1
|
Qi M, Won J, Rodriguez C, Storace DA. Glutamatergic heterogeneity in the neuropeptide projections from the lateral hypothalamus to the mouse olfactory bulb. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.16.638511. [PMID: 39990441 PMCID: PMC11844501 DOI: 10.1101/2025.02.16.638511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The direct pathway from the lateral hypothalamus to the mouse olfactory bulb (OB) includes neurons that express the neuropeptide orexin-A, and others that do not. The OB-projecting neurons that do not express orexin-A are present in an area of the lateral hypothalamus known to contain neurons that express the neuropeptide melanin-concentrating hormone (MCH). We used virally mediated anterograde tract tracing and immunohistochemistry for orexin-A and MCH to demonstrate that the OB is broadly innervated by axon projections from both populations of neurons. Orexin-A and MCH were expressed in each OB layer across its anterior to posterior axis. Both orexin-A and MCH neurons are genetically heterogeneous, with subsets that co-express an isoform of vesicular glutamate transporter (VGLUT). We used high-resolution confocal imaging to test whether the projections from orexin-A and MCH neurons to the OB reflect this glutamatergic heterogeneity. The majority (~57%) of putative orexin-A axon terminals overlapped with VGLUT2, with smaller proportions that co-expressed VGLUT1, or that did not overlap with either VGLUT1 or VGLUT2. In contrast, only ~26% of putative MCH axon terminals overlapped with VGLUT2, with the majority not overlapping with either VGLUT. Therefore, the projections from the lateral hypothalamus to the OB are genetically heterogeneous and include neurons that can release two different neuropeptides. The projections from both populations are themselves genetically heterogeneous with distinct ratios of glutamatergic and non-glutamatergic axon terminals.
Collapse
Affiliation(s)
- Meizhu Qi
- Department of Biological Science, Florida State University, Tallahassee, FL
- Program in Neuroscience, Florida State University, Tallahassee, FL
| | - Julia Won
- Department of Biological Science, Florida State University, Tallahassee, FL
| | | | - Douglas A. Storace
- Department of Biological Science, Florida State University, Tallahassee, FL
- Program in Neuroscience, Florida State University, Tallahassee, FL
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL
| |
Collapse
|
2
|
Richards BK, Ch'ng SS, Simon AB, Pang TY, Kim JH, Lawrence AJ, Perry CJ. Relaxin family peptide receptor 3 (RXFP3) expressing cells in the zona incerta/lateral hypothalamus augment behavioural arousal. J Neurochem 2025; 169:e16217. [PMID: 39233365 PMCID: PMC11658188 DOI: 10.1111/jnc.16217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/15/2024] [Accepted: 08/17/2024] [Indexed: 09/06/2024]
Abstract
Fear-related psychopathologies, such as post-traumatic stress disorder, are linked to dysfunction in neural circuits that govern fear memory and arousal. The lateral hypothalamus (LH) and zona incerta (ZI) regulate fear, but our understanding of the precise neural circuits and cell types involved remains limited. Here, we examined the role of relaxin family peptide receptor 3 (RXFP3) expressing cells in the LH/ZI in conditioned fear expression and general arousal in male RXFP3-Cre mice. We found that LH/ZI RXFP3+ (LH/ZIRXFP3) cells projected strongly to fear learning, stress, and arousal centres, notably, the periaqueductal grey, lateral habenula, and nucleus reuniens. These cells do not express hypocretin/orexin or melanin-concentrating hormone but display putative efferent connectivity with LH hypocretin/orexin+ neurons and dopaminergic A13 cells. Following Pavlovian fear conditioning, chemogenetically activating LH/ZIRXFP3 cells reduced fear expression (freezing) overall but also induced jumping behaviour and increased locomotor activity. Therefore, the decreased freezing was more likely to reflect enhanced arousal rather than reduced fear. Indeed, stimulating these cells produced distinct patterns of coactivation between several motor, stress, and arousal regions, as measured by Fos expression. These results suggest that activating LH/ZIRXFP3 cells generates brain-wide activation patterns that augment behavioural arousal.
Collapse
Affiliation(s)
- Brandon K. Richards
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- School of Psychological SciencesMacquarie UniversityNorth RydeNew South WalesAustralia
| | - Sarah S. Ch'ng
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Ariel B. Simon
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Terence Y. Pang
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- Institute of Health and Sports (IHES)Victoria UniversityFootscrayVictoriaAustralia
| | - Jee Hyun Kim
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- IMPACT—The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin UniversityGeelongVictoriaAustralia
| | - Andrew J. Lawrence
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Christina J. Perry
- The Florey Institute of Neuroscience and Mental HealthParkvilleVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- School of Psychological SciencesMacquarie UniversityNorth RydeNew South WalesAustralia
| |
Collapse
|
3
|
Vringer M, Zhou J, Gool JK, Bijlenga D, Lammers GJ, Fronczek R, Schinkelshoek MS. Recent insights into the pathophysiology of narcolepsy type 1. Sleep Med Rev 2024; 78:101993. [PMID: 39241492 DOI: 10.1016/j.smrv.2024.101993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/09/2024]
Abstract
Narcolepsy type 1 (NT1) is a sleep-wake disorder in which people typically experience excessive daytime sleepiness, cataplexy and other sleep-wake disturbances impairing daily life activities. NT1 symptoms are due to hypocretin deficiency. The cause for the observed hypocretin deficiency remains unclear, even though the most likely hypothesis is that this is due to an auto-immune process. The search for autoantibodies and autoreactive T-cells has not yet produced conclusive evidence for or against the auto-immune hypothesis. Other mechanisms, such as reduced corticotrophin-releasing hormone production in the paraventricular nucleus have recently been suggested. There is no reversive treatment, and the therapeutic approach is symptomatic. Early diagnosis and appropriate NT1 treatment is essential, especially in children to prevent impaired cognitive, emotional and social development. Hypocretin receptor agonists have been designed to replace the attenuated hypocretin signalling. Pre-clinical and clinical trials have shown encouraging initial results. A better understanding of NT1 pathophysiology may contribute to faster diagnosis or treatments, which may cure or prevent it.
Collapse
Affiliation(s)
- Marieke Vringer
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jingru Zhou
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jari K Gool
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands; Department of Anatomy & Neurosciences, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Compulsivity, Impulsivity and Attention, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Denise Bijlenga
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Gert Jan Lammers
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Rolf Fronczek
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Mink S Schinkelshoek
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
4
|
Heiss JE, Zhong P, Lee SM, Yamanaka A, Kilduff TS. Distinct lateral hypothalamic CaMKIIα neuronal populations regulate wakefulness and locomotor activity. Proc Natl Acad Sci U S A 2024; 121:e2316150121. [PMID: 38593074 PMCID: PMC11032496 DOI: 10.1073/pnas.2316150121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/14/2024] [Indexed: 04/11/2024] Open
Abstract
For nearly a century, evidence has accumulated indicating that the lateral hypothalamus (LH) contains neurons essential to sustain wakefulness. While lesion or inactivation of LH neurons produces a profound increase in sleep, stimulation of inhibitory LH neurons promotes wakefulness. To date, the primary wake-promoting cells that have been identified in the LH are the hypocretin/orexin (Hcrt) neurons, yet these neurons have little impact on total sleep or wake duration across the 24-h period. Recently, we and others have identified other LH populations that increase wakefulness. In the present study, we conducted microendoscopic calcium imaging in the LH concomitant with EEG and locomotor activity (LMA) recordings and found that a subset of LH neurons that express Ca2+/calmodulin-dependent protein kinase IIα (CaMKIIα) are preferentially active during wakefulness. Chemogenetic activation of these neurons induced sustained wakefulness and greatly increased LMA even in the absence of Hcrt signaling. Few LH CaMKIIα-expressing neurons are hypocretinergic or histaminergic while a small but significant proportion are GABAergic. Ablation of LH inhibitory neurons followed by activation of the remaining LH CaMKIIα neurons induced similar levels of wakefulness but blunted the LMA increase. Ablated animals showed no significant changes in sleep architecture but both spontaneous LMA and high theta (8 to 10 Hz) power during wakefulness were reduced. Together, these findings indicate the existence of two subpopulations of LH CaMKIIα neurons: an inhibitory population that promotes locomotion without affecting sleep architecture and an excitatory population that promotes prolonged wakefulness even in the absence of Hcrt signaling.
Collapse
Affiliation(s)
- Jaime E. Heiss
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA94025
| | - Peng Zhong
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA94025
| | - Stephanie M. Lee
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA94025
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya464-8601, Japan
| | - Thomas S. Kilduff
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA94025
| |
Collapse
|
5
|
Kim WJ, Kim HS. Emerging and upcoming therapies in insomnia. Transl Clin Pharmacol 2024; 32:1-17. [PMID: 38586124 PMCID: PMC10990727 DOI: 10.12793/tcp.2024.32.e5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 04/09/2024] Open
Abstract
Insomnia, commonly treated with benzodiazepine (BZD) receptor agonists, presents challenges due to associated serious side effects such as abuse and dependence. To address these concerns, many researches have been conducted to develop and advance both pharmacological and non-pharmacological interventions. Dual orexin receptor antagonists (DORAs), which include suvorexant, daridorexant and lemborexant, have recently been approved by United States Food and Drug Administration (US FDA) as a novel pharmacotherapeutic alternative. Unlike BZD receptor agonists that act as positive allosteric modulators of the gamma-aminobutyric acid type A subunit alpha 1 receptor, DORAs function by binding to both orexin receptor types 1 and 2, and inhibiting the action of the wake-promoting orexin neuropeptide. These drugs induce normal sleep without sleep stage change, do not impair attention and memory performance, and facilitate easier awakening. However, more real-world safety information is needed. Selective orexin-2 receptor antagonists (2-SORAs) is under clinical developments. This review provides an overview of the mechanism of action in relation to insomnia, pharmacokinetics, efficacy and safety information of DORAs and SORA. According to insomnia management guidelines, the first-line treatment for chronic insomnia is cognitive behavioral therapy for insomnia (CBT-I). Although it has proven effective in improving sleep-related quality of life, it has several restrictions limitations due to a face-to-face format. Recently, prescription digital therapy such as Somryst® was approved by US FDA. Somryst®, a smartphone app-based CBT-I, demonstrated meaningful responses in patients. However, digital limitations may impact scalability. Overall, these developments offer promising alternatives for insomnia treatment, emphasizing safety, efficacy, and accessibility.
Collapse
Affiliation(s)
- Woo-Ju Kim
- Inje University College of Medicine, Busan, Korea
| | - Ho-Sook Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Korea
| |
Collapse
|
6
|
Narai E, Yoshimura Y, Honaga T, Mizoguchi H, Yamanaka A, Hiyama TY, Watanabe T, Koba S. Orexinergic neurons contribute to autonomic cardiovascular regulation for locomotor exercise. J Physiol 2024. [PMID: 38380995 DOI: 10.1113/jp285791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/26/2024] [Indexed: 02/22/2024] Open
Abstract
While the hypothalamic orexinergic nervous system is established as having a pivotal role in the long-term regulation of various organismic functions, including wakefulness, metabolism and hypertensive states, whether this system contributes to the rapid autonomic cardiovascular regulation during physical activity remains elusive. This study aimed to elucidate the role of the orexinergic nervous system in transmitting volitional motor signals, i.e. central command, to drive somatomotor and sympathetic cardiovascular responses. We first found that this system is activated by voluntary locomotor exercise as evidenced by an increased expression of Fos, a marker of neural activation, in the orexinergic neurons of Sprague-Dawley rats engaged in spontaneous wheel running. Next, using transgenic Orexin-Cre rats for optogenetic manipulation of orexinergic neurons, we found that optogenetic excitation of orexinergic neurons caused sympathoexcitation on a subsecond timescale under anaesthesia. In freely moving conscious rats, this excitatory stimulation rapidly elicited exploration-like behaviours, predominantly locomotor activity, along with pressor and tachycardiac responses. Meanwhile, optogenetic inhibition of orexinergic neurons during spontaneous wheel running immediately suppressed locomotor activities and blood pressure elevation without affecting basal cardiovascular homeostasis. Taken together, these findings demonstrate the essential role of the orexinergic nervous system in the central circuitry that transmits central command signals for locomotor exercise. This study not only offers insights into the brain circuit mechanisms precisely regulating autonomic cardiovascular systems during voluntary exercise but also likely contributes to our understanding of brain mechanisms underlying abnormal cardiovascular adjustments to exercise in pathological conditions, such as hypertension. KEY POINTS: The hypothalamic orexinergic nervous system plays various roles in the long-term regulation of autonomic and endocrine functions, as well as motivated behaviours. We present a novel, rapid role of the orexinergic nervous system, revealing its significance as a crucial substrate in the brain circuit mechanisms that coordinate somatomotor and autonomic cardiovascular controls for locomotor exercise. Our data demonstrate that orexinergic neurons relay volitional motor signals, playing a necessary and sufficient role in the autonomic cardiovascular regulation required for locomotor exercise in rats. The findings contribute to our understanding of how the brain precisely regulates autonomic cardiovascular systems during voluntary exercise, providing insights into the central neural mechanisms that enhance physical performance moment-by-moment during exercise.
Collapse
Affiliation(s)
- Emi Narai
- Division of Integrative Physiology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Yuki Yoshimura
- Division of Integrative Physiology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Takaho Honaga
- Division of Integrative Physiology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Hiroyuki Mizoguchi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Akihiro Yamanaka
- Chinese Institute for Brain Research, Beijing (CIBR), Beijing, China
| | - Takeshi Y Hiyama
- Division of Integrative Physiology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Tatsuo Watanabe
- Division of Integrative Physiology, Tottori University Faculty of Medicine, Yonago, Japan
| | - Satoshi Koba
- Division of Integrative Physiology, Tottori University Faculty of Medicine, Yonago, Japan
- Division of Veterinary Physiology, Tottori University Faculty of Agriculture, Tottori, Japan
| |
Collapse
|
7
|
Li HT, Viskaitis P, Bracey E, Peleg-Raibstein D, Burdakov D. Transient targeting of hypothalamic orexin neurons alleviates seizures in a mouse model of epilepsy. Nat Commun 2024; 15:1249. [PMID: 38341419 PMCID: PMC10858876 DOI: 10.1038/s41467-024-45515-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Lateral hypothalamic (LH) hypocretin/orexin neurons (HONs) control brain-wide electrical excitation. Abnormally high excitation produces epileptic seizures, which affect millions of people and need better treatments. HON population activity spikes from minute to minute, but the role of this in seizures is unknown. Here, we describe correlative and causal links between HON activity spikes and seizures. Applying temporally-targeted HON recordings and optogenetic silencing to a male mouse model of acute epilepsy, we found that pre-seizure HON activity predicts and controls the electrophysiology and behavioral pathology of subsequent seizures. No such links were detected for HON activity during seizures. Having thus defined the time window where HONs influence seizures, we targeted it with LH deep brain stimulation (DBS), which inhibited HON population activity, and produced seizure protection. Collectively, these results uncover a feature of brain activity linked to seizures, and demonstrate a proof-of-concept treatment that controls this feature and alleviates epilepsy.
Collapse
Affiliation(s)
- Han-Tao Li
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology | ETH Zurich, 8603, Schwerzenbach, Switzerland
- Section of Epilepsy, Department of Neurology, Chang Gung Memorial Hospital at Linkou Medical Center and Chang Gung University College of Medicine, 333, Taoyuan, Taiwan
| | - Paulius Viskaitis
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology | ETH Zurich, 8603, Schwerzenbach, Switzerland
| | - Eva Bracey
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology | ETH Zurich, 8603, Schwerzenbach, Switzerland
| | - Daria Peleg-Raibstein
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology | ETH Zurich, 8603, Schwerzenbach, Switzerland
| | - Denis Burdakov
- Department of Health Sciences and Technology, Swiss Federal Institute of Technology | ETH Zurich, 8603, Schwerzenbach, Switzerland.
| |
Collapse
|
8
|
Pintwala SK, Peever J. Brain Circuits Underlying Narcolepsy. Neuroscientist 2023; 29:751-766. [PMID: 34704497 DOI: 10.1177/10738584211052263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Narcolepsy is a sleep disorder manifesting symptoms such as excessive daytime sleepiness and often cataplexy, a sudden and involuntary loss of muscle activity during wakefulness. The underlying neuropathological basis of narcolepsy is the loss of orexin neurons from the lateral hypothalamus. To date numerous animal models of narcolepsy have been produced in the laboratory, being invaluable tools for delineating the brain circuits of narcolepsy. This review will examine the evidence regarding the function of the orexin system, and how loss of this wake-promoting system manifests in excessive daytime sleepiness. This review will also outline the brain circuits controlling cataplexy, focusing on the contribution of orexin signaling loss in narcolepsy. Although our understanding of the brain circuits of narcolepsy has made great progress in recent years, much remains to be understood.
Collapse
Affiliation(s)
| | - John Peever
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Adamantidis AR, de Lecea L. Sleep and the hypothalamus. Science 2023; 382:405-412. [PMID: 37883555 DOI: 10.1126/science.adh8285] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/08/2023] [Indexed: 10/28/2023]
Abstract
Neural substrates of wakefulness, rapid eye movement sleep (REMS), and non-REMS (NREMS) in the mammalian hypothalamus overlap both anatomically and functionally with cellular networks that support physiological and behavioral homeostasis. Here, we review the roles of sleep neurons of the hypothalamus in the homeostatic control of thermoregulation or goal-oriented behaviors during wakefulness. We address how hypothalamic circuits involved in opposing behaviors such as core body temperature and sleep compute conflicting information and provide a coherent vigilance state. Finally, we highlight some of the key unresolved questions and challenges, and the promise of a more granular view of the cellular and molecular diversity underlying the integrative role of the hypothalamus in physiological and behavioral homeostasis.
Collapse
Affiliation(s)
- Antoine R Adamantidis
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Luis de Lecea
- Department of Psychiatry and Behavioural Sciences, Stanford, CA, USA
- Wu Tsai Neurosciences Institute Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
10
|
Dawson M, Terstege DJ, Jamani N, Tsutsui M, Pavlov D, Bugescu R, Epp JR, Leinninger GM, Sargin D. Hypocretin/orexin neurons encode social discrimination and exhibit a sex-dependent necessity for social interaction. Cell Rep 2023; 42:112815. [PMID: 37459234 DOI: 10.1016/j.celrep.2023.112815] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 05/20/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023] Open
Abstract
The hypothalamus plays a crucial role in the modulation of social behavior by encoding internal states. The hypothalamic hypocretin/orexin neurons, initially identified as regulators of sleep and appetite, are important for emotional and motivated behaviors. However, their role in social behavior remains unclear. Using fiber photometry and behavioral analysis, we show here that hypocretin neurons differentially encode social discrimination based on the nature of social encounters. The optogenetic inhibition of hypocretin neuron activity or blocking of hcrt-1 receptors reduces the amount of time mice are engaged in social interaction in males but not in females. Reduced hcrt-1 receptor signaling during social interaction is associated with altered activity in the insular cortex and ventral tegmental area in males. Our data implicating hypocretin neurons as sexually dimorphic regulators within social networks have significant implications for the treatment of neuropsychiatric diseases with social dysfunction, particularly considering varying prevalence among sexes.
Collapse
Affiliation(s)
- Matthew Dawson
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Dylan J Terstege
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Naila Jamani
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Mio Tsutsui
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Dmitrii Pavlov
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Raluca Bugescu
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Jonathan R Epp
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Gina M Leinninger
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Derya Sargin
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
11
|
Saitoh T, Sakurai T. The Present and Future of Synthetic Orexin Receptor Agonists. Peptides 2023:171051. [PMID: 37422012 DOI: 10.1016/j.peptides.2023.171051] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/16/2023] [Accepted: 07/02/2023] [Indexed: 07/10/2023]
Abstract
The neuropeptide orexin/hypocretin plays a crucial role in various physiological processes, including the regulation of sleep/wakefulness, appetite, emotion and the reward system. Dysregulation of orexin signaling has been implicated in hypersomnia, especially in narcolepsy, which is a chronic neurological disorder characterized by excessive daytime sleepiness (EDS), sudden loss of muscle tone while awake (cataplexy), sleep paralysis, and hallucinations. Small-molecule orexin receptor agonists have emerged as promising therapeutics for these disorders, and significant progress has been made in this field in the past decade. This review summarizes recent advances in the design and synthesis of orexin receptor agonists, with a focus on peptidic and small-molecule OX2R-selective, dual, and OX1R-selective agonists. The review discusses the key structural features and pharmacological properties of these agonists, as well as their potential therapeutic applications. DATA AVAILABILITY: No data was used for the research described in the article.
Collapse
Affiliation(s)
- Tsuyoshi Saitoh
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Takeshi Sakurai
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
12
|
Khouma A, Moeini MM, Plamondon J, Richard D, Caron A, Michael NJ. Histaminergic regulation of food intake. Front Endocrinol (Lausanne) 2023; 14:1202089. [PMID: 37448468 PMCID: PMC10338010 DOI: 10.3389/fendo.2023.1202089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/06/2023] [Indexed: 07/15/2023] Open
Abstract
Histamine is a biogenic amine that acts as a neuromodulator within the brain. In the hypothalamus, histaminergic signaling contributes to the regulation of numerous physiological and homeostatic processes, including the regulation of energy balance. Histaminergic neurons project extensively throughout the hypothalamus and two histamine receptors (H1R, H3R) are strongly expressed in key hypothalamic nuclei known to regulate energy homeostasis, including the paraventricular (PVH), ventromedial (VMH), dorsomedial (DMH), and arcuate (ARC) nuclei. The activation of different histamine receptors is associated with differential effects on neuronal activity, mediated by their different G protein-coupling. Consequently, activation of H1R has opposing effects on food intake to that of H3R: H1R activation suppresses food intake, while H3R activation mediates an orexigenic response. The central histaminergic system has been implicated in atypical antipsychotic-induced weight gain and has been proposed as a potential therapeutic target for the treatment of obesity. It has also been demonstrated to interact with other major regulators of energy homeostasis, including the central melanocortin system and the adipose-derived hormone leptin. However, the exact mechanisms by which the histaminergic system contributes to the modification of these satiety signals remain underexplored. The present review focuses on recent advances in our understanding of the central histaminergic system's role in regulating feeding and highlights unanswered questions remaining in our knowledge of the functionality of this system.
Collapse
Affiliation(s)
- Axelle Khouma
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Moein Minbashi Moeini
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Julie Plamondon
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
| | - Denis Richard
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Medicine, Université Laval, Québec, QC, Canada
| | - Alexandre Caron
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
- Montreal Diabetes Research Center, Montreal, QC, Canada
| | - Natalie Jane Michael
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Quebec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| |
Collapse
|
13
|
Kniazkina M, Dyachuk V. Does EGFR Signaling Mediate Orexin System Activity in Sleep Initiation? Int J Mol Sci 2023; 24:ijms24119505. [PMID: 37298454 DOI: 10.3390/ijms24119505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/21/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Sleep-wake cycle disorders are an important symptom of many neurological diseases, including Parkinson's disease, Alzheimer's disease, and multiple sclerosis. Circadian rhythms and sleep-wake cycles play a key role in maintaining the health of organisms. To date, these processes are still poorly understood and, therefore, need more detailed elucidation. The sleep process has been extensively studied in vertebrates, such as mammals and, to a lesser extent, in invertebrates. A complex, multi-step interaction of homeostatic processes and neurotransmitters provides the sleep-wake cycle. Many other regulatory molecules are also involved in the cycle regulation, but their functions remain largely unclear. One of these signaling systems is epidermal growth factor receptor (EGFR), which regulates the activity of neurons in the modulation of the sleep-wake cycle in vertebrates. We have evaluated the possible role of the EGFR signaling pathway in the molecular regulation of sleep. Understanding the molecular mechanisms that underlie sleep-wake regulation will provide critical insight into the fundamental regulatory functions of the brain. New findings of sleep-regulatory pathways may provide new drug targets and approaches for the treatment of sleep-related diseases.
Collapse
Affiliation(s)
- Marina Kniazkina
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Vyacheslav Dyachuk
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| |
Collapse
|
14
|
Sheibani M, Shayan M, Khalilzadeh M, Ghasemi M, Dehpour AR. Orexin receptor antagonists in the pathophysiology and treatment of sleep disorders and epilepsy. Neuropeptides 2023; 99:102335. [PMID: 37003137 DOI: 10.1016/j.npep.2023.102335] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023]
Abstract
The correlation between sleep and epilepsy has been argued over the past decades among scientists. Although the similarities and contrasts between sleep and epilepsy had been considered, their intertwined nature was not revealed until the nineteenth century. Sleep is recognized as a recurring state of mind and body through alternating brain electrical activities. It is documented that sleep disorders are associated with epilepsy. The origin, suppression, and spread of seizures are affected by sleep. As such, in patients with epilepsy, sleep disorders are a frequent comorbidity. Meanwhile, orexin, a wake-promoting neuropeptide, provides a bidirectional effect on both sleep and epilepsy. Orexin and its cognate receptors, orexin receptor type 1 (OX1R) and type 2 (OX2R), orchestrate their effects by activating various downstream signaling pathways. Although orexin was considered a therapeutic target in insomnia shortly after its discovery, its potential usefulness for psychiatric disorders and epileptic seizures has been suggested in the pre-clinical studies. This review aimed to discuss whether the relationship between sleep, epilepsy, and orexin is clearly reciprocal.
Collapse
Affiliation(s)
- Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Shayan
- Experimental Medicine Research Centre, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Khalilzadeh
- Experimental Medicine Research Centre, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Ghasemi
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Centre, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Mogavero MP, Silvani A, Lanza G, DelRosso LM, Ferini-Strambi L, Ferri R. Targeting Orexin Receptors for the Treatment of Insomnia: From Physiological Mechanisms to Current Clinical Evidence and Recommendations. Nat Sci Sleep 2023; 15:17-38. [PMID: 36713640 PMCID: PMC9879039 DOI: 10.2147/nss.s201994] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/08/2023] [Indexed: 01/23/2023] Open
Abstract
After a detailed description of orexins and their roles in sleep and other medical disorders, we discuss here the current clinical evidence on the effects of dual (DORAs) or selective (SORAs) orexin receptor antagonists on insomnia with the aim to provide recommendations for their further assessment in a context of personalized and precision medicine. In the last decade, many trials have been conducted with orexin receptor antagonists, which represent an innovative and valid therapeutic option based on the multiple mechanisms of action of orexins on different biological circuits, both centrally and peripherally, and their role in a wide range of medical conditions which are often associated with insomnia. A very interesting aspect of this new category of drugs is that they have limited abuse liability and their discontinuation does not seem associated with significant rebound effects. Further studies on the efficacy of DORAs are required, especially on children and adolescents and in particular conditions, such as menopause. Which DORA is most suitable for each patient, based on comorbidities and/or concomitant treatments, should be the focus of further careful research. On the contrary, studies on SORAs, some of which seem to be appropriate also in insomnia in patients with psychiatric diseases, are still at an early stage and, therefore, do not allow to draw definite conclusions.
Collapse
Affiliation(s)
- Maria P Mogavero
- Vita-Salute San Raffaele University, Milan, Italy
- Sleep Disorders Center, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Silvani
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe Lanza
- Sleep Research Centre, Oasi Research Institute - IRCCS, Troina, Italy
- Department of Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Lourdes M DelRosso
- Pulmonary and Sleep Medicine, University of California San Francisco-Fresno, Fresno, CA, USA
| | - Luigi Ferini-Strambi
- Vita-Salute San Raffaele University, Milan, Italy
- Sleep Disorders Center, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Raffaele Ferri
- Sleep Research Centre, Oasi Research Institute - IRCCS, Troina, Italy
| |
Collapse
|
16
|
Shao YQ, Fan L, Wu WY, Zhu YJ, Xu HT. A developmental switch between electrical and neuropeptide communication in the ventromedial hypothalamus. Curr Biol 2022; 32:3137-3145.e3. [PMID: 35659861 DOI: 10.1016/j.cub.2022.05.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/15/2022] [Accepted: 05/11/2022] [Indexed: 12/29/2022]
Abstract
Dissecting neural connectivity patterns within local brain regions is an essential step to understanding the function of the brain.1 Neural microcircuits in brain regions, such as the neocortex and the hippocampus, have been extensively studied.2 By contrast, the microcircuit in the hypothalamus remains largely uncharacterized. The hypothalamus is crucial for animals' survival and reproduction.3 Knowledge of how different hypothalamic nuclei coordinate with each other and outside brain regions for hypothalamus-related functions has been significantly advanced.4-9 Although there are limited studies on the neural microcircuit in the lateral hypothalamus (LHA)10,11 and the suprachiasmatic nucleus (SCN),12,13 the patterns of neural microcircuits in most of the given hypothalamic nuclei remain largely unknown. This study applied combinatory approaches to address the local neural circuit pattern in the ventromedial hypothalamus (VMH) and other hypothalamic nuclei. We discovered a unique neural circuit design in the VMH. Neurons in the VMH were electrically coupled at the early postnatal stage like ones in the neocortex.14 However, unlike neocortical neurons,14,15 they developed very few chemical synapses after the disappearance of electrical synapses. Instead, VMH neurons communicated with neuropeptides. The similar scarceness of synaptic connectivity found in other hypothalamic nuclei further indicated that the lack of synaptic connections is a unique feature for local neural circuits in most adult hypothalamic nuclei. Thus, our findings provide a solid synaptic basis at the cellular level to understand hypothalamic functions better.
Collapse
Affiliation(s)
- Yin-Qi Shao
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liu Fan
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wen-Yan Wu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi-Jun Zhu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hua-Tai Xu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China.
| |
Collapse
|
17
|
Elahdadi Salmani M, Sarfi M, Goudarzi I. Hippocampal orexin receptors: Localization and function. VITAMINS AND HORMONES 2022; 118:393-421. [PMID: 35180935 DOI: 10.1016/bs.vh.2021.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Orexin (hypocretin) is secreted from the perifornical/lateral hypothalamus and is well known for sleep regulation. Orexin has two, orexin A and B, transcripts and two receptors, type 1 and 2 (OX1R and OX2R), located in the plasma membrane of neurons in different brain areas, including the hippocampus involved in learning, memory, seizures, and epilepsy, as physiologic and pathologic phenomena. OX1R is expressed in the dentate gyrus and CA1 and the OX2R in the CA3 areas. Orexin enhances learning and memory as well as reward, stress, seizures, and epilepsy, partly through OX1Rs, while either aggravating or alleviating those phenomena via OX2Rs. OX1Rs activation induces long-term changes of synaptic responses in the hippocampus, an age and concentration-dependent manner. Briefly, we will review the localization and functions of hippocampal orexin receptors, their role in learning, memory, stress, reward, seizures, epilepsy, and hippocampal synaptic plasticity.
Collapse
Affiliation(s)
| | | | - Iran Goudarzi
- School of Biology, Damghan University, Damghan, Iran
| |
Collapse
|
18
|
Sotelo MI, Tyan J, Markunas C, Sulaman BA, Horwitz L, Lee H, Morrow JG, Rothschild G, Duan B, Eban-Rothschild A. Lateral hypothalamic neuronal ensembles regulate pre-sleep nest-building behavior. Curr Biol 2022; 32:806-822.e7. [PMID: 35051354 PMCID: PMC10455050 DOI: 10.1016/j.cub.2021.12.053] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 11/17/2021] [Accepted: 12/21/2021] [Indexed: 01/08/2023]
Abstract
The transition from wakefulness to sleep requires striking alterations in brain activity, physiology, and behavior, yet the precise neuronal circuit elements facilitating this transition remain unclear. Prior to sleep onset, many animal species display characteristic behaviors, including finding a safe location, performing hygiene-related behaviors, and preparing a space for sleep. It has been proposed that the pre-sleep period is a transitional phase in which engaging in a specific behavioral repertoire de-arouses the brain and facilitates the wake-to-sleep transition, yet both causal evidence for this premise and an understanding of the neuronal circuit elements involved are lacking. Here, we combine detailed behavioral observations, EEG-EMG recordings, selective targeting, and activity modulation of pre-sleep-active neurons to reveal the behaviors preceding sleep initiation and their underlying neurobiological mechanisms. We show that mice engage in temporally structured behaviors with stereotypic EEG signatures prior to sleep and that nest-building and grooming become significantly more prevalent with sleep proximity. We next demonstrate that the ability to build a nest promotes the initiation and consolidation of sleep and that the lack of nesting material chronically fragments sleep. Lastly, we identify broadly projecting and predominantly glutamatergic neuronal ensembles in the lateral hypothalamus that regulate the motivation to engage in pre-sleep nest-building behavior and gate sleep initiation and intensity. Our study provides causal evidence for the facilitatory role of pre-sleep behaviors in sleep initiation and consolidation and a functional characterization of the neuronal underpinnings regulating a sleep-related and goal-directed complex behavior.
Collapse
Affiliation(s)
- Maria I Sotelo
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jean Tyan
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chelsea Markunas
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bibi A Sulaman
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lorraine Horwitz
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hankyu Lee
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Joshua G Morrow
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gideon Rothschild
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA; Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bo Duan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
19
|
Thomas CS, Mohammadkhani A, Rana M, Qiao M, Baimel C, Borgland SL. Optogenetic stimulation of lateral hypothalamic orexin/dynorphin inputs in the ventral tegmental area potentiates mesolimbic dopamine neurotransmission and promotes reward-seeking behaviours. Neuropsychopharmacology 2022; 47:728-740. [PMID: 34663867 PMCID: PMC8782948 DOI: 10.1038/s41386-021-01196-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 01/30/2023]
Abstract
Reward and reinforcement processes are critical for survival and propagation of genes. While numerous brain systems underlie these processes, a cardinal role is ascribed to mesolimbic dopamine. However, ventral tegmental area (VTA) dopamine neurons receive complex innervation and various neuromodulatory factors, including input from lateral hypothalamic (LH) orexin/hypocretin neurons which also express and co-release the neuropeptide, dynorphin. Dynorphin in the VTA induces aversive conditioning through the Kappa opioid receptor (KOR) and decreases dopamine when administered intra-VTA. Exogenous application of orexin or orexin 1 receptor (oxR1) antagonists in the VTA bidirectionally modulates dopamine-driven motivation and reward-seeking behaviours, including the attribution of motivational value to primary rewards and associated conditioned stimuli. However, the effect of endogenous stimulation of LH orexin/dynorphin-containing projections to the VTA and the potential contribution of co-released dynorphin on mesolimbic dopamine and reward related processes remains uncharacterised. We combined optogenetic, electrochemical, and behavioural approaches to examine this. We found that optical stimulation of LH orexin/dynorphin inputs in the VTA potentiates mesolimbic dopamine neurotransmission in the nucleus accumbens (NAc) core, produces real time and conditioned place preference, and increases the food cue-directed orientation in a Pavlovian conditioning procedure. LH orexin/dynorphin potentiation of NAc dopamine release and real time place preference was blocked by an oxR1, but not KOR antagonist. Thus, rewarding effects associated with optical stimulation of LH orexin/dynorphin inputs in the VTA are predominantly driven by orexin rather than dynorphin.
Collapse
Affiliation(s)
- Catherine S. Thomas
- grid.22072.350000 0004 1936 7697Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1 Canada
| | - Aida Mohammadkhani
- grid.22072.350000 0004 1936 7697Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1 Canada
| | - Madiha Rana
- grid.22072.350000 0004 1936 7697Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1 Canada
| | - Min Qiao
- grid.22072.350000 0004 1936 7697Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1 Canada
| | - Corey Baimel
- grid.22072.350000 0004 1936 7697Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1 Canada
| | - Stephanie L. Borgland
- grid.22072.350000 0004 1936 7697Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1 Canada
| |
Collapse
|
20
|
Arrigoni E, Fuller PM. The Role of the Central Histaminergic System in Behavioral State Control. Curr Top Behav Neurosci 2022; 59:447-468. [PMID: 34595740 DOI: 10.1007/7854_2021_263] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Histamine is a small monoamine signaling molecule that plays a role in many peripheral and central physiological processes, including the regulation of wakefulness. The tuberomammillary nucleus is the sole neuronal source of histamine in the brain, and histamine neurons are thought to promote wakefulness and vigilance maintenance - under certain environmental and/or behavioral contexts - through their diffuse innervation of the cortex and other wake-promoting brain circuits. Histamine neurons also contain a number of other putative neurotransmitters, although the functional role of these co-transmitters remains incompletely understood. Within the brain histamine operates through three receptor subtypes that are located on pre- and post-synaptic membranes. Some histamine receptors exhibit constitutive activity, and hence exist in an activated state even in the absence of histamine. Newer medications used to reduce sleepiness in narcolepsy patients in fact enhance histamine signaling by blunting the constitutive activity of these histamine receptors. In this chapter, we provide an overview of the central histamine system with an emphasis on its role in behavioral state regulation and how drugs targeting histamine receptors are used clinically to treat a wide range of sleep-wake disorders.
Collapse
Affiliation(s)
- Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Patrick M Fuller
- Department of Neurological Surgery, University of California Davis School of Medicine, Davis, CA, USA
| |
Collapse
|
21
|
Peleg-Raibstein D, Burdakov D. Do orexin/hypocretin neurons signal stress or reward? Peptides 2021; 145:170629. [PMID: 34416308 DOI: 10.1016/j.peptides.2021.170629] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/14/2021] [Indexed: 12/23/2022]
Abstract
Hypothalamic neurons that produce the peptide transmitters orexins/hypocretins (HONs) broadcast their predominantly neuroexcitatory outputs to the entire brain via their extremely wide axonal projections. HONs were originally reported to be activated by food deprivation, and to stimulate arousal, energy expenditure, and eating. This led to extensive studies of HONs in the context of nutrient-sensing and energy balance control. While activation of HONs by body energy depletion continues to be supported by experimental evidence, it has also become clear that HONs are robustly activated not only by nutrient depletion, but also by diverse sensory stimuli (both neutral and those associated with rewarding or aversive events), seemingly unrelated to each other or to energy balance. One theory that could unify these findings is that all these stimuli signal "stress" - defined either as a potentially harmful state, or an awareness of reward deficiency. If HON activity is conceptualized as a cumulative representation of stress, then many of the reported HONs outputs - including EEG arousal, sympathetic activation, place avoidance, and exploratory behaviours - could be viewed as logical stress-counteracting responses. We discuss evidence for and against this unifying theory of HON function, including the alterations in HON activity observed in anxiety and depression disorders. We propose that, in order to orchestrate stress-countering responses, HONs need to coactivate motivation and aversion brain systems, and the impact of HON stimulation on affective states may be perceived as rewarding or aversive depending on the baseline HON activity.
Collapse
Affiliation(s)
| | - Denis Burdakov
- Department of Health Sciences and Technology, ETH Zürich, Switzerland.
| |
Collapse
|
22
|
Pittaras E, Colas D, Chuluun B, Allocca G, Heller C. Enhancing sleep after training improves memory in Down syndrome model mice. Sleep 2021; 45:6383427. [PMID: 34618890 DOI: 10.1093/sleep/zsab247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 09/09/2021] [Indexed: 11/13/2022] Open
Abstract
Down syndrome (DS) is a genetic disorder caused by the presence of all or part of a third copy of chromosome 21. DS is associated with cognitive disabilities, for which there are no drug therapies. In spite of significant behavioral and pharmacological efforts to treat cognitive disabilities, new and continued efforts are still necessary. Over sixty percent of children with DS are reported to have sleep apnea that disrupt normal sleep. Normal and adequate sleep is necessary to maintain optimal cognitive functions. Therefore, we asked whether improved quality and/or quantity of sleep could improve cognitive capacities of people with DS. To investigate this possibility, we used the Ts65Dn mouse model of DS and applied two methods for enhancing their sleep following training on mouse memory tasks. A behavioral method was to impose sleep deprivation prior to training resulting in sleep rebound following the training. A pharmacologic method, hypocretin receptor 2 antagonist, was used immediately after the training to enhance subsequent sleep knowing that hypocretin is involved in the maintenance of wake. Our behavioral method resulted in a sleep reorganization that decreased wake and increased REM sleep following the training associated with an improvement of recognition memory and spatial memory in the DS model mice. Our pharmacologic approach decreased wake and increased NREM sleep and was associated with improvement only in the spatial memory task. These results show that enhancing sleep after the training in a memory task improves memory consolidation in a mouse model of DS.
Collapse
Affiliation(s)
- E Pittaras
- Stanford University, Department: Biology, Stanford, CA, USA
| | | | - B Chuluun
- Stanford University, Department: Biology, Stanford, CA, USA
| | - G Allocca
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia and School of Biomedical Sciences, University of Melbourne, Parkville, VIC, Australia and Somnivore Pty. Ltd., Bacchus Marsh, VIC, Australia
| | - C Heller
- Stanford University, Department: Biology, Stanford, CA, USA
| |
Collapse
|
23
|
Ye Q, Zhang X. Serotonin activates paraventricular thalamic neurons through direct depolarization and indirect disinhibition from zona incerta. J Physiol 2021; 599:4883-4900. [PMID: 34510418 DOI: 10.1113/jp282088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/10/2021] [Indexed: 12/11/2022] Open
Abstract
Paraventricular thalamus (PVT) is a midline thalamic area that receives dense GABA projections from zona incerta (ZI) for the regulation of feeding behaviours. Activation of central serotonin (5-HT) signalling is known to inhibit food intake. Although previous studies have reported both 5-HT fibres and receptors in the PVT, it remains unknown how 5-HT regulates PVT neurons and whether PVT 5-HT signalling is involved in the control of food intake. Using slice patch-clamp recordings in combination with optogenetics, we found that 5-HT not only directly excited PVT neurons by activating 5-HT7 receptors to modulate hyperpolarization-activated cyclic nucleotide-gated (HCN) channels but also disinhibited these neurons by acting on presynaptic 5-HT1A receptors to reduce GABA inhibition. Specifically, 5-HT depressed photostimulation-evoked inhibitory postsynaptic currents (eIPSCs) in PVT neurons innervated by channelrhodopsin-2-positive GABA axons from ZI. Using paired-pulse photostimulation, we found 5-HT increased paired-pulse ratios of eIPSCs, suggesting 5-HT decreases ZI-PVT GABA release. Furthermore, we found that exposure to a high-fat-high-sucrose diet for 2 weeks impaired both 5-HT inhibition of ZI-PVT GABA transmission and 5-HT excitation of PVT neurons. Using retrograde tracer in combination with immunocytochemistry and slice electrophysiology, we found that PVT-projecting dorsal raphe neurons expressed 5-HT and were inhibited by food deprivation. Together, our study reveals the mechanism by which 5-HT activates PVT neurons through both direct excitation and indirect disinhibition from the ZI. The downregulation in 5-HT excitation and disinhibition of PVT neurons may contribute to the development of overeating and obesity after chronic high-fat diet. KEY POINTS: Serotonin (5-HT) depolarizes and excites paraventricular thalamus (PVT) neurons. 5-HT7 receptors are responsible for 5-HT excitation of PVT neurons and the coupling of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels to 5-HT receptors in part mediates the excitatory effect of 5-HT. 5-HT depresses the frequency of spontaneous inhibitory but not excitatory postsynaptic currents in PVT neurons. 5-HT1A receptors contribute to the depressive effect of 5-HT on inhibitory transmissions. 5-HT inhibits GABA release from zona incerta (ZI) GABA terminals in PVT. Chronic high-fat diet not only impairs 5-HT inhibition of the ZI-PVT GABA transmission but also downregulates 5-HT excitation of PVT neurons. PVT-projecting dorsal raphe neurons express 5-HT and are inhibited by food deprivation.
Collapse
Affiliation(s)
- Qiying Ye
- Department of Psychology, Florida State University, Tallahassee, FL, USA
| | - Xiaobing Zhang
- Department of Psychology, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
24
|
Concetti C, Burdakov D. Orexin/Hypocretin and MCH Neurons: Cognitive and Motor Roles Beyond Arousal. Front Neurosci 2021; 15:639313. [PMID: 33828450 PMCID: PMC8019792 DOI: 10.3389/fnins.2021.639313] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/01/2021] [Indexed: 02/01/2023] Open
Abstract
The lateral hypothalamus (LH) is classically implicated in sleep-wake control. It is the main source of orexin/hypocretin and melanin-concentrating hormone (MCH) neuropeptides in the brain, which have been both implicated in arousal state switching. These neuropeptides are produced by non-overlapping LH neurons, which both project widely throughout the brain, where release of orexin and MCH activates specific postsynaptic G-protein-coupled receptors. Optogenetic manipulations of orexin and MCH neurons during sleep indicate that they promote awakening and REM sleep, respectively. However, recordings from orexin and MCH neurons in awake, moving animals suggest that they also act outside sleep/wake switching. Here, we review recent studies showing that both orexin and MCH neurons can rapidly (sub-second-timescale) change their firing when awake animals experience external stimuli, or during self-paced exploration of objects and places. However, the sensory-behavioral correlates of orexin and MCH neural activation can be quite different. Orexin neurons are generally more dynamic, with about 2/3rds of them activated before and during self-initiated running, and most activated by sensory stimulation across sensory modalities. MCH neurons are activated in a more select manner, for example upon self-paced investigation of novel objects and by certain other novel stimuli. We discuss optogenetic and chemogenetic manipulations of orexin and MCH neurons, which combined with pharmacological blockade of orexin and MCH receptors, imply that these rapid LH dynamics shape fundamental cognitive and motor processes due to orexin and MCH neuropeptide actions in the awake brain. Finally, we contemplate whether the awake control of psychomotor brain functions by orexin and MCH are distinct from their “arousal” effects.
Collapse
Affiliation(s)
- Cristina Concetti
- Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Denis Burdakov
- Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
25
|
Wang RF, Guo H, Jiang SY, Liu ZL, Qu WM, Huang ZL, Wang L. Control of wakefulness by lateral hypothalamic glutamatergic neurons in male mice. J Neurosci Res 2021; 99:1689-1703. [PMID: 33713502 DOI: 10.1002/jnr.24828] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/21/2021] [Accepted: 03/01/2021] [Indexed: 11/10/2022]
Abstract
The lateral hypothalamus (LH) plays a key role in the maintenance of cortical activation and wakefulness. In the LH, the two main neuronal cell populations consist of excitatory glutamatergic neurons and inhibitory GABAergic neurons. Recent studies have shown that inhibitory LH GABAergic neurons are wake-promoting. However, the mechanism by which excitatory LH glutamatergic neurons contribute to sleep-wake regulation remains unclear. Using fiber photometry in male mice, we demonstrated that LH glutamatergic neurons exhibited high activities during both wakefulness and rapid eye movement sleep. Chemogenetic activation of LH glutamatergic neurons induced an increase in wakefulness that lasted for 6 hr, whereas suppression of LH glutamatergic neuronal activity caused a reduction in wakefulness. Brief optogenetic activation of LH glutamatergic neurons induced an immediate transition from slow-wave sleep to wakefulness, and long-lasting optogenetic stimulation of these neurons maintained wakefulness. Moreover, we found that LH-locus coeruleus/parabrachial nucleus and LH-basal forebrain projections mediated the wake-promoting effects of LH glutamatergic neurons. Taken together, our data indicate that LH glutamatergic neurons are essential for the induction and maintenance of wakefulness. The results presented here may advance our understanding of the role of LH in the control of wakefulness.
Collapse
Affiliation(s)
- Ren-Fei Wang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Han Guo
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Shi-Yu Jiang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Zi-Long Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Zhi-Li Huang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lu Wang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| |
Collapse
|
26
|
Yaeger JD, Krupp KT, Gale JJ, Summers CH. Counterbalanced microcircuits for Orx1 and Orx2 regulation of stress reactivity. MEDICINE IN DRUG DISCOVERY 2020. [DOI: 10.1016/j.medidd.2020.100059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
27
|
Burdakov D, Peleg-Raibstein D. The hypothalamus as a primary coordinator of memory updating. Physiol Behav 2020; 223:112988. [DOI: 10.1016/j.physbeh.2020.112988] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/05/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022]
|
28
|
Fast sensory representations in the lateral hypothalamus and their roles in brain function. Physiol Behav 2020; 222:112952. [DOI: 10.1016/j.physbeh.2020.112952] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/26/2020] [Accepted: 04/28/2020] [Indexed: 01/12/2023]
|
29
|
Michael NJ, Zigman JM, Williams KW, Elmquist JK. Electrophysiological Properties of Genetically Identified Histaminergic Neurons. Neuroscience 2020; 444:183-195. [PMID: 32599122 DOI: 10.1016/j.neuroscience.2020.06.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 11/27/2022]
Abstract
Histaminergic neurons of the tuberomammillary nucleus (TMN) are important regulators of behavioral and homeostatic processes. Previous work suggested that histaminergic neurons exhibit a characteristic electrophysiological signature, allowing for their identification in brain slice preparations. However, these previous investigations focused on neurons in the ventral subregion of the TMN of rats. Consequently, it remains unclear whether such electrophysiological properties extend to mice, including other subregions of the TMN, and the potential for differences between males and females. To further characterize the electrophysiological properties of histaminergic neurons, we performed whole-cell patch-clamp recordings on transgenic mice expressing Cre recombinase in histidine decarboxylase (HDC)-expressing cells; the sole enzyme for histamine synthesis (Hdc-cre::tdTomato). Despite similarities with the electrophysiological properties reported in rats, we observed considerable variability in mouse HDC neuron passive membrane properties, action potential firing, and intrinsic subthreshold active membrane properties. Overall, the electrophysiological properties of HDC neurons appeared similar across subregions of the TMN, consistent with a lack of topographical organization in this nucleus. Moreover, we found no obvious sex differences in the electrical excitability of HDC neurons. However, our data reveal a diversity in the electrophysiological properties of genetically identified histaminergic neurons from mice not previously appreciated from rat studies. Thus, these data highlight the utility of mouse genetics to target the widespread histaminergic neuronal population within the TMN and support the idea that histaminergic neurons are a heterogeneous neuronal population.
Collapse
Affiliation(s)
- Natalie J Michael
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390-9077, United States; Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390-9077, United States
| | - Kevin W Williams
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390-9077, United States.
| | - Joel K Elmquist
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390-9077, United States; Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390-9077, United States.
| |
Collapse
|
30
|
Michael NJ, Caron A, Lee CE, Castorena CM, Lee S, Zigman JM, Williams KW, Elmquist JK. Melanocortin regulation of histaminergic neurons via perifornical lateral hypothalamic melanocortin 4 receptors. Mol Metab 2020; 35:100956. [PMID: 32244183 PMCID: PMC7082550 DOI: 10.1016/j.molmet.2020.01.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/23/2020] [Accepted: 01/28/2020] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Histaminergic neurons of the tuberomammillary nucleus (TMN) are wake-promoting and contribute to the regulation of energy homeostasis. Evidence indicates that melanocortin 4 receptors (MC4R) are expressed within the TMN. However, whether the melanocortin system influences the activity and function of TMN neurons expressing histidine decarboxylase (HDC), the enzyme required for histamine synthesis, remains undefined. METHODS We utilized Hdc-Cre mice in combination with whole-cell patch-clamp electrophysiology and in vivo chemogenetic techniques to determine whether HDC neurons receive metabolically relevant information via the melanocortin system. RESULTS We found that subsets of HDC-expressing neurons were excited by melanotan II (MTII), a non-selective melanocortin receptor agonist. Use of melanocortin receptor selective agonists (THIQ, [D-Trp8]-γ-MSH) and inhibitors of synaptic transmission (TTX, CNQX, AP5) indicated that the effect was mediated specifically by MC4Rs and involved a glutamatergic dependent presynaptic mechanism. MTII enhanced evoked excitatory post-synaptic currents (EPSCs) originating from electrical stimulation of the perifornical lateral hypothalamic area (PeFLH), supportive of melanocortin effects on the glutamatergic PeFLH projection to the TMN. Finally, in vivo chemogenetic inhibition of HDC neurons strikingly enhanced the anorexigenic effects of intracerebroventricular administration of MTII, suggesting that MC4R activation of histaminergic neurons may restrain the anorexigenic effects of melanocortin system activation. CONCLUSIONS These experiments identify a functional interaction between the melanocortin and histaminergic systems and suggest that HDC neurons act naturally to restrain the anorexigenic effect of melanocortin system activation. These findings may have implications for the control of arousal and metabolic homeostasis, especially in the context of obesity, in which both processes are subjected to alterations.
Collapse
MESH Headings
- Animals
- Behavior, Animal/drug effects
- Eating/drug effects
- Excitatory Postsynaptic Potentials/drug effects
- Histamine/metabolism
- Histidine Decarboxylase/genetics
- Histidine Decarboxylase/metabolism
- Hypothalamic Area, Lateral/cytology
- Hypothalamic Area, Lateral/metabolism
- Locomotion/drug effects
- Male
- Melanocortins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neurons/drug effects
- Neurons/metabolism
- Peptides, Cyclic/pharmacology
- Receptor, Melanocortin, Type 4/agonists
- Receptor, Melanocortin, Type 4/genetics
- Receptor, Melanocortin, Type 4/metabolism
- alpha-MSH/analogs & derivatives
- alpha-MSH/pharmacology
Collapse
Affiliation(s)
- Natalie J Michael
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, 75390-9077, USA
| | - Alexandre Caron
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, 75390-9077, USA
| | - Charlotte E Lee
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, 75390-9077, USA
| | - Carlos M Castorena
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, 75390-9077, USA
| | - Syann Lee
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, 75390-9077, USA
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, 75390-9077, USA
| | - Kevin W Williams
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, 75390-9077, USA.
| | - Joel K Elmquist
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, 75390-9077, USA; Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, 75390-9077, USA.
| |
Collapse
|
31
|
Hung CJ, Ono D, Kilduff TS, Yamanaka A. Dual orexin and MCH neuron-ablated mice display severe sleep attacks and cataplexy. eLife 2020; 9:54275. [PMID: 32314734 PMCID: PMC7173968 DOI: 10.7554/elife.54275] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 04/03/2020] [Indexed: 12/25/2022] Open
Abstract
Orexin/hypocretin-producing and melanin-concentrating hormone-producing (MCH) neurons are co-extensive in the hypothalamus and project throughout the brain to regulate sleep/wakefulness. Ablation of orexin neurons decreases wakefulness and results in a narcolepsy-like phenotype, whereas ablation of MCH neurons increases wakefulness. Since it is unclear how orexin and MCH neurons interact to regulate sleep/wakefulness, we generated transgenic mice in which both orexin and MCH neurons could be ablated. Double-ablated mice exhibited increased wakefulness and decreased both rapid eye movement (REM) and non-REM (NREM) sleep. Double-ablated mice showed severe cataplexy compared with orexin neuron-ablated mice, suggesting that MCH neurons normally suppress cataplexy. Double-ablated mice also showed frequent sleep attacks with elevated spectral power in the delta and theta range, a unique state that we call 'delta-theta sleep'. Together, these results indicate a functional interaction between orexin and MCH neurons in vivo that suggests the synergistic involvement of these neuronal populations in the sleep/wakefulness cycle.
Collapse
Affiliation(s)
- Chi Jung Hung
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan.,CREST, JST, Honcho Kawaguchi, Saitama, Japan
| | - Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan.,CREST, JST, Honcho Kawaguchi, Saitama, Japan
| | - Thomas S Kilduff
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, United States
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan.,CREST, JST, Honcho Kawaguchi, Saitama, Japan
| |
Collapse
|
32
|
Karnani MM, Schöne C, Bracey EF, González JA, Viskaitis P, Li HT, Adamantidis A, Burdakov D. Role of spontaneous and sensory orexin network dynamics in rapid locomotion initiation. Prog Neurobiol 2020; 187:101771. [PMID: 32058043 PMCID: PMC7086232 DOI: 10.1016/j.pneurobio.2020.101771] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/04/2022]
Abstract
Appropriate motor control is critical for normal life, and requires hypothalamic hypocretin/orexin neurons (HONs). HONs are slowly regulated by nutrients, but also display rapid (subsecond) activity fluctuations in vivo. The necessity of these activity bursts for sensorimotor control and their roles in specific phases of movement are unknown. Here we show that temporally-restricted optosilencing of spontaneous or sensory-evoked HON bursts disrupts locomotion initiation, but does not affect ongoing locomotion. Conversely, HON optostimulation initiates locomotion with subsecond delays in a frequency-dependent manner. Using 2-photon volumetric imaging of activity of >300 HONs during sensory stimulation and self-initiated locomotion, we identify several locomotion-related HON subtypes, which distinctly predict the probability of imminent locomotion initiation, display distinct sensory responses, and are differentially modulated by food deprivation. By causally linking HON bursts to locomotion initiation, these findings reveal the sensorimotor importance of rapid spontaneous and evoked fluctuations in HON ensemble activity.
Collapse
Affiliation(s)
- Mahesh M Karnani
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland; The Francis Crick Institute, London, UK; Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK.
| | - Cornelia Schöne
- The Francis Crick Institute, London, UK; Systems Neuroscience, University of Göttingen, Germany
| | - Edward F Bracey
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland; The Francis Crick Institute, London, UK
| | - J Antonio González
- The Francis Crick Institute, London, UK; The Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, UK
| | - Paulius Viskaitis
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Han-Tao Li
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Antoine Adamantidis
- Department of Neurology, Inselspital, University of Bern, Switzerland; Department of Biomedical Research, University of Bern, Switzerland
| | - Denis Burdakov
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland; The Francis Crick Institute, London, UK; Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK; Neuroscience Center Zürich (ZNZ), ETH Zürich and University of Zürich, Zürich, Switzerland
| |
Collapse
|
33
|
Li SB, de Lecea L. The hypocretin (orexin) system: from a neural circuitry perspective. Neuropharmacology 2020; 167:107993. [PMID: 32135427 DOI: 10.1016/j.neuropharm.2020.107993] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/23/2020] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
Hypocretin/orexin neurons are distributed restrictively in the hypothalamus, a brain region known to orchestrate diverse functions including sleep, reward processing, food intake, thermogenesis, and mood. Since the hypocretins/orexins were discovered more than two decades ago, extensive studies have accumulated concrete evidence showing the pivotal role of hypocretin/orexin in diverse neural modulation. New method of viral-mediated tracing system offers the possibility to map the monosynaptic inputs and detailed anatomical connectivity of Hcrt neurons. With the development of powerful research techniques including optogenetics, fiber-photometry, cell-type/pathway specific manipulation and neuronal activity monitoring, as well as single-cell RNA sequencing, the details of how hypocretinergic system execute functional modulation of various behaviors are coming to light. In this review, we focus on the function of neural pathways from hypocretin neurons to target brain regions. Anatomical and functional inputs to hypocretin neurons are also discussed. We further briefly summarize the development of pharmaceutical compounds targeting hypocretin signaling. This article is part of the special issue on Neuropeptides.
Collapse
Affiliation(s)
- Shi-Bin Li
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305, USA.
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305, USA.
| |
Collapse
|
34
|
Negishi K, Payant MA, Schumacker KS, Wittmann G, Butler RM, Lechan RM, Steinbusch HWM, Khan AM, Chee MJ. Distributions of hypothalamic neuron populations coexpressing tyrosine hydroxylase and the vesicular GABA transporter in the mouse. J Comp Neurol 2020; 528:1833-1855. [PMID: 31950494 DOI: 10.1002/cne.24857] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/20/2019] [Accepted: 01/03/2020] [Indexed: 12/21/2022]
Abstract
The hypothalamus contains catecholaminergic neurons marked by the expression of tyrosine hydroxylase (TH). As multiple chemical messengers coexist in each neuron, we determined if hypothalamic TH-immunoreactive (ir) neurons express vesicular glutamate or GABA transporters. We used Cre/loxP recombination to express enhanced GFP (EGFP) in neurons expressing the vesicular glutamate (vGLUT2) or GABA transporter (vGAT), then determined whether TH-ir neurons colocalized with native EGFPVglut2 - or EGFPVgat -fluorescence, respectively. EGFPVglut2 neurons were not TH-ir. However, discrete TH-ir signals colocalized with EGFPVgat neurons, which we validated by in situ hybridization for Vgat mRNA. To contextualize the observed pattern of colocalization between TH-ir and EGFPVgat , we first performed Nissl-based parcellation and plane-of-section analysis, and then mapped the distribution of TH-ir EGFPVgat neurons onto atlas templates from the Allen Reference Atlas (ARA) for the mouse brain. TH-ir EGFPVgat neurons were distributed throughout the rostrocaudal extent of the hypothalamus. Within the ARA ontology of gray matter regions, TH-ir neurons localized primarily to the periventricular hypothalamic zone, periventricular hypothalamic region, and lateral hypothalamic zone. There was a strong presence of EGFPVgat fluorescence in TH-ir neurons across all brain regions, but the most striking colocalization was found in a circumscribed portion of the zona incerta (ZI)-a region assigned to the hypothalamus in the ARA-where every TH-ir neuron expressed EGFPVgat . Neurochemical characterization of these ZI neurons revealed that they display immunoreactivity for dopamine but not dopamine β-hydroxylase. Collectively, these findings indicate the existence of a novel mouse hypothalamic population that may signal through the release of GABA and/or dopamine.
Collapse
Affiliation(s)
- Kenichiro Negishi
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences, and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas
| | - Mikayla A Payant
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Kayla S Schumacker
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Gabor Wittmann
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tufts Medical Center, Boston, Massachusetts
| | - Rebecca M Butler
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Ronald M Lechan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tufts Medical Center, Boston, Massachusetts
| | - Harry W M Steinbusch
- Department of Psychiatry and Neuropsychology, Section Cellular Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Arshad M Khan
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences, and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas
| | - Melissa J Chee
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
35
|
Transgenic Archaerhodopsin-3 Expression in Hypocretin/Orexin Neurons Engenders Cellular Dysfunction and Features of Type 2 Narcolepsy. J Neurosci 2019; 39:9435-9452. [PMID: 31628177 DOI: 10.1523/jneurosci.0311-19.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 02/08/2023] Open
Abstract
Narcolepsy, characterized by excessive daytime sleepiness, is associated with dysfunction of the hypothalamic hypocretin/orexin (Hcrt) system, either due to extensive loss of Hcrt cells (Type 1, NT1) or hypothesized Hcrt signaling impairment (Type 2, NT2). Accordingly, efforts to recapitulate narcolepsy-like symptoms in mice have involved ablating these cells or interrupting Hcrt signaling. Here, we describe orexin/Arch mice, in which a modified archaerhodopsin-3 gene was inserted downstream of the prepro-orexin promoter, resulting in expression of the yellow light-sensitive Arch-3 proton pump specifically within Hcrt neurons. Histological examination along with ex vivo and in vivo electrophysiological recordings of male and female orexin/Arch mice demonstrated silencing of Hcrt neurons when these cells were photoilluminated. However, high expression of the Arch transgene affected cellular and physiological parameters independent of photoillumination. The excitability of Hcrt neurons was reduced, and both circadian and metabolic parameters were perturbed in a subset of orexin/Arch mice that exhibited high levels of Arch expression. Orexin/Arch mice also had increased REM sleep under baseline conditions but did not exhibit cataplexy, a sudden loss of muscle tone during wakefulness characteristic of NT1. These aberrations resembled some aspects of mouse models with Hcrt neuron ablation, yet the number of Hcrt neurons in orexin/Arch mice was not reduced. Thus, orexin/Arch mice may be useful to investigate Hcrt system dysfunction when these neurons are intact, as is thought to occur in narcolepsy without cataplexy (NT2). These results also demonstrate the utility of extended phenotypic screening of transgenic models when specific neural circuits have been manipulated.SIGNIFICANCE STATEMENT Optogenetics has become an invaluable tool for functional dissection of neural circuitry. While opsin expression is often achieved by viral injection, stably integrated transgenes offer some practical advantages. Here, we demonstrate successful transgenic expression of an inhibitory opsin in hypocretin/orexin neurons, which are thought to promote or maintain wakefulness. Both brief and prolonged illumination resulted in inhibition of these neurons and induced sleep. However, even in the absence of illumination, these cells exhibited altered electrical characteristics, particularly when transgene expression was high. These aberrant properties affected metabolism and sleep, resulting in a phenotype reminiscent of the narcolepsy Type 2, a sleep disorder for which no good animal model currently exists.
Collapse
|
36
|
Yin D, Dong H, Wang TX, Hu ZZ, Cheng NN, Qu WM, Huang ZL. Glutamate Activates the Histaminergic Tuberomammillary Nucleus and Increases Wakefulness in Rats. Neuroscience 2019; 413:86-98. [DOI: 10.1016/j.neuroscience.2019.05.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 05/10/2019] [Accepted: 05/17/2019] [Indexed: 01/23/2023]
|
37
|
Kosse C, Burdakov D. Natural hypothalamic circuit dynamics underlying object memorization. Nat Commun 2019; 10:2505. [PMID: 31175285 PMCID: PMC6555780 DOI: 10.1038/s41467-019-10484-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 05/08/2019] [Indexed: 12/23/2022] Open
Abstract
Brain signals that govern memory formation remain incompletely identified. The hypothalamus is implicated in memory disorders, but how its rapidly changing activity shapes memorization is unknown. During encounters with objects, hypothalamic melanin-concentrating hormone (MCH) neurons emit brief signals that reflect object novelty. Here we show that targeted optogenetic silencing of these signals, performed selectively during the initial object encounters (i.e. memory acquisition), prevents future recognition of the objects. We identify an upstream inhibitory microcircuit from hypothalamic GAD65 neurons to MCH neurons, which constrains the memory-promoting MCH cell bursts. Finally, we demonstrate that silencing the GAD65 cells during object memory acquisition improves future object recognition through MCH-receptor-dependent pathways. These results provide causal evidence that object-associated signals in genetically distinct but interconnected hypothalamic neurons differentially control whether the brain forms object memories. This gating of memory formation by hypothalamic activity establishes appropriate behavioral responses to novel and familiar objects.
Collapse
Affiliation(s)
- Christin Kosse
- The Francis Crick Institute, London, NW1 1AT, UK
- Laboratory of Molecular Genetics, The Rockefeller University, New York, NY, 10065, USA
| | - Denis Burdakov
- The Francis Crick Institute, London, NW1 1AT, UK.
- Neurobehavioural Dynamics Lab, Institute for Neuroscience, D-HEST, Swiss Federal Institute of Technology / ETH Zürich, Zürich, 8603, Switzerland.
| |
Collapse
|
38
|
Briggs C, Bowes SC, Semba K, Hirasawa M. Sleep deprivation-induced pre- and postsynaptic modulation of orexin neurons. Neuropharmacology 2018; 154:50-60. [PMID: 30586566 DOI: 10.1016/j.neuropharm.2018.12.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 10/27/2022]
Abstract
Sleep/wake states are controlled by sleep- and wake-promoting systems, and transitions between states are thought to be regulated by their reciprocal inhibition and homeostatic sleep need. Orexin neurons are known to promote wake maintenance and stabilize the sleep/wake switch. Thus, we asked whether orexin neurons are modulated by homeostatic sleep need. Rats were sleep deprived or left undisturbed to rest for 6 h, then acute brain slices were generated for patch clamp recordings. We found that sleep deprivation increased firing and reduced spike frequency adaptation in response to excitatory drive in orexin neurons. These changes were specific to D-type orexin neurons which, unlike H-type orexin neurons, lack A-type current. In D-type orexin neurons, sleep deprivation decreased afterhyperpolarizing potential, which was associated with increased gain, measured as the slope of the input-output relationship. These effects were mimicked by inhibition of SK channels. Furthermore, sleep deprivation resulted in presynaptic inhibition of excitatory inputs to both D-type and H-type orexin neurons, which preferentially affected sparse synaptic inputs while sparing high frequency synaptic activities. Taken together, our results indicate that sleep deprivation modulates the gain control and synaptic gating in orexin neurons. These pre-and postsynaptic changes would tune orexin neurons to strong wake-promoting excitatory signals, while dampening weak synaptic inputs to allow transition to sleep in the absence of such strong signals. These mechanisms are consistent with a role of orexin neurons not only as a key state stabilizer, but also as a homeostatic wake integrator in the sleep/wake switch. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.
Collapse
Affiliation(s)
- Chantalle Briggs
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada; Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, 5850 College Street, Halifax, NS, B3H 4R2, Canada
| | - Sherri C Bowes
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada
| | - Kazue Semba
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, 5850 College Street, Halifax, NS, B3H 4R2, Canada; Department of Psychiatry, Faculty of Medicine, Dalhousie University, 5909 Veterans' Memorial Lane, Halifax, NS, B3H 2E2, Canada; Department of Psychology and Neuroscience, Faculty of Science, Dalhousie University, 1355 Oxford Street, Halifax, NS, B3H 4R2, Canada
| | - Michiru Hirasawa
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL, A1B 3V6, Canada.
| |
Collapse
|
39
|
Arrigoni E, Chee MJS, Fuller PM. To eat or to sleep: That is a lateral hypothalamic question. Neuropharmacology 2018; 154:34-49. [PMID: 30503993 DOI: 10.1016/j.neuropharm.2018.11.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 12/15/2022]
Abstract
The lateral hypothalamus (LH) is a functionally and anatomically complex brain region that is involved in the regulation of many behavioral and physiological processes including feeding, arousal, energy balance, stress, reward and motivated behaviors, pain perception, body temperature regulation, digestive functions and blood pressure. Despite noteworthy experimental efforts over the past decades, the circuit, cellular and synaptic bases by which these different processes are regulated by the LH remains incompletely understood. This knowledge gap links in large part to the high cellular heterogeneity of the LH. Fortunately, the rapid evolution of newer genetic and electrophysiological tools is now permitting the selective manipulation, typically genetically-driven, of discrete LH cell populations. This, in turn, permits not only assignment of function to discrete cell groups, but also reveals that considerable synergistic and antagonistic interactions exist between key LH cell populations that regulate feeding and arousal. For example, we now know that while LH melanin-concentrating hormone (MCH) and orexin/hypocretin neurons both function as sensors of the internal metabolic environment, their roles regulating sleep and arousal are actually opposing. Additional studies have uncovered similarly important roles for subpopulations of LH GABAergic cells in the regulation of both feeding and arousal. Herein we review the role of LH MCH, orexin/hypocretin and GABAergic cell populations in the regulation of energy homeostasis (including feeding) and sleep-wake and discuss how these three cell populations, and their subpopulations, may interact to optimize and coordinate metabolism, sleep and arousal. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.
Collapse
Affiliation(s)
- Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02215, USA.
| | - Melissa J S Chee
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | - Patrick M Fuller
- Department of Neurology, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02215, USA
| |
Collapse
|
40
|
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: 2.4] [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
|
41
|
Burdakov D. Reactive and predictive homeostasis: Roles of orexin/hypocretin neurons. Neuropharmacology 2018; 154:61-67. [PMID: 30347195 DOI: 10.1016/j.neuropharm.2018.10.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/14/2018] [Accepted: 10/16/2018] [Indexed: 11/30/2022]
Abstract
Homeostasis is the maintenance of a healthy physiological equilibrium in a changing world. Reactive (feedback, counter-regulatory) and predictive (feedforward, anticipatory) homeostatic control strategies are both important for survival. For example, in energy homeostasis, the pancreas reacts to ingested glucose by releasing insulin, whereas the brain prepares the body for ingestion through anticipatory salivation based on food-associated cues. Reactive control is largely innate, whereas predictive control is often acquired or modified through associative learning, though some important predictive control strategies are innate, e.g. avoidance of fox scent in mice that never met a fox. Traditionally, the hypothalamus has been viewed as a reactive controller, sensing deviations from homeostasis to elicit counter-regulatory responses, while "higher" areas such as the cortex have been viewed as predictive controllers. However, experimental evidence argues against such neuroanatomical segregation: for example, receptors for internal homeostatic indicators are found throughout the brain, while key interoceptive hypothalamic cells also rapidly sense external cues. Here a model is proposed where the brain-wide-projecting, non-neuroendocrine, neurons of the hypothalamus, exemplified by orexin/hypocretin neurons, function as "brain government" systems that convert integrated internal and external information into reactive and predictive autonomic, cognitive, and behavioural adaptations that ensure homeostasis. Like regions of a country without a government, individual brain regions can function normally without hypothalamic guidance, but these functions are uncoordinated, producing mismatch between supply and demand of arousal, and derailing decision-making as seen in orexin-deficient narcolepsy. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.
Collapse
Affiliation(s)
- Denis Burdakov
- Swiss Federal Institute of Technology / ETH Zürich, D-HEST, Institute for Neuroscience, Schorenstrasse 16, Schwerzenbach 8603, Switzerland.
| |
Collapse
|
42
|
Liu JJ, Mirabella VR, Pang ZP. Cell type- and pathway-specific synaptic regulation of orexin neurocircuitry. Brain Res 2018; 1731:145974. [PMID: 30296428 DOI: 10.1016/j.brainres.2018.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 12/19/2022]
Abstract
Orexin-expressing neurons are located exclusively in the lateral hypothalamic and perifornical areas and exhibit complex connectivity. The intricate wiring pattern is evident from a diverse function for orexin neurons in regulating many physiological processes and behaviors including sleep, metabolism, circadian cycles, anxiety, and reward. Nevertheless, the precise synaptic and circuitry-level mechanisms mediating these processes remain enigmatic, partially due to the wide spread connectivity of the orexin system, complex neurochemistry of orexin neurons, and previous lack of suitable tools to address its complexity. Here we summarize recent advances, focusing on synaptic regulatory mechanisms in the orexin neurocircuitry, including both the synaptic inputs to orexin neurons as well as their downstream targets in the brain. A clear and detailed elucidation of these mechanisms will likely provide novel insight into how dysfunction in orexin-mediated signaling leads to human disease and may ultimately be treated with more precise strategies.
Collapse
Affiliation(s)
- Jing-Jing Liu
- Child Health Institute of New Jersey, Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
| | - Vincent R Mirabella
- Child Health Institute of New Jersey, Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Zhiping P Pang
- Child Health Institute of New Jersey, Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| |
Collapse
|
43
|
Li SB, Nevárez N, Giardino WJ, de Lecea L. Optical probing of orexin/hypocretin receptor antagonists. Sleep 2018; 41:5060288. [PMID: 30060151 PMCID: PMC6454482 DOI: 10.1093/sleep/zsy141] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/10/2018] [Indexed: 01/17/2023] Open
Abstract
Study Objectives The present study investigated the function of Hypocretin (Hcrt or Orexin/OX) receptor antagonists in sleep modulation and memory function with optical methods in transgenic mice. Methods We used Hcrt-IRES-Cre knock-in mice and AAV vectors expressing channelrhodopsin-2 (ChR2) to render Hcrt neurons sensitive to blue light stimulation. We optogenetically stimulated Hcrt neurons and measured latencies to wakefulness in the presence or absence of OX1/2R antagonists and Zolpidem. We also examined endogenous Hcrt neuronal activity with fiber photometry. Changes in memory after optogenetic sleep disruption were evaluated by the novel object recognition test (NOR) and compared for groups treated with vehicle, OX1/2R antagonists, or Zolpidem. We also analyzed electroencephalogram (EEG) power spectra of wakefulness, rapid eye movement (REM) sleep, and non-REM (NREM) sleep following the injections of vehicle, OX1/2R antagonists, and Zolpidem in young adult mice. Results Acute optogenetic stimulation of Hcrt neurons at different frequencies resulted in wakefulness. Treatment with dual OX1/2R antagonists (DORAs) DORA12 and MK6096, as well as selective OX2R antagonist MK1064 and Zolpidem, but not selective OX1R antagonist 1SORA1, significantly reduced the bout length of optogenetic stimulation-evoked wakefulness episode. Fiber photometry recordings of GCaMP6f signals showed that Hcrt neurons are active during wakefulness, even in the presence of OXR antagonists. Treatment with dual OX1/2R antagonists improved memory function despite optogenetic sleep fragmentation caused impaired memory function in a NOR test. Conclusions Our results show DORAs and selective OX2R antagonists stabilize sleep and improve sleep-dependent cognitive processes even when challenged by optogenetic stimulation mimicking highly arousing stimuli.
Collapse
Affiliation(s)
- Shi-Bin Li
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| | - Natalie Nevárez
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| | - William J Giardino
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
44
|
Azeez IA, Del Gallo F, Cristino L, Bentivoglio M. Daily Fluctuation of Orexin Neuron Activity and Wiring: The Challenge of "Chronoconnectivity". Front Pharmacol 2018; 9:1061. [PMID: 30319410 PMCID: PMC6167434 DOI: 10.3389/fphar.2018.01061] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022] Open
Abstract
In the heterogeneous hub represented by the lateral hypothalamus, neurons containing the orexin/hypocretin peptides play a key role in vigilance state transitions and wakefulness stability, energy homeostasis, and other functions relevant for motivated behaviors. Orexin neurons, which project widely to the neuraxis, are innervated by multiple extra- and intra-hypothalamic sources. A key property of the adaptive capacity of orexin neurons is represented by daily variations of activity, which is highest in the period of the animal’s activity and wakefulness. These sets of data are here reviewed. They concern the discharge profile during the sleep/wake cycle, spontaneous Fos induction, peptide synthesis and release reflected by immunostaining intensity and peptide levels in the cerebrospinal fluid as well as postsynaptic effects. At the synaptic level, adaptive capacity of orexin neurons subserved by remodeling of excitatory and inhibitory inputs has been shown in response to changes in the nutritional status and prolonged wakefulness. The present review wishes to highlight that synaptic plasticity in the wiring of orexin neurons also occurs in unperturbed conditions and could account for diurnal variations of orexin neuron activity. Data in zebrafish larvae have shown rhythmic changes in the density of inhibitory innervation of orexin dendrites in relation to vigilance states. Recent findings in mice have indicated a diurnal reorganization of the excitatory/inhibitory balance in the perisomatic innervation of orexin neurons. Taken together these sets of data point to “chronoconnectivity,” i.e., a synaptic rearrangement of inputs to orexin neurons over the course of the day in relation to sleep and wake states. This opens questions on the underlying circadian and homeostatic regulation and on the involved players at synaptic level, which could implicate dual transmitters, cytoskeletal rearrangements, hormonal regulation, as well as surrounding glial cells and extracellular matrix. Furthermore, the question arises of a “chronoconnectivity” in the wiring of other neuronal cell groups of the sleep-wake-regulatory network, many of which are characterized by variations of their firing rate during vigilance states.
Collapse
Affiliation(s)
- Idris A Azeez
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Federico Del Gallo
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | | | - Marina Bentivoglio
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.,National Institute of Neuroscience, Verona Unit, Verona, Italy
| |
Collapse
|
45
|
Parallel Arousal Pathways in the Lateral Hypothalamus. eNeuro 2018; 5:eN-NWR-0228-18. [PMID: 30225361 PMCID: PMC6140123 DOI: 10.1523/eneuro.0228-18.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/11/2018] [Accepted: 07/12/2018] [Indexed: 11/29/2022] Open
Abstract
Until recently, hypocretin (Hcrt) neurons were the only known wake-promoting neuronal population in the lateral hypothalamus (LH), but subpopulations of inhibitory neurons in this area and glutamatergic neurons in the nearby supramammillary nucleus (SuM) have recently been found that also promote wakefulness. We performed chemogenetic excitation of LH neurons in mice and observed increased wakefulness that lasted more than 4 h without unusual behavior or EEG anomalies. The increased wakefulness was similar in the presence or absence of the dual orexin receptor blocker almorexant (ALM). Analysis of hM3Dq transfection and c-FOS expression in LH inhibitory neurons and in the SuM failed to confirm that the increased wakefulness was due to these wake-promoting populations, although this possibility cannot be completely excluded. To evaluate the relationship to the Hcrt system, we repeated the study in Orexin-tTA mice in the presence or absence of dietary doxycycline (DOX), which enabled us to manipulate the percentage of Hcrt neurons that expressed hM3Dq. In DOX-fed mice, 18% of Hcrt neurons as well as many other LH neurons expressed hM3Dq; these mice showed a profound increase in wake after hM3Dq activation even in the presence of ALM. In mice switched to normal chow, 62% of Hcrt neurons expressed hM3Dq along with other LH cells; chemogenetic activation produced even more sustained arousal which could be reduced to previous levels by ALM treatment. Together, these results indicate an LH neuron population that promotes wakefulness through an Hcrt-independent pathway that can act synergistically with the Hcrt system to prolong arousal.
Collapse
|
46
|
Takács VT, Cserép C, Schlingloff D, Pósfai B, Szőnyi A, Sos KE, Környei Z, Dénes Á, Gulyás AI, Freund TF, Nyiri G. Co-transmission of acetylcholine and GABA regulates hippocampal states. Nat Commun 2018; 9:2848. [PMID: 30030438 PMCID: PMC6054650 DOI: 10.1038/s41467-018-05136-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 06/12/2018] [Indexed: 12/13/2022] Open
Abstract
The basal forebrain cholinergic system is widely assumed to control cortical functions via non-synaptic transmission of a single neurotransmitter. Yet, we find that mouse hippocampal cholinergic terminals invariably establish GABAergic synapses, and their cholinergic vesicles dock at those synapses only. We demonstrate that these synapses do not co-release but co-transmit GABA and acetylcholine via different vesicles, whose release is triggered by distinct calcium channels. This co-transmission evokes composite postsynaptic potentials, which are mutually cross-regulated by presynaptic autoreceptors. Although postsynaptic cholinergic receptor distribution cannot be investigated, their response latencies suggest a focal, intra- and/or peri-synaptic localisation, while GABAA receptors are detected intra-synaptically. The GABAergic component alone effectively suppresses hippocampal sharp wave-ripples and epileptiform activity. Therefore, the differentially regulated GABAergic and cholinergic co-transmission suggests a hitherto unrecognised level of control over cortical states. This novel model of hippocampal cholinergic neurotransmission may lead to alternative pharmacotherapies after cholinergic deinnervation seen in neurodegenerative disorders.
Collapse
Affiliation(s)
- Virág T Takács
- Laboratory of Cerebral Cortex Research Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u 43, Budapest, 1083, Hungary
| | - Csaba Cserép
- Laboratory of Cerebral Cortex Research Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u 43, Budapest, 1083, Hungary.,Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u 43, Budapest, 1083, Hungary
| | - Dániel Schlingloff
- Laboratory of Cerebral Cortex Research Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u 43, Budapest, 1083, Hungary.,János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, 1085, Hungary
| | - Balázs Pósfai
- Laboratory of Cerebral Cortex Research Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u 43, Budapest, 1083, Hungary
| | - András Szőnyi
- Laboratory of Cerebral Cortex Research Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u 43, Budapest, 1083, Hungary.,János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, 1085, Hungary
| | - Katalin E Sos
- Laboratory of Cerebral Cortex Research Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u 43, Budapest, 1083, Hungary.,János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, 1085, Hungary
| | - Zsuzsanna Környei
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u 43, Budapest, 1083, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u 43, Budapest, 1083, Hungary
| | - Attila I Gulyás
- Laboratory of Cerebral Cortex Research Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u 43, Budapest, 1083, Hungary
| | - Tamás F Freund
- Laboratory of Cerebral Cortex Research Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u 43, Budapest, 1083, Hungary
| | - Gábor Nyiri
- Laboratory of Cerebral Cortex Research Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u 43, Budapest, 1083, Hungary.
| |
Collapse
|
47
|
Tyree SM, Borniger JC, de Lecea L. Hypocretin as a Hub for Arousal and Motivation. Front Neurol 2018; 9:413. [PMID: 29928253 PMCID: PMC5997825 DOI: 10.3389/fneur.2018.00413] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/18/2018] [Indexed: 01/01/2023] Open
Abstract
The lateral hypothalamus is comprised of a heterogeneous mix of neurons that serve to integrate and regulate sleep, feeding, stress, energy balance, reward, and motivated behavior. Within these populations, the hypocretin/orexin neurons are among the most well studied. Here, we provide an overview on how these neurons act as a central hub integrating sensory and physiological information to tune arousal and motivated behavior accordingly. We give special attention to their role in sleep-wake states and conditions of hyper-arousal, as is the case with stress-induced anxiety. We further discuss their roles in feeding, drug-seeking, and sexual behavior, which are all dependent on the motivational state of the animal. We further emphasize the application of powerful techniques, such as optogenetics, chemogenetics, and fiber photometry, to delineate the role these neurons play in lateral hypothalamic functions.
Collapse
Affiliation(s)
- Susan M Tyree
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Jeremy C Borniger
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| |
Collapse
|
48
|
Moorman DE. The hypocretin/orexin system as a target for excessive motivation in alcohol use disorders. Psychopharmacology (Berl) 2018; 235:1663-1680. [PMID: 29508004 PMCID: PMC5949267 DOI: 10.1007/s00213-018-4871-2] [Citation(s) in RCA: 17] [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] [Received: 10/30/2017] [Accepted: 02/20/2018] [Indexed: 12/17/2022]
Abstract
The hypocretin/orexin (ORX) system has been repeatedly demonstrated to regulate motivation for drugs of abuse, including alcohol. In particular, ORX seems to be critically involved in highly motivated behaviors, as is observed in high-seeking individuals in a population, in the seeking of highly palatable substances, and in models of dependence. It seems logical that this system could be considered as a potential target for treatment for addiction, particularly alcohol addiction, as ORX pharmacological manipulations significantly reduce drinking. However, the ORX system also plays a role in a wide range of other behaviors, emotions, and physiological functions and is disrupted in a number of non-dependence-associated disorders. It is therefore important to consider how the ORX system might be optimally targeted for potential treatment for alcohol use disorders either in combination with or separate from its role in other functions or diseases. This review will focus on the role of ORX in alcohol-associated behaviors and whether and how this system could be targeted to treat alcohol use disorders while avoiding impacts on other ORX-relevant functions. A brief overview of the ORX system will be followed by a discussion of some of the factors that makes it particularly intriguing as a target for alcohol addiction treatment, a consideration of some potential challenges associated with targeting this system and, finally, some future directions to optimize new treatments.
Collapse
Affiliation(s)
- David E Moorman
- Department of Psychological and Brain Sciences, Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, 528 Tobin Hall, 135 Hicks Way, Amherst, MA, 01003, USA.
| |
Collapse
|
49
|
Schwartz MD, Palmerston JB, Lee DL, Hoener MC, Kilduff TS. Deletion of Trace Amine-Associated Receptor 1 Attenuates Behavioral Responses to Caffeine. Front Pharmacol 2018; 9:35. [PMID: 29456505 PMCID: PMC5801540 DOI: 10.3389/fphar.2018.00035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/12/2018] [Indexed: 12/18/2022] Open
Abstract
Trace amines (TAs), endogenous amino acid metabolites that are structurally similar to the biogenic amines, are endogenous ligands for trace amine-associated receptor 1 (TAAR1), a GPCR that modulates dopaminergic, serotonergic, and glutamatergic activity. Selective TAAR1 full and partial agonists exhibit similar pro-cognitive, antidepressant- and antipsychotic-like properties in rodents and non-human primates, suggesting TAAR1 as a novel target for the treatment of neurological and psychiatric disorders. We previously reported that TAAR1 partial agonists are wake-promoting in rats and mice, and that TAAR1 knockout (KO) and overexpressing mice exhibit altered sleep-wake and EEG spectral composition. Here, we report that locomotor and EEG spectral responses to the psychostimulants modafinil and caffeine are attenuated in TAAR1 KO mice. TAAR1 KO mice and WT littermates were instrumented for EEG and EMG recording and implanted with telemetry transmitters for monitoring locomotor activity (LMA) and core body temperature (Tb). Following recovery, mice were administered modafinil (25, 50, 100 mg/kg), caffeine (2.5, 10, 20 mg/kg) or vehicle p.o. at ZT6 in balanced order. In WT mice, both modafinil and caffeine dose-dependently increased LMA for up to 6 h following dosing, whereas only the highest dose of each drug increased LMA in KO mice, and did so for less time after dosing. This effect was particularly pronounced following caffeine, such that total LMA response was significantly attenuated in KO mice compared to WT at all doses of caffeine and did not differ from Vehicle treatment. Tb increased comparably in both genotypes in a dose-dependent manner. TAAR1 deletion was associated with reduced wake consolidation following both drugs, but total time in wakefulness did not differ between KO and WT mice. Furthermore, gamma band EEG activity following both modafinil and caffeine treatment was attenuated in TAAR1 KO compared to WT mice. Our results show that TAAR1 is a critical component of the behavioral and cortical arousal associated with two widely used psychostimulants with very different mechanisms of action. Together with our previous findings, these data suggest that TAAR1 is a previously unrecognized component of an endogenous wake-modulating system.
Collapse
Affiliation(s)
- Michael D Schwartz
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA, United States
| | - Jeremiah B Palmerston
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA, United States
| | - Diana L Lee
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA, United States
| | - Marius C Hoener
- Neuroscience, Ophthalmology and Rare Diseases Discovery and Translational Area, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Ltd., Basel, Switzerland
| | - Thomas S Kilduff
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA, United States
| |
Collapse
|
50
|
Alvarez B, Barrientos T, Gac L, Teske J, Perez-Leighton C. Effects on Hedonic Feeding, Energy Expenditure and Balance of the Non-opioid Peptide DYN-A2-17. Neuroscience 2018; 371:337-345. [DOI: 10.1016/j.neuroscience.2017.11.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 11/11/2017] [Accepted: 11/27/2017] [Indexed: 11/28/2022]
|