in midline thalamic nuclei, which are thought to modulate arousal and homeostasis. One midline nucleus that has been implicated in mediating stress responses is the paraventricular nucleus of the thalamus (PVT). Like other thalamic neurons, these neurons display two distinct firing modes, burst and tonic. In contrast to burst firing, little is known about the ionic mechanisms modulating tonic firing in these cells. Here we performed a series of whole cell recordings to characterize tonic firing in PVT neurons in acute rat brain slices. We found that PVT neurons are able to fire sustained, low-frequency, weakly accommodating trains of action potentials in response to a depolarizing stimulus. Unexpectedly, PVT neurons displayed a very high propensity to enter depolarization block, occurring at stimulus intensities that would elicit tonic firing in other thalamic neurons. The tonic firing behavior of these cells is modulated by a functional interplay between N-type Ca2+ channels and downstream activation of small-conductance Ca2+-dependent K+ (SK) channels and a transient receptor potential (TRP)-like conductance. Thus these ionic conductances endow PVT neurons with a narrow dynamic range, which may have fundamental implications for the integrative properties of this nucleus.
Collapse
Affiliation(s)
| | | | - Stephen Clarke
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Kevin F. H. Lee
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Jean-Claude Béïque
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Richard Bergeron
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
37
|
Different levels in orexin concentrations and risk factors associated with higher orexin levels: comparison between detoxified opiate and methamphetamine addicts in 5 Chinese cities. BIOMED RESEARCH INTERNATIONAL 2013; 2013:282641. [PMID: 24102051 PMCID: PMC3786501 DOI: 10.1155/2013/282641] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 08/01/2013] [Indexed: 11/17/2022]
Abstract
This study sought to explore the degree of orexin levels in Chinese opiate and methamphetamine addicts and the differences between them. The cross-sectional study was conducted among detoxified drug addicts from Mandatory Detoxification Center (MDC) in five Chinese cities. Orexin levels were assayed with radioimmunoassay (RIA). Mann-Whitney U test and Kruskal-Wallis test were used to detect differences across groups, and logistic regression was used to explore the association between orexin levels and characteristics of demographic and drug abuse. Between November 2009 and January 2011, 285 opiates addicts, 112 methamphetamine addicts, and 79 healthy controls were enrolled. At drug withdrawal period, both opiate and methamphetamine addicts had lower median orexin levels than controls, and median orexin levels in opiate addicts were higher than those in methamphetamine addicts (all above P < 0.05). Adjusted odds of the above median concentration of orexin were higher for injection than "chasing the dragon" (AOR = 3.1, 95% CI = 1.2-7.9). No significant factors associated with orexin levels of methamphetamine addicts were found. Development of intervention method on orexin system by different administration routes especially for injected opiate addicts at detoxification phase may be significant and was welcome.
Collapse
|
38
|
Tsuneki H, Tokai E, Sugawara C, Wada T, Sakurai T, Sasaoka T. Hypothalamic orexin prevents hepatic insulin resistance induced by social defeat stress in mice. Neuropeptides 2013; 47:213-9. [PMID: 23510906 DOI: 10.1016/j.npep.2013.02.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 01/28/2013] [Accepted: 02/20/2013] [Indexed: 12/28/2022]
Abstract
Depression is associated with insulin resistance and type 2 diabetes, although the molecular mechanism behind the pathological link remains unclear. Orexin, a hypothalamic neuropeptide regulating energy and glucose homeostasis, has been implicated in the endogenous antidepressant mechanism. To clarify whether orexin is involved in the coordination between mental and metabolic functions, we investigated the influence of orexin deficiency on social interaction behavior and glucose metabolism in mice subjected to chronic social defeat stress. Chronic stress-induced glucose intolerance and systemic insulin resistance as well as social avoidance were ameliorated by calorie restriction in an orexin-dependent manner. Moreover, orexin-deficient mice maintained under ad libitum-fed conditions after defeat stress exhibited hyperinsulinemia and elevated HOMA-IR (homeostasis model assessment for insulin resistance), despite normal fasting blood glucose levels. In a pyruvate tolerance test to evaluate hepatic insulin sensitivity, chronic stress-induced abnormal glucose elevation was observed in orexin-deficient but not wild-type mice, although both types of mice were susceptible to chronic stress. In addition, insulin-induced phosphorylation of Akt in the liver was impaired in orexin-deficient but not wild-type mice after chronic stress. These results demonstrate that the central physiological actions of orexin under ad libitum-fed conditions are required for the adaptive response to chronic defeat stress, which can prevent the development of hepatic insulin resistance but not social avoidance behavior. Moreover, calorie restriction, a paradigm to strongly activate orexin neurons, appears to prevent the persistence of depression-like behavior per se, leading to the amelioration of impaired glucose metabolism after chronic stress; therefore, we suggest that hypothalamic orexin system is the key for inhibiting the exacerbating link between depression and type 2 diabetes.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama, Japan.
| | | | | | | | | | | |
Collapse
|
39
|
Steiner MA, Sciarretta C, Brisbare-Roch C, Strasser DS, Studer R, Jenck F. Examining the role of endogenous orexins in hypothalamus-pituitary-adrenal axis endocrine function using transient dual orexin receptor antagonism in the rat. Psychoneuroendocrinology 2013; 38:560-71. [PMID: 22917622 DOI: 10.1016/j.psyneuen.2012.07.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 07/24/2012] [Accepted: 07/25/2012] [Indexed: 11/25/2022]
Abstract
The orexin neuropeptide system regulates wakefulness and contributes to physiological and behavioral stress responses. Moreover, a role for orexins in modulating hypothalamus-pituitary-adrenal (HPA) axis activity has been proposed. Brain penetrating dual orexin receptor (OXR) antagonists such as almorexant decrease vigilance and have emerged as a novel therapeutic class for the treatment of insomnia. Almorexant was used here as a pharmacological tool to examine the role of endogenous orexin signaling in HPA axis endocrine function under natural conditions. After confirming the expression of prepro-orexin and OXR-1 and OXR-2 mRNA in hypothalamus, pituitary and adrenal glands, the effects of systemic almorexant were investigated on peripheral HPA axis hormone release in the rat under baseline, stress and pharmacological challenge conditions. Almorexant did not alter basal or stress-induced corticosterone release despite affecting wake and sleep stages (detected by radiotelemetric electroencephalography/electromyography) during the stress exposure. Moreover, almorexant did not affect the release of adrenocorticotropin (ACTH) and corticosterone at different time points along the diurnal rhythm, nor corticotrophin-releasing hormone (CRH)- and ACTH-stimulated neuroendocrine responses, measured in vivo under stress-free conditions. These results illustrate that dual OXR antagonists, despite modulating stress-induced wakefulness, do not interfere with endocrine HPA axis function in the rat. They converge to suggest that endogenous orexin signaling plays a minor role in stress hormone release under basal conditions and under challenge.
Collapse
Affiliation(s)
- Michel A Steiner
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, 4123 Allschwil, Switzerland.
| | | | | | | | | | | |
Collapse
|
40
|
The dual orexin receptor antagonist almorexant, alone and in combination with morphine, cocaine and amphetamine, on conditioned place preference and locomotor sensitization in the rat. Int J Neuropsychopharmacol 2013; 16:417-32. [PMID: 22436395 DOI: 10.1017/s1461145712000193] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Dual orexin receptor (OXR) antagonists emerge as a novel therapeutic class to treat insomnia that, based on anti-addictive effects of selective OXR type 1 antagonists in rats, might be associated with less abuse liability than commonly used γ-aminobutyric acid (GABA) receptor modulators. Here, we studied the effects of the sleep-enabling dual OXR antagonist almorexant on conditioned place preference (CPP) and locomotor sensitization in rats. First, we compared almorexant to the GABA metabolite γ-hydroxybutyrate (GHB), which is clinically used as a sleep-inducing drug and which is associated with mild abuse liability. Whereas conditioning with GHB induced significant place preference, conditioning with almorexant did not. Second, we tested the potential of almorexant to interfere with the conditioned rewarding or locomotor sensitizing effects related to psychostimulants or opiates. Almorexant attenuated the expression of CPP to high doses of cocaine (15 mg/kg) and d.l-amphetamine (2 mg/kg), but not to high dose of morphine (10 mg/kg). Conversely, almorexant interfered with the expression of locomotor sensitization to morphine, but not with that to cocaine and d.l-amphetamine. Third, we observed that chronic almorexant (12 d) treatment in morphine, cocaine or amphetamine pre-conditioned and locomotor-sensitized rats had no influence on the maintenance of CPP and locomotor sensitization when tested after almorexant washout. Our findings suggest that almorexant itself does not exert conditioned rewarding effects in the rat and that it may acutely interfere with the expression of CPP or locomotor sensitization in a drug-dependent manner (monoaminergic psychostimulants vs. opiates).
Collapse
|
41
|
Xiao F, Jiang M, Du D, Xia C, Wang J, Cao Y, Shen L, Zhu D. Orexin A regulates cardiovascular responses in stress-induced hypertensive rats. Neuropharmacology 2012; 67:16-24. [PMID: 23147417 DOI: 10.1016/j.neuropharm.2012.10.021] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 10/16/2012] [Accepted: 10/20/2012] [Indexed: 01/22/2023]
Abstract
Several pieces of evidence indicate that the rostral ventrolateral medulla (RVLM) is probably one of the key neural structures mediating the pressor effects of orexins in the brain. Nitric oxide synthase/nitric oxide (NOS/NO) system in the RVLM modulates cardiovascular activities. Our experiments were designed to test the hypothesis that orexin-A (OXA) is involved in the mechanism of stress-induced hypertension (SIH) by adjusting NOS/NO system in the RVLM. The stress-induced hypertensive rats (SIHR) model was established by electric foot-shocks and noises. Here we examined the expression of OXA immunoreactive (OXA-IR) cells in the lateral hypothalamus (LH) and the protein level of orexin 1 receptor (OX1R) in the RVLM of SIHR, and we found that the expressions of OXA-IR and OX1R were higher than those of the control group. The double-staining immunohistochemical evidence showed that OX1R immunoreactive (OX1R-IR) cells and neuronal nitric oxide synthase (nNOS) or inducible nitric oxide synthase (iNOS) immunoreactive (IR) cells were co-localizated in the RVLM. Microinjection of OXA (10, 50, 100 pmol/100 nl) into the unilateral (right) RVLM of control rats or SIHR produced pressor and tachycardiac effects in a dose-dependent manner. SB-408124 (100 pmol/100 nl, an antagonist of OX1R) or TCS OX2 29 (100 pmol/100 nl, an antagonist of OX2R) partly abolished the cardiovascular effects of exogenously-administrated OXA into the RVLM of control rats and SIHR, and lowered the increased systolic blood pressure (SBP) and heart rate (HR) of SIHR, with no difference in statistical significance between the two antagonists' effects. Microinjection into the RVLM of both control and SIHR groups of 7-Ni (0.05 pmol/100 nl, nNOS inhibitor) or Methylene Blue [100 pmol/100 nl, an inhibitor of soluble guanylate cyclase (sGC)] suppressed the OXA-induced increase of SBP and HR, whereas microinjection of AG (1, 10, 100 pmol/100 nl) had no obvious effects on the OXA-induced increase of SBP and HR. Our results indicate that OXA in the RVLM may participate in the central regulation of cardiovascular activities in SIHR, and OX1R and OX2R both have important roles in it. The cardiovascular effects of OXA in the RVLM may be induced by nNOS-derived NO, which activated sGC-associated signaling pathway.
Collapse
Affiliation(s)
- Fen Xiao
- Department of Physiology and Pathophysiology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Jakubcakova V, Flachskamm C, Landgraf R, Kimura M. Sleep phenotyping in a mouse model of extreme trait anxiety. PLoS One 2012; 7:e40625. [PMID: 22808211 PMCID: PMC3394752 DOI: 10.1371/journal.pone.0040625] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 06/11/2012] [Indexed: 01/19/2023] Open
Abstract
Background There is accumulating evidence that anxiety impairs sleep. However, due to high sleep variability in anxiety disorders, it has been difficult to state particular changes in sleep parameters caused by anxiety. Sleep profiling in an animal model with extremely high vs. low levels of trait anxiety might serve to further define sleep patterns associated with this psychopathology. Methodology/Principal Findings Sleep-wake behavior in mouse lines with high (HAB), low (LAB) and normal (NAB) anxiety-related behaviors was monitored for 24 h during baseline and recovery after 6 h sleep deprivation (SD). The amounts of each vigilance state, sleep architecture, and EEG spectral variations were compared between the mouse lines. In comparison to NAB mice, HAB mice slept more and exhibited consistently increased delta power during non-rapid eye movement (NREM) sleep. Their sleep patterns were characterized by heavy fragmentation, reduced maintenance of wakefulness, and frequent intrusions of rapid eye movement (REM) sleep. In contrast, LAB mice showed a robust sleep-wake rhythm with remarkably prolonged sleep latency and a long, persistent period of wakefulness. In addition, the accumulation of delta power after SD was impaired in the LAB line, as compared to HAB mice. Conclusions/Significance Sleep-wake patterns were significantly different between HAB and LAB mice, indicating that the genetic predisposition to extremes in trait anxiety leaves a biological scar on sleep quality. The enhanced sleep demand observed in HAB mice, with a strong drive toward REM sleep, may resemble a unique phenotype reflecting not only elevated anxiety but also a depression-like attribute.
Collapse
Affiliation(s)
| | | | | | - Mayumi Kimura
- Max Planck Institute of Psychiatry, Munich, Germany
- * E-mail:
| |
Collapse
|
43
|
Abstract
Driven by the demands and opportunities of modern life, many people habitually sleep less than 6 h a night. In the sleep clinic, chronic sleep restriction is recognized by the diagnosis of insufficient sleep syndrome (ICSD-9, 307.49-4), which is receiving increased scrutiny as a potential risk to metabolic health. Its relevance for the practicing endocrinologist is highlighted by a stream of epidemiological data that show an association of insufficient sleep with increased incidence of obesity and related morbidities. A central theme of this update is the notion that sleep loss incurs additional metabolic cost, which triggers a set of neuroendocrine, metabolic, and behavioral adaptations aimed at increasing food intake and conserving energy. Although this coordinated response may have evolved to offset the metabolic demands of extended wakefulness in natural habitats with limited food availability, it can be maladaptive in the context of a modern environment that allows many to overeat while maintaining a sedentary lifestyle without sufficient sleep. Importantly, such sleep loss-related metabolic adaptation may undermine the success of behavioral interventions based on reduced caloric intake and increased physical activity to lower metabolic risk in obesity-prone individuals. This emerging perspective is based on data from recently published human interventional studies and requires further experimental support. Nevertheless, it now seems prudent to recommend that overweight and obese individuals attempting to reduce their caloric intake and maintain increased physical activity should obtain adequate sleep and, if needed, seek effective treatment for any coexisting sleep disorders.
Collapse
Affiliation(s)
- Plamen D Penev
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Chicago, MC-1027, 5841 South Maryland Avenue, Chicago, Illinois 60637, USA.
| |
Collapse
|
44
|
Origins of arousal: roles for medullary reticular neurons. Trends Neurosci 2012; 35:468-76. [PMID: 22626543 DOI: 10.1016/j.tins.2012.04.008] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 04/18/2012] [Accepted: 04/23/2012] [Indexed: 01/12/2023]
Abstract
The existence of a primitive CNS function involved in the activation of all vertebrate behaviors, generalized arousal (GA), has been proposed. Here, we provide an overview of the neuroanatomical, neurophysiological and molecular properties of reticular neurons within the nucleus gigantocellularis (NGC) of the mammalian medulla, and propose that the properties of these neurons equip them to contribute powerfully to GA. We also explore the hypothesis that these neurons may have evolved from the Mauthner cell in the medulla of teleost fish, although NGC neurons have a wider range of action far beyond the specific escape network served by Mauthner cells. Understanding the neuronal circuits that control and regulate GA is central to understanding how motivated behaviors such as hunger, thirst and sexual behaviors arise.
Collapse
|
45
|
Laorden ML, Ferenczi S, Pintér-Kübler B, González-Martín LL, Lasheras MC, Kovács KJ, Milanés MV, Núñez C. Hypothalamic orexin--a neurons are involved in the response of the brain stress system to morphine withdrawal. PLoS One 2012; 7:e36871. [PMID: 22590628 PMCID: PMC3348891 DOI: 10.1371/journal.pone.0036871] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 04/09/2012] [Indexed: 11/18/2022] Open
Abstract
Both the hypothalamus-pituitary-adrenal (HPA) axis and the extrahypothalamic brain stress system are key elements of the neural circuitry that regulates the negative states during abstinence from chronic drug exposure. Orexins have recently been hypothesized to modulate the extended amygdala and to contribute to the negative emotional state associated with dependence. This study examined the impact of chronic morphine and withdrawal on the lateral hypothalamic (LH) orexin A (OXA) gene expression and activity as well as OXA involvement in the brain stress response to morphine abstinence. Male Wistar rats received chronic morphine followed by naloxone to precipitate withdrawal. The selective OX1R antagonist SB334867 was used to examine whether orexins' activity is related to somatic symptoms of opiate withdrawal and alterations in HPA axis and extended amygdala in rats dependent on morphine. OXA mRNA was induced in the hypothalamus during morphine withdrawal, which was accompanied by activation of OXA neurons in the LH. Importantly, SB334867 attenuated the somatic symptoms of withdrawal, and reduced morphine withdrawal-induced c-Fos expression in the nucleus accumbens (NAc) shell, bed nucleus of stria terminalis, central amygdala and hypothalamic paraventricular nucleus, but did not modify the HPA axis activity. These results highlight a critical role of OXA signalling, via OX1R, in activation of brain stress system to morphine withdrawal and suggest that all orexinergic subpopulations in the lateral hypothalamic area contribute in this response.
Collapse
Affiliation(s)
- M. Luisa Laorden
- Cellular and Molecular Pharmacology Laboratory, Faculty of Medicine, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Szilamér Ferenczi
- Molecular Neuroendocrinology Laboratory, Institute of Experimental Medicine, Budapest, Hungary
| | - Bernadett Pintér-Kübler
- Molecular Neuroendocrinology Laboratory, Institute of Experimental Medicine, Budapest, Hungary
| | - Laura L. González-Martín
- Cellular and Molecular Pharmacology Laboratory, Faculty of Medicine, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - M. Carmen Lasheras
- Cellular and Molecular Pharmacology Laboratory, Faculty of Medicine, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Krisztina J. Kovács
- Molecular Neuroendocrinology Laboratory, Institute of Experimental Medicine, Budapest, Hungary
| | - M. Victoria Milanés
- Cellular and Molecular Pharmacology Laboratory, Faculty of Medicine, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
- * E-mail:
| | - Cristina Núñez
- Cellular and Molecular Pharmacology Laboratory, Faculty of Medicine, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| |
Collapse
|
46
|
Abstract
Central chemoreception traditionally refers to a change in ventilation attributable to changes in CO2/H(+) detected within the brain. Interest in central chemoreception has grown substantially since the previous Handbook of Physiology published in 1986. Initially, central chemoreception was localized to areas on the ventral medullary surface, a hypothesis complemented by the recent identification of neurons with specific phenotypes near one of these areas as putative chemoreceptor cells. However, there is substantial evidence that many sites participate in central chemoreception some located at a distance from the ventral medulla. Functionally, central chemoreception, via the sensing of brain interstitial fluid H(+), serves to detect and integrate information on (i) alveolar ventilation (arterial PCO2), (ii) brain blood flow and metabolism, and (iii) acid-base balance, and, in response, can affect breathing, airway resistance, blood pressure (sympathetic tone), and arousal. In addition, central chemoreception provides a tonic "drive" (source of excitation) at the normal, baseline PCO2 level that maintains a degree of functional connectivity among brainstem respiratory neurons necessary to produce eupneic breathing. Central chemoreception responds to small variations in PCO2 to regulate normal gas exchange and to large changes in PCO2 to minimize acid-base changes. Central chemoreceptor sites vary in function with sex and with development. From an evolutionary perspective, central chemoreception grew out of the demands posed by air versus water breathing, homeothermy, sleep, optimization of the work of breathing with the "ideal" arterial PCO2, and the maintenance of the appropriate pH at 37°C for optimal protein structure and function.
Collapse
Affiliation(s)
- Eugene Nattie
- Dartmouth Medical School, Department of Physiology, Lebanon, New Hampshire, USA.
| | | |
Collapse
|
47
|
Abstract
Central chemoreception traditionally refers to a change in ventilation attributable to changes in CO2/H(+) detected within the brain. Interest in central chemoreception has grown substantially since the previous Handbook of Physiology published in 1986. Initially, central chemoreception was localized to areas on the ventral medullary surface, a hypothesis complemented by the recent identification of neurons with specific phenotypes near one of these areas as putative chemoreceptor cells. However, there is substantial evidence that many sites participate in central chemoreception some located at a distance from the ventral medulla. Functionally, central chemoreception, via the sensing of brain interstitial fluid H(+), serves to detect and integrate information on (i) alveolar ventilation (arterial PCO2), (ii) brain blood flow and metabolism, and (iii) acid-base balance, and, in response, can affect breathing, airway resistance, blood pressure (sympathetic tone), and arousal. In addition, central chemoreception provides a tonic "drive" (source of excitation) at the normal, baseline PCO2 level that maintains a degree of functional connectivity among brainstem respiratory neurons necessary to produce eupneic breathing. Central chemoreception responds to small variations in PCO2 to regulate normal gas exchange and to large changes in PCO2 to minimize acid-base changes. Central chemoreceptor sites vary in function with sex and with development. From an evolutionary perspective, central chemoreception grew out of the demands posed by air versus water breathing, homeothermy, sleep, optimization of the work of breathing with the "ideal" arterial PCO2, and the maintenance of the appropriate pH at 37°C for optimal protein structure and function.
Collapse
Affiliation(s)
- Eugene Nattie
- Dartmouth Medical School, Department of Physiology, Lebanon, New Hampshire, USA.
| | | |
Collapse
|
48
|
Schmidt FM, Arendt E, Steinmetzer A, Bruegel M, Kratzsch J, Strauss M, Baum P, Hegerl U, Schönknecht P. CSF-hypocretin-1 levels in patients with major depressive disorder compared to healthy controls. Psychiatry Res 2011; 190:240-3. [PMID: 21757240 DOI: 10.1016/j.psychres.2011.06.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 05/13/2011] [Accepted: 06/05/2011] [Indexed: 01/25/2023]
Abstract
Depressive patients exhibit symptoms of impaired regulation of wakefulness with hyperarousal and agitation as well as difficulties to falling asleep and preserving sleep continuity. Changes in hypocretin (hcrt) levels as polypeptides with impact on arousal and sleep-wake-regulation have been discussed in affective disorders but have not been investigated in patients with solely unipolar depression in comparison to healthy controls. In the present study, cerebrospinal fluid (CSF) levels of hcrt-1 for the first time were analyzed in patients with major depressive disorder (MDD) without psychiatric comorbidities and compared with levels in healthy controls. In 17 inpatients with MDD (mean Hamilton Depression Rating Scale 13.9 ± 7.4) and 10 healthy controls, CSF-hcrt-1 levels were measured using a fluorescence immunoassay (FIA). The mean hcrt-1 CSF levels in patients with MDD (74.3 ± 17.8pg/ml) did not differ compared to that of healthy controls (82.8 ± 22.1pg/ml). Hcrt-1 levels did not correlate with the severity of depressive episode, the symptoms of depression or the number of episodes. Although autonomic and neurohumoral signs of hyperarousal are common in MDD, hcrt-1 levels in CSF were not found to be altered in MDD compared to healthy controls. Whether hcrt-1 levels are altered in depressive patients exhibiting impaired vigilance regulation has to be investigated in further studies combining measures of CSF-hcrt-1 with electroencephalography.
Collapse
Affiliation(s)
- Frank Martin Schmidt
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Leipzig, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Duguay D, Bélanger-Nelson E, Mongrain V, Beben A, Khatchadourian A, Cermakian N. Dynein light chain Tctex-type 1 modulates orexin signaling through its interaction with orexin 1 receptor. PLoS One 2011; 6:e26430. [PMID: 22028875 PMCID: PMC3197643 DOI: 10.1371/journal.pone.0026430] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 09/27/2011] [Indexed: 11/19/2022] Open
Abstract
Orexins (OX-A, OX-B) are neuropeptides involved in the regulation of the sleep-wake cycle, feeding and reward, via activation of orexin receptors 1 and 2 (OX1R, OX2R). The loss of orexin peptides or functional OX2R has been shown to cause the sleep disorder, narcolepsy. Since the regulation of orexin receptors remains largely undefined, we searched for novel protein partners of the intracellular tail of orexin receptors. Using a yeast two-hybrid screening strategy in combination with co-immunoprecipitation experiments, we found interactions between OX1R and the dynein light chains Tctex-type 1 and 3 (Dynlt1, Dynlt3). These interactions were mapped to the C-terminal region of the dynein light chains and to specific residues within the last 10 amino acids of OX1R. Hence, we hypothesized that dynein light chains could regulate orexin signaling. In HEK293 cells expressing OX1R, stimulation with OX-A produced a less sustained extracellular signal-regulated kinases 1/2 (ERK1/2) activation when Dynlt1 was co-expressed, while it was prolonged under reduced Dynlt1 expression. The amount of OX1R located at the plasma membrane as well as the kinetics and extent of OX-A-induced internalization of OX1R (disappearance from membrane) were not altered by Dynlt1. However, Dynlt1 reduced the localization of OX1R in early endosomes following initial internalization. Taken together, these data suggest that Dynlt1 modulates orexin signaling by regulating OX1R, namely its intracellular localization following ligand-induced internalization.
Collapse
Affiliation(s)
- David Duguay
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Quebec, Canada
- Department of Psychiatry, McGill University, Montréal, Quebec, Canada
| | - Erika Bélanger-Nelson
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| | - Valérie Mongrain
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Quebec, Canada
- Department of Psychiatry, McGill University, Montréal, Quebec, Canada
| | - Anna Beben
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Quebec, Canada
| | - Armen Khatchadourian
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Quebec, Canada
| | - Nicolas Cermakian
- Laboratory of Molecular Chronobiology, Douglas Mental Health University Institute, Montréal, Quebec, Canada
- Department of Psychiatry, McGill University, Montréal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
50
|
Droogleever Fortuyn H, Mulders P, Renier W, Buitelaar J, Overeem S. Narcolepsy and psychiatry: An evolving association of increasing interest. Sleep Med 2011; 12:714-9. [PMID: 21689985 DOI: 10.1016/j.sleep.2011.01.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Revised: 12/13/2010] [Accepted: 01/05/2011] [Indexed: 11/26/2022]
|