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
|
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
|
3
|
Li Z, Zheng L, Wang J, Wang L, Qi Y, Amin B, Zhu J, Zhang N. Dopamine in the regulation of glucose and lipid metabolism: a narrative review. Obesity (Silver Spring) 2024; 32:1632-1645. [PMID: 39081007 DOI: 10.1002/oby.24068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVE Owing to the global obesity epidemic, understanding the regulatory mechanisms of glucose and lipid metabolism has become increasingly important. The dopaminergic system, including dopamine, dopamine receptors, dopamine transporters, and other components, is involved in numerous physiological and pathological processes. However, the mechanism of action of the dopaminergic system in glucose and lipid metabolism is poorly understood. In this review, we examine the role of the dopaminergic system in glucose and lipid metabolism. RESULTS The dopaminergic system regulates glucose and lipid metabolism through several mechanisms. It regulates various activities at the central level, including appetite control and decision-making, which contribute to regulating body weight and energy metabolism. In the pituitary gland, dopamine inhibits prolactin production and promotes insulin secretion through dopamine receptor 2. Furthermore, it can influence various physiological components in the peripheral system, such as pancreatic β cells, glucagon-like peptide-1, adipocytes, hepatocytes, and muscle, by regulating insulin and glucagon secretion, glucose uptake and use, and fatty acid metabolism. CONCLUSIONS The role of dopamine in regulating glucose and lipid metabolism has significant implications for the physiology and pathogenesis of disease. The potential therapeutic value of dopamine lies in its effects on metabolic disorders.
Collapse
Affiliation(s)
- Zhehong Li
- Surgery Centre of Diabetes Mellitus, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Lifei Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jing Wang
- Surgery Centre of Diabetes Mellitus, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Liang Wang
- Surgery Centre of Diabetes Mellitus, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yao Qi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Buhe Amin
- Surgery Centre of Diabetes Mellitus, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jinxia Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Nengwei Zhang
- Surgery Centre of Diabetes Mellitus, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Payant MA, Spencer CD, Ly NKK, Chee MJ. Inhibitory actions of melanin-concentrating hormone in the lateral septum. J Physiol 2024; 602:3545-3574. [PMID: 38874572 DOI: 10.1113/jp284845] [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: 04/11/2023] [Accepted: 05/21/2024] [Indexed: 06/15/2024] Open
Abstract
Melanin-concentrating hormone (MCH) neurons can co-express several neuropeptides or neurotransmitters and send widespread projections throughout the brain. Notably, there is a dense cluster of nerve terminals from MCH neurons in the lateral septum (LS) that innervate LS cells by glutamate release. The LS is also a key region integrating stress- and anxiety-like behaviours, which are also emerging roles of MCH neurons. However, it is not known if or where the MCH peptide acts within the LS. We analysed the projections from MCH neurons in male and female mice anteroposteriorly throughout the LS and found spatial overlap between the distribution pattern of MCH-immunoreactive (MCH-ir) fibres with MCH receptor Mchr1 mRNA hybridization or MCHR1-ir cells. This overlap was most prominent along the ventral and lateral border of the rostral part of the LS (LSr). Most MCHR1-labelled LS neurons lay adjacent to passing MCH-ir fibres, but some MCH-ir varicosities directly contacted the soma or cilium of MCHR1-labelled LS neurons. We thus performed whole-cell patch-clamp recordings from MCHR1-rich LSr regions to determine if and how LS cells respond to MCH. Bath application of MCH to acute brain slices activated a bicuculline-sensitive chloride current that directly hyperpolarized LS cells. This MCH-mediated hyperpolarization was blocked by calphostin C, which suggested that the inhibitory actions of MCH were mediated by protein kinase C-dependent activation of GABAA receptors. Taken together, these findings define potential hotspots within the LS that may elucidate the contributions of MCH to stress- or anxiety-related feeding behaviours. KEY POINTS: Melanin-concentrating hormone (MCH) neurons have dense nerve terminals within the lateral septum (LS), a key region underlying stress- and anxiety-like behaviours that are emerging roles of the MCH system, but the function of MCH in the LS is not known. We found spatial overlap between MCH-immunoreactive fibres, Mchr1 mRNA, and MCHR1 protein expression along the lateral border of the LS. Within MCHR1-rich regions, MCH directly inhibited LS cells by increasing chloride conductance via GABAA receptor activation in a protein kinase C-dependent manner. Electrophysiological MCH effects in brain slices have been elusive, and few studies have described the mechanisms of MCH action. Our findings demonstrated, to our knowledge, the first description of MCHR1 Gq-coupling in brain slices, which was previously predicted in cell or primary culture models only. Together, these findings defined hotspots and mechanistic underpinnings for MCH effects such as in feeding and anxiety-related behaviours.
Collapse
Affiliation(s)
- Mikayla A Payant
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - C Duncan Spencer
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Nikita K Koziel Ly
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Melissa J Chee
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
5
|
Concetti C, Peleg-Raibstein D, Burdakov D. Hypothalamic MCH Neurons: From Feeding to Cognitive Control. FUNCTION 2023; 5:zqad059. [PMID: 38020069 PMCID: PMC10667013 DOI: 10.1093/function/zqad059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Modern neuroscience is progressively elucidating that the classic view positing distinct brain regions responsible for survival, emotion, and cognitive functions is outdated. The hypothalamus demonstrates the interdependence of these roles, as it is traditionally known for fundamental survival functions like energy and electrolyte balance, but is now recognized to also play a crucial role in emotional and cognitive processes. This review focuses on lateral hypothalamic melanin-concentrating hormone (MCH) neurons, producing the neuropeptide MCH-a relatively understudied neuronal population with integrative functions related to homeostatic regulation and motivated behaviors, with widespread inputs and outputs throughout the entire central nervous system. Here, we review early findings and recent literature outlining their role in the regulation of energy balance, sleep, learning, and memory processes.
Collapse
Affiliation(s)
- Cristina Concetti
- Neurobehavioural Dynamics Laboratory, ETH Zürich, Schorenstrasse 16, Schwerzenbach 8603, Switzerland
| | - Daria Peleg-Raibstein
- Neurobehavioural Dynamics Laboratory, ETH Zürich, Schorenstrasse 16, Schwerzenbach 8603, Switzerland
| | - Denis Burdakov
- Neurobehavioural Dynamics Laboratory, ETH Zürich, Schorenstrasse 16, Schwerzenbach 8603, Switzerland
| |
Collapse
|
6
|
Somach RT, Jean ID, Farrugia AM, Cohen AS. Mild Traumatic Brain Injury Affects Orexin/Hypocretin Physiology Differently in Male and Female Mice. J Neurotrauma 2023; 40:2146-2163. [PMID: 37476962 PMCID: PMC10701510 DOI: 10.1089/neu.2023.0125] [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] [Indexed: 07/22/2023] Open
Abstract
Traumatic brain injury (TBI) is known to affect the physiology of neural circuits in several brain regions, which can contribute to behavioral changes after injury. Disordered sleep is a behavior that is often seen after TBI, but there is little research into how injury affects the circuitry that contributes to disrupted sleep regulation. Orexin/hypocretin neurons (hereafter referred to as orexin neurons) located in the lateral hypothalamus normally stabilize wakefulness in healthy animals and have been suggested as a source of dysregulated sleep behavior. Despite this, few studies have examined how TBI affects orexin neuron circuitry. Further, almost no animal studies of orexin neurons after TBI have included female animals. Here, we address these gaps by studying changes to orexin physiology using ex vivo acute brain slices and whole-cell patch clamp recording. We hypothesized that orexin neurons would have reduced afferent excitatory activity after injury. Ultimately, this hypothesis was supported but there were additional physiological changes that occurred that we did not originally hypothesize. We studied physiological properties in orexin neurons approximately 1 week after mild traumatic brain injury (mTBI) in 6-8-week-old male and female mice. mTBI was performed with a lateral fluid percussion injury between 1.4 and 1.6 atmospheres. Mild TBI increased the size of action potential afterhyperpolarization in orexin neurons from female mice, but not male mice and reduced the action potential threshold in male mice, but not in female mice. Mild TBI reduced afferent excitatory activity and increased afferent inhibitory activity onto orexin neurons. Alterations in afferent excitatory activity occurred in different parameters in male and female animals. The increased afferent inhibitory activity after injury is more pronounced in recordings from female animals. Our results indicate that mTBI changes the physiology of orexin neuron circuitry and that these changes are not the same in male and female animals.
Collapse
Affiliation(s)
- Rebecca T. Somach
- Department of Anesthesiology and Critical Care Medicine, the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ian D. Jean
- Department of Anesthesiology and Critical Care Medicine, the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Anthony M. Farrugia
- Department of Anesthesiology and Critical Care Medicine, the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Akiva S. Cohen
- Department of Anesthesiology and Critical Care Medicine, the Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Guo R, Vaughan DT, Rojo ALA, Huang YH. Sleep-mediated regulation of reward circuits: implications in substance use disorders. Neuropsychopharmacology 2023; 48:61-78. [PMID: 35710601 PMCID: PMC9700806 DOI: 10.1038/s41386-022-01356-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 12/11/2022]
Abstract
Our modern society suffers from both pervasive sleep loss and substance abuse-what may be the indications for sleep on substance use disorders (SUDs), and could sleep contribute to the individual variations in SUDs? Decades of research in sleep as well as in motivated behaviors have laid the foundation for us to begin to answer these questions. This review is intended to critically summarize the circuit, cellular, and molecular mechanisms by which sleep influences reward function, and to reveal critical challenges for future studies. The review also suggests that improving sleep quality may serve as complementary therapeutics for treating SUDs, and that formulating sleep metrics may be useful for predicting individual susceptibility to SUDs and other reward-associated psychiatric diseases.
Collapse
Affiliation(s)
- Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Allen Institute, Seattle, WA, 98109, USA
| | - Dylan Thomas Vaughan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- The Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Ana Lourdes Almeida Rojo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- The Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Yanhua H Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
- The Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
8
|
Guo R, Wang Y, Yan R, Chen B, Ding W, Gorczyca MT, Ozsoy S, Cai L, Hines RL, Tseng GC, Allocca G, Dong Y, Fang J, Huang YH. Rapid Eye Movement Sleep Engages Melanin-Concentrating Hormone Neurons to Reduce Cocaine Seeking. Biol Psychiatry 2022; 92:880-894. [PMID: 35953320 PMCID: PMC9872495 DOI: 10.1016/j.biopsych.2022.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Persistent sleep disruptions following withdrawal from abused drugs may hold keys to battle drug relapse. It is posited that there may be sleep signatures that predict relapse propensity, identifying which may open new avenues for treating substance use disorders. METHODS We trained male rats (approximately postnatal day 56) to self-administer cocaine. After long-term drug withdrawal (approximately postnatal day 100), we examined the correlations between the intensity of cocaine seeking and key sleep features. To test for causal relationships, we then used behavioral, chemogenetic, or optogenetic methods to selectively increase rapid eye movement sleep (REMS) and measured behavioral and electrophysiological outcomes to probe for cellular and circuit mechanisms underlying REMS-mediated regulation of cocaine seeking. RESULTS A selective set of REMS features was preferentially associated with the intensity of cue-induced cocaine seeking after drug withdrawal. Moreover, selectively increasing REMS time and continuity by environmental warming attenuated a withdrawal time-dependent intensification of cocaine seeking, or incubation of cocaine craving, suggesting that REMS may benefit withdrawal. Warming increased the activity of lateral hypothalamic melanin-concentrating hormone (MCH) neurons selectively during prolonged REMS episodes and counteracted cocaine-induced synaptic accumulation of calcium-permeable AMPA receptors in the nucleus accumbens-a critical substrate for incubation. Finally, the warming effects were partly mimicked by chemogenetic or optogenetic stimulations of MCH neurons during sleep, or intra-accumbens infusions of MCH peptide during the rat's inactive phase. CONCLUSIONS REMS may encode individual vulnerability to relapse, and MCH neuron activities can be selectively targeted during REMS to reduce drug relapse.
Collapse
Affiliation(s)
- Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yao Wang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rongzhen Yan
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bo Chen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wanqiao Ding
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael T Gorczyca
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sahin Ozsoy
- Somnivore Pty. Ltd., Bacchus Marsh, Victoria, Australia
| | - Li Cai
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rachel L Hines
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Giancarlo Allocca
- Somnivore Pty. Ltd., Bacchus Marsh, Victoria, Australia; Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Yan Dong
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jidong Fang
- Department of Psychiatry and Behavioral Health, Penn State College of Medicine, Hershey, Pennsylvania
| | - Yanhua H Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
9
|
Liu J, Lai F, Hou Y, Zheng R. Leptin signaling and leptin resistance. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:363-384. [PMID: 37724323 PMCID: PMC10388810 DOI: 10.1515/mr-2022-0017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/12/2022] [Indexed: 09/20/2023]
Abstract
With the prevalence of obesity and associated comorbidities, studies aimed at revealing mechanisms that regulate energy homeostasis have gained increasing interest. In 1994, the cloning of leptin was a milestone in metabolic research. As an adipocytokine, leptin governs food intake and energy homeostasis through leptin receptors (LepR) in the brain. The failure of increased leptin levels to suppress feeding and elevate energy expenditure is referred to as leptin resistance, which encompasses complex pathophysiological processes. Within the brain, LepR-expressing neurons are distributed in hypothalamus and other brain areas, and each population of the LepR-expressing neurons may mediate particular aspects of leptin effects. In LepR-expressing neurons, the binding of leptin to LepR initiates multiple signaling cascades including janus kinase (JAK)-signal transducers and activators of transcription (STAT) phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT), extracellular regulated protein kinase (ERK), and AMP-activated protein kinase (AMPK) signaling, etc., mediating leptin actions. These findings place leptin at the intersection of metabolic and neuroendocrine regulations, and render leptin a key target for treating obesity and associated comorbidities. This review highlights the main discoveries that shaped the field of leptin for better understanding of the mechanism governing metabolic homeostasis, and guides the development of safe and effective interventions to treat obesity and associated diseases.
Collapse
Affiliation(s)
- Jiarui Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Futing Lai
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Yujia Hou
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
- Key Laboratory for Neuroscience of Ministry of Education, Peking University, Beijing, China
- Key Laboratory for Neuroscience of National Health Commission, Peking University, Beijing 100191, China
| |
Collapse
|
10
|
Gao XB, Horvath TL. From Molecule to Behavior: Hypocretin/orexin Revisited From a Sex-dependent Perspective. Endocr Rev 2022; 43:743-760. [PMID: 34792130 PMCID: PMC9277634 DOI: 10.1210/endrev/bnab042] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Indexed: 11/19/2022]
Abstract
The hypocretin/orexin (Hcrt/Orx) system in the perifornical lateral hypothalamus has been recognized as a critical node in a complex network of neuronal systems controlling both physiology and behavior in vertebrates. Our understanding of the Hcrt/Orx system and its array of functions and actions has grown exponentially in merely 2 decades. This review will examine the latest progress in discerning the roles played by the Hcrt/Orx system in regulating homeostatic functions and in executing instinctive and learned behaviors. Furthermore, the gaps that currently exist in our knowledge of sex-related differences in this field of study are discussed.
Collapse
Affiliation(s)
- Xiao-Bing Gao
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
11
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
12
|
Linehan V, Hirasawa M. Short-term fasting induces alternate activation of orexin and melanin-concentrating hormone neurons in rats. Neuroscience 2022; 491:156-165. [DOI: 10.1016/j.neuroscience.2022.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/02/2022] [Accepted: 04/04/2022] [Indexed: 11/27/2022]
|
13
|
Characterization of Hypothalamic MCH Neuron Development in a 3D Differentiation System of Mouse Embryonic Stem Cells. eNeuro 2022; 9:ENEURO.0442-21.2022. [PMID: 35437265 PMCID: PMC9047030 DOI: 10.1523/eneuro.0442-21.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 01/20/2023] Open
Abstract
Hypothalamic melanin-concentrating hormone (MCH) neurons are important regulators of multiple physiological processes, such as sleep, feeding, and memory. Despite the increasing interest in their neuronal functions, the molecular mechanism underlying MCH neuron development remains poorly understood. We report that a three-dimensional culture of mouse embryonic stem cells (mESCs) can generate hypothalamic-like tissues containing MCH-positive neurons, which reproduce morphologic maturation, neuronal connectivity, and neuropeptide/neurotransmitter phenotype of native MCH neurons. Using this in vitro system, we demonstrate that Hedgehog (Hh) signaling serves to produce major neurochemical subtypes of MCH neurons characterized by the presence or absence of cocaine- and amphetamine-regulated transcript (CART). Without exogenous Hh signals, mESCs initially differentiated into dorsal hypothalamic/prethalamic progenitors and finally into MCH+CART+ neurons through a specific intermediate progenitor state. Conversely, activation of the Hh pathway specified ventral hypothalamic progenitors that generate both MCH+CART− and MCH+CART+ neurons. These results suggest that in vivo MCH neurons may originate from multiple cell lineages that arise through early dorsoventral patterning of the hypothalamus. Additionally, we found that Hh signaling supports the differentiation of mESCs into orexin/hypocretin neurons, a well-defined cell group intermingled with MCH neurons in the lateral hypothalamic area (LHA). The present study highlights and improves the utility of mESC culture in the analysis of the developmental programs of specific hypothalamic cell types.
Collapse
|
14
|
Bakshi A, Singh R, Rai U. Trajectory of leptin and leptin receptor in vertebrates: Structure, function and their regulation. Comp Biochem Physiol B Biochem Mol Biol 2021; 257:110652. [PMID: 34343670 DOI: 10.1016/j.cbpb.2021.110652] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 06/23/2021] [Accepted: 07/29/2021] [Indexed: 12/11/2022]
Abstract
The present review provides a comparative insight into structure, function and control of leptin system in fishes, herptiles, birds and mammals. In general, leptin acts as an anorexigenic hormone since its administration results in decrease of food intake in vertebrates. Nonetheless, functional paradox arises in fishes from contradictory observations on level of leptin during fasting and re-feeding. In addition, leptin is shown to modulate metabolic functions in fishes, reptiles, birds and mammals. Leptin also regulates reproductive and immune functions though more studies are warranted in non-mammalian vertebrates. The expression of leptin and its receptor is influenced by numerous factors including sex steroids, stress and stress-induced catecholamines and glucocorticoids though their effect in non-mammalian vertebrates is hard to be generalized due to limited studies.
Collapse
Affiliation(s)
- Amrita Bakshi
- Department of Zoology, University of Delhi, Delhi 110007, India
| | - Rajeev Singh
- Satyawati College, University of Delhi, Delhi 110052, India
| | - Umesh Rai
- Department of Zoology, University of Delhi, Delhi 110007, India.
| |
Collapse
|
15
|
Tan Y, Hang F, Liu ZW, Stoiljkovic M, Wu M, Tu Y, Han W, Lee AM, Kelley C, Hajós M, Lu L, de Lecea L, De Araujo I, Picciotto MR, Horvath TL, Gao XB. Impaired hypocretin/orexin system alters responses to salient stimuli in obese male mice. J Clin Invest 2021; 130:4985-4998. [PMID: 32516139 DOI: 10.1172/jci130889] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 06/03/2020] [Indexed: 12/27/2022] Open
Abstract
The brain has evolved in an environment where food sources are scarce, and foraging for food is one of the major challenges for survival of the individual and species. Basic and clinical studies show that obesity or overnutrition leads to overwhelming changes in the brain in animals and humans. However, the exact mechanisms underlying the consequences of excessive energy intake are not well understood. Neurons expressing the neuropeptide hypocretin/orexin (Hcrt) in the lateral/perifonical hypothalamus (LH) are critical for homeostatic regulation, reward seeking, stress response, and cognitive functions. In this study, we examined adaptations in Hcrt cells regulating behavioral responses to salient stimuli in diet-induced obese mice. Our results demonstrated changes in primary cilia, synaptic transmission and plasticity, cellular responses to neurotransmitters necessary for reward seeking, and stress responses in Hcrt neurons from obese mice. Activities of neuronal networks in the LH and hippocampus were impaired as a result of decreased hypocretinergic function. The weakened Hcrt system decreased reward seeking while altering responses to acute stress (stress-coping strategy), which were reversed by selectively activating Hcrt cells with chemogenetics. Taken together, our data suggest that a deficiency in Hcrt signaling may be a common cause of behavioral changes (such as lowered arousal, weakened reward seeking, and altered stress response) in obese animals.
Collapse
Affiliation(s)
- Ying Tan
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Fu Hang
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Guangxi Reproductive Medical Research Center, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhong-Wu Liu
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Milan Stoiljkovic
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Mingxing Wu
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Ophthalmology, Second Affiliate Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Tu
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Traditional Chinese Medicine Health Preservation, Second Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wenfei Han
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Angela M Lee
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Craig Kelley
- Joint Biomedical Engineering Program, SUNY Downstate and NYU Tandon, Brooklyn, New York, USA
| | - Mihály Hajós
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Ivan De Araujo
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Xiao-Bing Gao
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
16
|
De Pablo-Fernández E, Warner TT. Hypothalamic α-synuclein and its relation to autonomic symptoms and neuroendocrine abnormalities in Parkinson disease. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:223-233. [PMID: 34266594 DOI: 10.1016/b978-0-12-819973-2.00015-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder presenting with defining motor features and a variable combination of nonmotor symptoms. There is growing evidence suggesting that hypothalamic involvement in PD may contribute to the pathogenesis of nonmotor symptoms. Initial neuropathologic studies demonstrated histologic involvement of hypothalamic nuclei by Lewy pathology, i.e., neuronal aggregates including Lewy bodies (round eosinophilic inclusions with a halo found in the neuronal perikarya) and other inclusions in neuronal processes such as Lewy neurites. Recent studies using more sensitive immunohistochemistry have shown that synuclein deposition is common in all hypothalamic nuclei and can happen at preclinical stages of the disease. Several neuropathologic changes, including synuclein deposition, neuronal loss, and adaptative morphologic changes, have been described in neurochemically defined specific hypothalamic cell populations with a potential role in the pathogenesis of nonmotor symptoms such as autonomic dysfunction, blood pressure control, circadian rhythms, sleep, and body weight regulation. The clinical implications of these hypothalamic neuropathologic changes are not fully understood and a direct clinical correlation may be challenging due to the multifactorial pathogenesis of the symptomatology and the additional involvement of other peripheral regulatory mechanisms. Future neuropathologic research using histological and functional assessments should establish the potential role of hypothalamic dysfunction on clinical burden, symptomatic therapies, and disease biomarkers in PD.
Collapse
Affiliation(s)
- Eduardo De Pablo-Fernández
- Reta Lila Weston Institute and Queen Square Brain Bank, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Movement and Clinical Neuroscience, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Thomas T Warner
- Reta Lila Weston Institute and Queen Square Brain Bank, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Movement and Clinical Neuroscience, UCL Queen Square Institute of Neurology, London, United Kingdom.
| |
Collapse
|
17
|
Campbell EJ, Hill MK, Maddern XJ, Jin S, Pang TY, Lawrence AJ. Orexin-1 receptor signaling within the lateral hypothalamus, but not bed nucleus of the stria terminalis, mediates context-induced relapse to alcohol seeking. J Psychopharmacol 2020; 34:1261-1270. [PMID: 33063594 DOI: 10.1177/0269881120959638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The lateral hypothalamic orexin (hypocretin) system has a well-established role in the motivation for reward. This has particular relevance to substance use disorders since orexin-1 receptors play a critical role in alcohol-seeking behavior, acting at multiple nodes in relapse-associated networks. AIMS This study aimed to further our understanding of the role of orexin-1 receptor signaling within the lateral hypothalamus and bed nucleus of the stria terminalis, specifically in context-induced relapse to alcohol-seeking following punishment-imposed abstinence. METHODS We trained inbred male alcohol-preferring rats to self-administer alcohol in one environment or context (Context A) and subsequently punished their alcohol-reinforced lever presses in a different environment (Context B) using contingent foot shock punishment. Finally, we tested rats for relapse-like behavior in either context following systemic, intra-lateral hypothalamus or intra-bed nucleus of the stria terminalis orexin-1 receptor antagonism with SB-334867. RESULTS/OUTCOMES We found that systemic orexin-1 receptor antagonism significantly reduced alcohol-seeking in both contexts. Intra-lateral hypothalamus orexin-1 receptor antagonism significantly reduced alcohol-seeking in Context A whereas intra-bed nucleus of the stria terminalis orexin-1 receptor antagonism had no effect on alcohol-seeking behavior. CONCLUSIONS/INTERPRETATION Our results suggest a role for the orexin-1 receptor system in context-induced relapse to alcohol-seeking. Specifically, intra-lateral hypothalamus orexin microcircuits contribute to alcohol-seeking.
Collapse
Affiliation(s)
- Erin J Campbell
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Mitchell Kri Hill
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Xavier J Maddern
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Shubo Jin
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Terence Y Pang
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health and Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3052, Australia
| |
Collapse
|
18
|
Lee J, Raycraft L, Johnson AW. The dynamic regulation of appetitive behavior through lateral hypothalamic orexin and melanin concentrating hormone expressing cells. Physiol Behav 2020; 229:113234. [PMID: 33130035 DOI: 10.1016/j.physbeh.2020.113234] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
The lateral hypothalamic area (LHA) is a heterogeneous brain structure extensively studied for its potent role in regulating energy balance. The anatomical and molecular diversity of the LHA permits the orchestration of responses to energy sensing cues from the brain and periphery. Two of the primary cell populations within the LHA associated with integration of this information are Orexin (ORX) and Melanin Concentrating Hormone (MCH). While both of these non-overlapping populations exhibit orexigenic properties, the activities of these two systems support feeding behavior through contrasting mechanisms. We describe the anatomical and functional properties as well as interaction with other neuropeptides and brain reward and hedonic systems. Specific outputs relating to arousal, food seeking, feeding, and metabolism are coordinated through these mechanisms. We then discuss how both the ORX and MCH systems harmonize in a divergent yet overall cooperative manner to orchestrate feeding behavior through transitions between various appetitive states, and thus offer novel insights into LHA allostatic control of appetite.
Collapse
Affiliation(s)
| | | | - Alexander W Johnson
- Department of Psychology; Neuroscience Program, Michigan State University, East Lansing.
| |
Collapse
|
19
|
Bandaru SS, Khanday MA, Ibrahim N, Naganuma F, Vetrivelan R. Sleep-Wake Control by Melanin-Concentrating Hormone (MCH) Neurons: a Review of Recent Findings. Curr Neurol Neurosci Rep 2020; 20:55. [PMID: 33006677 PMCID: PMC11891936 DOI: 10.1007/s11910-020-01075-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE OF THE REVIEW Melanin-concentrating hormone (MCH)-expressing neurons located in the lateral hypothalamus are considered as an integral component of sleep-wake circuitry. However, the precise role of MCH neurons in sleep-wake regulation has remained unclear, despite several years of research employing a wide range of techniques. We review recent data on this aspect, which are mostly inconsistent, and propose a novel role for MCH neurons in sleep regulation. RECENT FINDINGS While almost all studies using "gain-of-function" approaches show an increase in rapid eye movement sleep (or paradoxical sleep; PS), loss-of-function approaches have not shown reductions in PS. Similarly, the reported changes in wakefulness or non-rapid eye movement sleep (slow-wave sleep; SWS) with manipulation of the MCH system using conditional genetic methods are inconsistent. Currently available data do not support a role for MCH neurons in spontaneous sleep-wake but imply a crucial role for them in orchestrating sleep-wake responses to changes in external and internal environments.
Collapse
Affiliation(s)
- Sathyajit S Bandaru
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA
| | - Mudasir A Khanday
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Nazifa Ibrahim
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA
- Department of Public Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Fumito Naganuma
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ramalingam Vetrivelan
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA.
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Crouse RB, Kim K, Batchelor HM, Girardi EM, Kamaletdinova R, Chan J, Rajebhosale P, Pittenger ST, Role LW, Talmage DA, Jing M, Li Y, Gao XB, Mineur YS, Picciotto MR. Acetylcholine is released in the basolateral amygdala in response to predictors of reward and enhances the learning of cue-reward contingency. eLife 2020; 9:e57335. [PMID: 32945260 PMCID: PMC7529459 DOI: 10.7554/elife.57335] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
The basolateral amygdala (BLA) is critical for associating initially neutral cues with appetitive and aversive stimuli and receives dense neuromodulatory acetylcholine (ACh) projections. We measured BLA ACh signaling and activity of neurons expressing CaMKIIα (a marker for glutamatergic principal cells) in mice during cue-reward learning using a fluorescent ACh sensor and calcium indicators. We found that ACh levels and nucleus basalis of Meynert (NBM) cholinergic terminal activity in the BLA (NBM-BLA) increased sharply in response to reward-related events and shifted as mice learned the cue-reward contingency. BLA CaMKIIα neuron activity followed reward retrieval and moved to the reward-predictive cue after task acquisition. Optical stimulation of cholinergic NBM-BLA terminal fibers led to a quicker acquisition of the cue-reward contingency. These results indicate BLA ACh signaling carries important information about salient events in cue-reward learning and provides a framework for understanding how ACh signaling contributes to shaping BLA responses to emotional stimuli.
Collapse
Affiliation(s)
- Richard B Crouse
- Department of Psychiatry, Yale UniversityNew HavenUnited States
- Yale Interdepartmental Neuroscience ProgramNew HavenUnited States
| | - Kristen Kim
- Department of Psychiatry, Yale UniversityNew HavenUnited States
- Yale Interdepartmental Neuroscience ProgramNew HavenUnited States
| | - Hannah M Batchelor
- Department of Psychiatry, Yale UniversityNew HavenUnited States
- Yale Interdepartmental Neuroscience ProgramNew HavenUnited States
| | - Eric M Girardi
- Department of Psychiatry, Yale UniversityNew HavenUnited States
| | - Rufina Kamaletdinova
- Department of Psychiatry, Yale UniversityNew HavenUnited States
- City University of New York, Hunter CollegeNew YorkUnited States
| | - Justin Chan
- Department of Psychiatry, Yale UniversityNew HavenUnited States
| | - Prithviraj Rajebhosale
- Program in Neuroscience, Stony Brook UniversityNew YorkUnited States
- National Institute of Neurological Disorders and Stroke (NINDS)BethesdaUnited States
| | | | - Lorna W Role
- National Institute of Neurological Disorders and Stroke (NINDS)BethesdaUnited States
| | - David A Talmage
- National Institute of Mental Health (NIMH)BethesdaUnited States
| | - Miao Jing
- Chinese Institute for Brain Research (CIBR)BeijingChina
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life SciencesBeijingChina
- PKU-IDG/McGovern Institute for Brain ResearchBeijingChina
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| | - Xiao-Bing Gao
- Section of Comparative Medicine, Yale University School of MedicineNew HavenUnited States
| | - Yann S Mineur
- Department of Psychiatry, Yale UniversityNew HavenUnited States
| | - Marina R Picciotto
- Department of Psychiatry, Yale UniversityNew HavenUnited States
- Yale Interdepartmental Neuroscience ProgramNew HavenUnited States
| |
Collapse
|
21
|
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: 22] [Impact Index Per Article: 4.4] [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
|
22
|
Naganuma F, Bandaru SS, Absi G, Mahoney CE, Scammell TE, Vetrivelan R. Melanin-concentrating hormone neurons contribute to dysregulation of rapid eye movement sleep in narcolepsy. Neurobiol Dis 2018; 120:12-20. [PMID: 30149182 PMCID: PMC6195361 DOI: 10.1016/j.nbd.2018.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 08/02/2018] [Accepted: 08/23/2018] [Indexed: 12/22/2022] Open
Abstract
The lateral hypothalamus contains neurons producing orexins that promote wakefulness and suppress REM sleep as well as neurons producing melanin-concentrating hormone (MCH) that likely promote REM sleep. Narcolepsy with cataplexy is caused by selective loss of the orexin neurons, and the MCH neurons appear unaffected. As the orexin and MCH systems exert opposing effects on REM sleep, we hypothesized that imbalance in this REM sleep-regulating system due to activity in the MCH neurons may contribute to the striking REM sleep dysfunction characteristic of narcolepsy. To test this hypothesis, we chemogenetically activated the MCH neurons and pharmacologically blocked MCH signaling in a murine model of narcolepsy and studied the effects on sleep-wake behavior and cataplexy. To chemoactivate MCH neurons, we injected an adeno-associated viral vector containing the hM3Dq stimulatory DREADD into the lateral hypothalamus of orexin null mice that also express Cre recombinase in the MCH neurons (MCH-Cre::OX-KO mice) and into control MCH-Cre mice with normal orexin expression. In both lines of mice, activation of MCH neurons by clozapine-N-oxide (CNO) increased rapid eye movement (REM) sleep without altering other states. In mice lacking orexins, activation of the MCH neurons also increased abnormal intrusions of REM sleep manifest as cataplexy and short latency transitions into REM sleep (SLREM). Conversely, a MCH receptor 1 antagonist, SNAP 94847, almost completely eliminated SLREM and cataplexy in OX-KO mice. These findings affirm that MCH neurons promote REM sleep under normal circumstances, and their activity in mice lacking orexins likely triggers abnormal intrusions of REM sleep into non-REM sleep and wake, resulting in the SLREM and cataplexy characteristic of narcolepsy.
Collapse
Affiliation(s)
- Fumito Naganuma
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston MA-02215, USA; Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1, Fukumuro, Miyagino-ku, Sendai 983-8536, Japan
| | - Sathyajit S Bandaru
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston MA-02215, USA
| | - Gianna Absi
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston MA-02215, USA
| | - Carrie E Mahoney
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston MA-02215, USA
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston MA-02215, USA
| | - Ramalingam Vetrivelan
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston MA-02215, USA.
| |
Collapse
|
23
|
Linehan V, Rowe TM, Hirasawa M. Dopamine modulates excitatory transmission to orexin neurons in a receptor subtype-specific manner. Am J Physiol Regul Integr Comp Physiol 2018; 316:R68-R75. [PMID: 30462527 DOI: 10.1152/ajpregu.00150.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Dopamine (DA) can promote or inhibit consummatory and reward-related behaviors by activating different receptor subtypes in the lateral hypothalamus and perifornical area (LH/PF). Because orexin neurons are involved in reward and localized in the LH/PF, DA may modulate these neurons to influence reward-related behaviors. To determine the cellular mechanism underlying dopaminergic modulation of orexin neurons, the effect of DA on excitatory transmission to these neurons was investigated using in vitro electrophysiology on rat brain slices. We found that low concentrations (0.1-1 µM) of DA increased evoked excitatory postsynaptic current amplitude while decreasing paired-pulse ratio. In contrast, high concentrations (10-100 µM) of DA did the opposite. The excitatory effect of low DA was blocked by the D1 receptor antagonist SCH-23390, whereas the inhibitory effect of high DA was blocked by the D2 receptor antagonist sulpiride. These results indicate distinct roles of D1 and D2 receptors in bidirectional presynaptic modulation of excitatory transmission. DA had stronger effects on isolated synaptic activity than repetitive ones, suggesting that sensitivity to dopaminergic modulation depends on the level of network activity. In orexin neurons from high-fat diet-fed rats, a high concentration of DA was less effective in suppressing repetitive synaptic activity compared with chow controls. Therefore, in diet-induced obesity, intense synaptic inputs may preferentially reach orexin neurons while intermittent signals are inhibited by high DA levels. In summary, our study provides a cellular mechanism by which DA may exert opposite behavioral effects in the LH/PF through bidirectional modulation of orexin neurons via different DA receptors.
Collapse
Affiliation(s)
- Victoria Linehan
- Division of Biomedical Sciences, Memorial University , St. John's, Newfoundland , Canada
| | - Todd M Rowe
- Division of Biomedical Sciences, Memorial University , St. John's, Newfoundland , Canada
| | - Michiru Hirasawa
- Division of Biomedical Sciences, Memorial University , St. John's, Newfoundland , Canada
| |
Collapse
|
24
|
Schmeichel BE, Matzeu A, Koebel P, Vendruscolo LF, Sidhu H, Shahryari R, Kieffer BL, Koob GF, Martin-Fardon R, Contet C. Knockdown of hypocretin attenuates extended access of cocaine self-administration in rats. Neuropsychopharmacology 2018; 43:2373-2382. [PMID: 29703996 PMCID: PMC6180106 DOI: 10.1038/s41386-018-0054-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 12/22/2022]
Abstract
The hypocretin/orexin (HCRT) neuropeptide system regulates feeding, arousal state, stress responses, and reward, especially under conditions of enhanced motivational relevance. In particular, HCRT neurotransmission facilitates drug-seeking behavior in circumstances that demand increased effort and/or motivation to take the drug. The present study used a shRNA-encoding adeno-associated viral vector to knockdown Hcrt expression throughout the dorsal hypothalamus in adult rats and determine the role of HCRT in cocaine self-administration. Chronic Hcrt silencing did not impact cocaine self-administration under short-access conditions, but robustly attenuated cocaine intake under extended access conditions, a model that mimics key features of compulsive cocaine taking. In addition, Hcrt silencing decreased motivation for both cocaine and a highly palatable food reward (i.e., sweetened condensed milk; SCM) under a progressive ratio schedule of reinforcement, but did not alter responding for SCM under a fixed ratio schedule. Importantly, Hcrt silencing did not affect food or water consumption, and had no consequence for general measures of arousal and stress reactivity. At the molecular level, chronic Hcrt knockdown reduced the number of neurons expressing dynorphin (DYN), and to a smaller extent melanin-concentrating hormone (MCH), in the dorsal hypothalamus. These original findings support the hypothesis that HCRT neurotransmission promotes operant responding for both drug and non-drug rewards, preferentially under conditions requiring a high degree of motivation. Furthermore, the current study provides compelling evidence for the involvement of the HCRT system in cocaine self-administration also under low-effort conditions in rats allowed extended access, possibly via functional interactions with DYN and MCH signaling.
Collapse
Affiliation(s)
- Brooke E Schmeichel
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA.
| | - Alessandra Matzeu
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Pascale Koebel
- Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, 67404, France
| | - Leandro F Vendruscolo
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
| | - Harpreet Sidhu
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Roxana Shahryari
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Brigitte L Kieffer
- Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, 67404, France
- Douglas Institute Research Centre, McGill University, Montréal, QC, Canada
| | - George F Koob
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
| | - Rémi Martin-Fardon
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Candice Contet
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
25
|
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
|
26
|
Latifi B, Adamantidis A, Bassetti C, Schmidt MH. Sleep-Wake Cycling and Energy Conservation: Role of Hypocretin and the Lateral Hypothalamus in Dynamic State-Dependent Resource Optimization. Front Neurol 2018; 9:790. [PMID: 30344503 PMCID: PMC6183196 DOI: 10.3389/fneur.2018.00790] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/31/2018] [Indexed: 12/23/2022] Open
Abstract
The hypocretin (Hcrt) system has been implicated in a wide range of physiological functions from sleep-wake regulation to cardiovascular, behavioral, metabolic, and thermoregulagtory control. These wide-ranging physiological effects have challenged the identification of a parsimonious function for Hcrt. A compelling hypothesis suggests that Hcrt plays a role in the integration of sleep-wake neurophysiology with energy metabolism. For example, Hcrt neurons promote waking and feeding, but are also sensors of energy balance. Loss of Hcrt function leads to an increase in REM sleep propensity, but a potential role for Hcrt linking energy balance with REM sleep expression has not been addressed. Here we examine a potential role for Hcrt and the lateral hypothalamus (LH) in state-dependent resource allocation as a means of optimizing resource utilization and, as a result, energy conservation. We review the energy allocation hypothesis of sleep and how state-dependent metabolic partitioning may contribute toward energy conservation, but with additional examination of how the loss of thermoregulatory function during REM sleep may impact resource optimization. Optimization of energy expenditures at the whole organism level necessitates a top-down network responsible for coordinating metabolic operations in a state-dependent manner across organ systems. In this context, we then specifically examine the potential role of the LH in regulating this output control, including the contribution from both Hcrt and melanin concentrating hormone (MCH) neurons among a diverse LH cell population. We propose that this hypothalamic integration system is responsible for global shifts in state-dependent resource allocations, ultimately promoting resource optimization and an energy conservation function of sleep-wake cycling.
Collapse
Affiliation(s)
- Blerina Latifi
- Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Antoine Adamantidis
- Department of Neurology, Center for Experimental Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department of Biomedical Research, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Claudio Bassetti
- Department of Neurology, Center for Experimental Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Markus H Schmidt
- Department of Neurology, Center for Experimental Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Ohio Sleep Medicine Institute, Dublin, OH, United States
| |
Collapse
|
27
|
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
|
28
|
Blanco-Centurion C, Bendell E, Zou B, Sun Y, Shiromani PJ, Liu M. VGAT and VGLUT2 expression in MCH and orexin neurons in double transgenic reporter mice. IBRO Rep 2018; 4:44-49. [PMID: 30155524 PMCID: PMC6111069 DOI: 10.1016/j.ibror.2018.05.001] [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: 03/30/2018] [Revised: 04/23/2018] [Accepted: 05/09/2018] [Indexed: 11/11/2022] Open
Abstract
MCH neurons contain neither VGAT nor VGLUT2. Majority of orexin neurons contain VGLUT2. MCH neurons do not contain orexin.
The neuropeptides orexin and melanin-concentrating hormone (MCH), as well as the neurotransmitters GABA (γ-Aminobutyric acid) and glutamate are chief modulators of the sleep-wake states in the posterior hypothalamus. To investigate co-expression of vesicular GABA transporter (VGAT, a marker of GABA neurons) and the vesicular glutamate transporter-2 (VGLUT2, a marker of glutamate neurons) in orexin and MCH neurons, we generated two transgenic mouse lines. One line selectively expressed the reporter gene EYFP in VGAT+ neurons, whereas the other line expressed reporter gene tdTomato in VGLUT2+ neurons. Co-localization between these genetic reporters and orexin or MCH immunofluorescent tags was determined using 3D computer reconstructions of Z stacks that were acquired using a multiphoton laser confocal microscope. Our results demonstrated that MCH neurons expressed neither VGAT nor VGLUT2, suggesting MCH neurons are a separate cluster of cells from VGAT+ GABAergic neurons and VGLUT2+ glutamatergic neurons. Moreover, most orexin neurons expressed VGLUT2, indicating these neurons are glutamatergic. Our data suggested that in the posterior hypothalamus there are four major distinct groups of neurons: VGAT+, orexin+/VGLUT2+, orexin-/VGLUT2+, and MCH neurons. This study facilitated our understanding of the role of these neurotransmitters and neuropeptides in relation to sleep/wake regulation.
Collapse
Key Words
- Arousal
- CeA, central nucleus of amygdala
- GABA
- GABA-γ, Aminobutyric acid
- GAD65, glutamic acid decarboxylase-65
- GAD67, glutamic acid decarboxylase-67
- Gad1, Glutamate decarboxylase 1
- Glutamate
- MCH, melanin concentrating hormone
- NREM, non-rapid eye movement
- REM, rapid eye movement
- RTN, reticular thalamic nucleus
- SSC, somatosensory cortex
- Sleep
- VGAT, vesicular GABA transporter
- VGLUT2, vesicular glutamate transporter-2
Collapse
Affiliation(s)
- Carlos Blanco-Centurion
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Emmaline Bendell
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Bingyu Zou
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Ying Sun
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Priyattam J Shiromani
- Ralph H. Johnson VA Medical Center, Charleston, SC, 29425, USA.,Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Meng Liu
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| |
Collapse
|
29
|
Sil’kis IG. A Neurochemical Approach to the Search for Drugs for the Treatment of Symptoms of Alzheimer’s Disease. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712418010130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Laperchia C, Xu YZ, Mumba Ngoyi D, Cotrufo T, Bentivoglio M. Neural Damage in Experimental Trypanosoma brucei gambiense Infection: Hypothalamic Peptidergic Sleep and Wake-Regulatory Neurons. Front Neuroanat 2018. [PMID: 29535612 PMCID: PMC5835115 DOI: 10.3389/fnana.2018.00013] [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] [Indexed: 01/08/2023] Open
Abstract
Neuron populations of the lateral hypothalamus which synthesize the orexin (OX)/hypocretin or melanin-concentrating hormone (MCH) peptides play crucial, reciprocal roles in regulating wake stability and sleep. The disease human African trypanosomiasis (HAT), also called sleeping sickness, caused by extracellular Trypanosoma brucei (T. b.) parasites, leads to characteristic sleep-wake cycle disruption and narcoleptic-like alterations of the sleep structure. Previous studies have revealed damage of OX and MCH neurons during systemic infection of laboratory rodents with the non-human pathogenic T. b. brucei subspecies. No information is available, however, on these peptidergic neurons after systemic infection with T. b. gambiense, the etiological agent of 97% of HAT cases. The present study was aimed at the investigation of immunohistochemically characterized OX and MCH neurons after T. b. gambiense or T. b. brucei infection of a susceptible rodent, the multimammate mouse, Mastomysnatalensis. Cell counts and evaluation of OX fiber density were performed at 4 and 8 weeks post-infection, when parasites had entered the brain parenchyma from the periphery. A significant decrease of OX neurons (about 44% reduction) and MCH neurons (about 54% reduction) was found in the lateral hypothalamus and perifornical area at 8 weeks in T. b. gambiense-infected M. natalensis. A moderate decrease (21% and 24% reduction, respectively), which did not reach statistical significance, was found after T. b. brucei infection. In two key targets of diencephalic orexinergic innervation, the peri-suprachiasmatic nucleus (SCN) region and the thalamic paraventricular nucleus (PVT), densitometric analyses showed a significant progressive decrease in the density of orexinergic fibers in both infection paradigms, and especially during T. b. gambiense infection. Altogether the findings provide novel information showing that OX and MCH neurons are highly vulnerable to chronic neuroinflammatory signaling caused by the infection of human-pathogenic African trypanosomes.
Collapse
Affiliation(s)
- Claudia Laperchia
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Yuan-Zhong Xu
- Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Dieudonné Mumba Ngoyi
- Institut National de Recherche Biomédicale (INRB), Kinshasa, Democratic Republic of Congo
| | - Tiziana Cotrufo
- 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 (INN), Verona Unit, Verona, Italy
| |
Collapse
|
31
|
Baimel C, Borgland SL. Hypocretin/Orexin and Plastic Adaptations Associated with Drug Abuse. Curr Top Behav Neurosci 2017; 33:283-304. [PMID: 28303403 DOI: 10.1007/7854_2016_44] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dopamine neurons in the ventral tegmental area (VTA) are a critical part of the neural circuits that underlie reward learning and motivation. Dopamine neurons send dense projections throughout the brain and recent observations suggest that both the intrinsic properties and the functional output of dopamine neurons are dependent on projection target and are subject to neuromodulatory influences. Lateral hypothalamic hypocretin (also termed orexin) neurons project to the VTA and contain both hypocretin and dynorphin peptides in the same dense core vesicles suggesting they may be co-released. Hypocretin peptides act at excitatory Gαq protein-coupled receptors and dynorphin acts at inhibitory Gαi/o protein-coupled receptors, which are both expressed on subpopulations of dopamine neurons. This review describes a role for neuromodulation of dopamine neurons and the influence on motivated behaviour in response to natural and drug rewards.
Collapse
Affiliation(s)
- Corey Baimel
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, Canada, T2N 4N1
| | - Stephanie L Borgland
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, Canada, T2N 4N1.
| |
Collapse
|
32
|
Optogenetic Investigation of Arousal Circuits. Int J Mol Sci 2017; 18:ijms18081773. [PMID: 28809797 PMCID: PMC5578162 DOI: 10.3390/ijms18081773] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/06/2017] [Accepted: 08/09/2017] [Indexed: 12/13/2022] Open
Abstract
Modulation between sleep and wake states is controlled by a number of heterogeneous neuron populations. Due to the topological proximity and genetic co-localization of the neurons underlying sleep-wake state modulation optogenetic methods offer a significant improvement in the ability to benefit from both the precision of genetic targeting and millisecond temporal control. Beginning with an overview of the neuron populations mediating arousal, this review outlines the progress that has been made in the investigation of arousal circuits since the incorporation of optogenetic techniques and the first in vivo application of optogenetic stimulation in hypocretin neurons in the lateral hypothalamus. This overview is followed by a discussion of the future progress that can be made by incorporating more recent technological developments into the research of neural circuits.
Collapse
|
33
|
Silkis IG. Hypothetical neurochemical mechanisms of paradoxical sleep deficiency in Alzheimer’s disease. NEUROCHEM J+ 2017. [DOI: 10.1134/s181971241702012x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
34
|
Equihua-Benítez AC, Guzmán-Vásquez K, Drucker-Colín R. Understanding sleep-wake mechanisms and drug discovery. Expert Opin Drug Discov 2017; 12:643-657. [PMID: 28511597 DOI: 10.1080/17460441.2017.1329818] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Although not discernible at first glance, sleep is a highly active and regulated brain state. Although we spend practically one third of our lifetimes in this stage, its importance is often taken for granted. Sleep loss can lead to disease, error and economic loss. Our understanding of how sleep is achieved has greatly advanced in recent years, and with that, the management of sleep disorders has improved. There is still room for improvement and recently many new compounds have reached clinical trials with a few being approved for commercial use. Areas covered: In this review, the authors make the case of sleep disorders as a matter of public health. The mechanisms of sleep transition are discussed emphasizing the wake and sleep promoting interaction of different brain regions. Finally, advances in pharmacotherapy are examined in the context of chronic insomnia and narcolepsy. Expert opinion: The orexinergic system is an example of a breakthrough in sleep medicine that has catalyzed drug development. Nevertheless, sleep is a topic still with many unanswered questions. That being said, the melanin-concentrating hormone system is becoming increasingly relevant and we speculate it will be the next target of sleep medication.
Collapse
Affiliation(s)
- Ana Clementina Equihua-Benítez
- a Departamento de Neuropatología Molecular , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Ciudad de México , México
| | - Khalil Guzmán-Vásquez
- a Departamento de Neuropatología Molecular , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Ciudad de México , México
| | - René Drucker-Colín
- a Departamento de Neuropatología Molecular , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Ciudad de México , México
| |
Collapse
|
35
|
Yamashita T, Yamanaka A. Lateral hypothalamic circuits for sleep-wake control. Curr Opin Neurobiol 2017; 44:94-100. [PMID: 28427008 DOI: 10.1016/j.conb.2017.03.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/31/2017] [Indexed: 12/12/2022]
Abstract
The lateral hypothalamic area (LHA) of the diencephalon is crucially involved in controlling instinctive behavior such as sleep-wake cycle and feeding behavior. LHA is a heterogeneous structure that contains spatially intermingled, genetically distinct cell populations. Among LHA neurons, orexin/hypocretin (OX) neuron is the key cell type that promotes waking, and specific loss of OX neurons results in narcolepsy. Melanin-concentrating hormone (MCH) containing neurons are known to be active during rapid eye movement (REM) sleep and stimulation of these neurons promotes REM sleep. Here we review the classical and more recent findings in this field and discuss the molecular and cellular network organization of LHA neurons that could ultimately regulate the switch between wakefulness and general states of sleep.
Collapse
Affiliation(s)
- Takayuki Yamashita
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan; PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan; CREST, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan; CREST, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
36
|
Narwade SC, Mallick BN, Deobagkar DD. Transcriptome Analysis Reveals Altered Expression of Memory and Neurotransmission Associated Genes in the REM Sleep Deprived Rat Brain. Front Mol Neurosci 2017; 10:67. [PMID: 28367113 PMCID: PMC5355427 DOI: 10.3389/fnmol.2017.00067] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/28/2017] [Indexed: 12/21/2022] Open
Abstract
Sleep disorders are associated with cognitive impairment. Selective rapid eye movement sleep (REMS) deprivation (REMSD) alters several physiological processes and behaviors. By employing NGS platform we carried out transcriptomic analysis in brain samples of control rats and those exposed to REMSD. The expression of genes involved in chromatin assembly, methylation, learning, memory, regulation of synaptic transmission, neuronal plasticity and neurohypophysial hormone synthesis were altered. Increased transcription of BMP4, DBH and ATP1B2 genes after REMSD supports our earlier findings and hypothesis. Alteration in the transcripts encoding histone subtypes and important players in chromatin remodeling was observed. The mRNAs which transcribe neurotransmitters such as OXT, AVP, PMCH and LNPEP and two small non-coding RNAs, namely RMRP and BC1 were down regulated. At least some of these changes are likely to regulate REMS and may participate in the consequences of REMS loss. Thus, the findings of this study have identified key epigenetic regulators and neuronal plasticity genes associated to REMS and its loss. This analysis provides a background and opens up avenues for unraveling their specific roles in the complex behavioral network particularly in relation to sustained REMS-loss associated changes.
Collapse
Affiliation(s)
- Santosh C Narwade
- Molecular Biology Research Laboratory, Center of Advanced Studies, Department of Zoology, Savitribai Phule Pune University Pune, India
| | | | - Deepti D Deobagkar
- Molecular Biology Research Laboratory, Center of Advanced Studies, Department of Zoology, Savitribai Phule Pune UniversityPune, India; Bioinformatics Center, Savitribai Phule Pune UniversityPune, India
| |
Collapse
|
37
|
Eban-Rothschild A, de Lecea L. Neuronal substrates for initiation, maintenance, and structural organization of sleep/wake states. F1000Res 2017; 6:212. [PMID: 28357049 PMCID: PMC5345773 DOI: 10.12688/f1000research.9677.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2017] [Indexed: 11/20/2022] Open
Abstract
Animals continuously alternate between sleep and wake states throughout their life. The daily organization of sleep and wakefulness is orchestrated by circadian, homeostatic, and motivational processes. Over the last decades, much progress has been made toward determining the neuronal populations involved in sleep/wake regulation. Here, we will discuss how the application of advanced
in vivo tools for cell type–specific manipulations now permits the functional interrogation of different features of sleep/wake state regulation: initiation, maintenance, and structural organization. We will specifically focus on recent studies examining the roles of wake-promoting neuronal populations.
Collapse
Affiliation(s)
- Ada Eban-Rothschild
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
38
|
Schrölkamp M, Jennum PJ, Gammeltoft S, Holm A, Kornum BR, Knudsen S. Normal Morning Melanin-Concentrating Hormone Levels and No Association with Rapid Eye Movement or Non-Rapid Eye Movement Sleep Parameters in Narcolepsy Type 1 and Type 2. J Clin Sleep Med 2017; 13:235-243. [PMID: 27855741 DOI: 10.5664/jcsm.6454] [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: 07/07/2016] [Accepted: 10/03/2016] [Indexed: 01/07/2023]
Abstract
STUDY OBJECTIVES Other than hypocretin-1 (HCRT-1) deficiency in narcolepsy type 1 (NT1), the neurochemical imbalance of NT1 and narcolepsy type 2 (NT2) with normal HCRT-1 levels is largely unknown. The neuropeptide melanin-concentrating hormone (MCH) is mainly secreted during sleep and is involved in rapid eye movement (REM) and non-rapid eye movement (NREM) sleep regulation. Hypocretin neurons reciprocally interact with MCH neurons. We hypothesized that altered MCH secretion contributes to the symptoms and sleep abnormalities of narcolepsy and that this is reflected in morning cerebrospinal fluid (CSF) MCH levels, in contrast to previously reported normal evening/afternoon levels. METHODS Lumbar CSF and plasma were collected from 07:00 to 10:00 from 57 patients with narcolepsy (subtypes: 47 NT1; 10 NT2) diagnosed according to International Classification of Sleep Disorders, Third Edition (ICSD-3) and 20 healthy controls. HCRT-1 and MCH levels were quantified by radioimmunoassay and correlated with clinical symptoms, polysomnography (PSG), and Multiple Sleep Latency Test (MSLT) parameters. RESULTS CSF and plasma MCH levels were not significantly different between narcolepsy patients regardless of ICSD-3 subtype, HCRT-1 levels, or compared to controls. CSF MCH and HCRT-1 levels were not significantly correlated. Multivariate regression models of CSF MCH levels, age, sex, and body mass index predicting clinical, PSG, and MSLT parameters did not reveal any significant associations to CSF MCH levels. CONCLUSIONS Our study shows that MCH levels in CSF collected in the morning are normal in narcolepsy and not associated with the clinical symptoms, REM sleep abnormalities, nor number of muscle movements during REM or NREM sleep of the patients. We conclude that morning lumbar CSF MCH measurement is not an informative diagnostic marker for narcolepsy.
Collapse
Affiliation(s)
- Maren Schrölkamp
- Molecular Sleep Laboratory, Department of Clinical Biochemistry, Rigshospitalet, Glostrup, Denmark.,FU-Berlin, Faculty Biology, Chemistry, Pharmacy, Takustr, Berlin, Germany
| | - Poul J Jennum
- Danish Center for Sleep Medicine, University of Copenhagen, Rigshospitalet, Glostrup, Denmark
| | - Steen Gammeltoft
- Molecular Sleep Laboratory, Department of Clinical Biochemistry, Rigshospitalet, Glostrup, Denmark
| | - Anja Holm
- Molecular Sleep Laboratory, Department of Clinical Biochemistry, Rigshospitalet, Glostrup, Denmark
| | - Birgitte R Kornum
- Molecular Sleep Laboratory, Department of Clinical Biochemistry, Rigshospitalet, Glostrup, Denmark
| | - Stine Knudsen
- Danish Center for Sleep Medicine, University of Copenhagen, Rigshospitalet, Glostrup, Denmark.,Norwegian Centre of Expertise for Neurodevelopmental Disorders and Hypersomnias (NevSom), Oslo University Hospital, Ullevål, Norway
| |
Collapse
|
39
|
Villano I, Messina A, Valenzano A, Moscatelli F, Esposito T, Monda V, Esposito M, Precenzano F, Carotenuto M, Viggiano A, Chieffi S, Cibelli G, Monda M, Messina G. Basal Forebrain Cholinergic System and Orexin Neurons: Effects on Attention. Front Behav Neurosci 2017; 11:10. [PMID: 28197081 PMCID: PMC5281635 DOI: 10.3389/fnbeh.2017.00010] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 01/12/2017] [Indexed: 12/27/2022] Open
Abstract
The basal forebrain (BF) cholinergic system has an important role in attentive functions. The cholinergic system can be activated by different inputs, and in particular, by orexin neurons, whose cell bodies are located within the postero-lateral hypothalamus. Recently the orexin-producing neurons have been proved to promote arousal and attention through their projections to the BF. The aim of this review article is to summarize the evidence showing that the orexin system contributes to attentional processing by an increase in cortical acetylcholine release and in cortical neurons activity.
Collapse
Affiliation(s)
- Ines Villano
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Antonietta Messina
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Fiorenzo Moscatelli
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy; Department of Motor, Human and Health Science, University of Rome, "Foro Italico"Rome, Italy
| | - Teresa Esposito
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Vincenzo Monda
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Maria Esposito
- Department of Mental Health, Physical and Preventive Medicine, Second University of Naples Naples, Italy
| | - Francesco Precenzano
- Department of Mental Health, Physical and Preventive Medicine, Second University of Naples Naples, Italy
| | - Marco Carotenuto
- Department of Mental Health, Physical and Preventive Medicine, Second University of NaplesNaples, Italy; Neapolitan Brain Group (NBG), Clinic of Child and Adolescent Neuropsychiatry, Department of Mental, Physical Health and Preventive Medicine, Second University of NaplesNaples, Italy
| | - Andrea Viggiano
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno Salerno, Italy
| | - Sergio Chieffi
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Giuseppe Cibelli
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Second University of Naples Naples, Italy
| | - Giovanni Messina
- Department of Experimental Medicine, Second University of NaplesNaples, Italy; Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| |
Collapse
|
40
|
Weber F, Dan Y. Circuit-based interrogation of sleep control. Nature 2016; 538:51-59. [PMID: 27708309 DOI: 10.1038/nature19773] [Citation(s) in RCA: 235] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/17/2016] [Indexed: 12/20/2022]
Abstract
Sleep is a fundamental biological process observed widely in the animal kingdom, but the neural circuits generating sleep remain poorly understood. Understanding the brain mechanisms controlling sleep requires the identification of key neurons in the control circuits and mapping of their synaptic connections. Technical innovations over the past decade have greatly facilitated dissection of the sleep circuits. This has set the stage for understanding how a variety of environmental and physiological factors influence sleep. The ability to initiate and terminate sleep on command will also help us to elucidate its functions within and beyond the brain.
Collapse
Affiliation(s)
- Franz Weber
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, California 94720, USA
| | - Yang Dan
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, California 94720, USA
| |
Collapse
|
41
|
Blanco-Centurion C, Liu M, Konadhode RP, Zhang X, Pelluru D, van den Pol AN, Shiromani PJ. Optogenetic activation of melanin-concentrating hormone neurons increases non-rapid eye movement and rapid eye movement sleep during the night in rats. Eur J Neurosci 2016; 44:2846-2857. [PMID: 27657541 DOI: 10.1111/ejn.13410] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/15/2016] [Accepted: 09/19/2016] [Indexed: 01/06/2023]
Abstract
Neurons containing melanin-concentrating hormone (MCH) are located in the hypothalamus. In mice, optogenetic activation of the MCH neurons induces both non-rapid eye movement (NREM) and rapid eye movement (REM) sleep at night, the normal wake-active period for nocturnal rodents [R. R. Konadhode et al. (2013) J. Neurosci., 33, 10257-10263]. Here we selectively activate these neurons in rats to test the validity of the sleep network hypothesis in another species. Channelrhodopsin-2 (ChR2) driven by the MCH promoter was selectively expressed by MCH neurons after injection of rAAV-MCHp-ChR2-EYFP into the hypothalamus of Long-Evans rats. An in vitro study confirmed that the optogenetic activation of MCH neurons faithfully triggered action potentials. In the second study, in Long-Evans rats, rAAV-MCH-ChR2, or the control vector, rAAV-MCH-EYFP, were delivered into the hypothalamus. Three weeks later, baseline sleep was recorded for 48 h without optogenetic stimulation (0 Hz). Subsequently, at the start of the lights-off cycle, the MCH neurons were stimulated at 5, 10, or 30 Hz (1 mW at tip; 1 min on - 4 min off) for 24 h. Sleep was recorded during the 24-h stimulation period. Optogenetic activation of MCH neurons increased both REM and NREM sleep at night, whereas during the day cycle, only REM sleep was increased. Delta power, an indicator of sleep intensity, was also increased. In control rats without ChR2, optogenetic stimulation did not increase sleep or delta power. These results lend further support to the view that sleep-active MCH neurons contribute to drive sleep in mammals.
Collapse
Affiliation(s)
- Carlos Blanco-Centurion
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, 114 Doughty Street, MSC 404/STB 404, Charleston, SC, 29425, USA
| | - Meng Liu
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, 114 Doughty Street, MSC 404/STB 404, Charleston, SC, 29425, USA
| | - Roda P Konadhode
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, 114 Doughty Street, MSC 404/STB 404, Charleston, SC, 29425, USA
| | - Xiaobing Zhang
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | - Dheeraj Pelluru
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, 114 Doughty Street, MSC 404/STB 404, Charleston, SC, 29425, USA
| | | | - Priyattam J Shiromani
- Department of Psychiatry & Behavioral Sciences, Medical University of South Carolina, 114 Doughty Street, MSC 404/STB 404, Charleston, SC, 29425, USA.,Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| |
Collapse
|
42
|
Arrigoni E, Chen MC, Fuller PM. The anatomical, cellular and synaptic basis of motor atonia during rapid eye movement sleep. J Physiol 2016; 594:5391-414. [PMID: 27060683 DOI: 10.1113/jp271324] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 03/02/2016] [Indexed: 01/14/2023] Open
Abstract
Rapid eye movement (REM) sleep is a recurring part of the sleep-wake cycle characterized by fast, desynchronized rhythms in the electroencephalogram (EEG), hippocampal theta activity, rapid eye movements, autonomic activation and loss of postural muscle tone (atonia). The brain circuitry governing REM sleep is located in the pontine and medullary brainstem and includes ascending and descending projections that regulate the EEG and motor components of REM sleep. The descending signal for postural muscle atonia during REM sleep is thought to originate from glutamatergic neurons of the sublaterodorsal nucleus (SLD), which in turn activate glycinergic pre-motor neurons in the spinal cord and/or ventromedial medulla to inhibit motor neurons. Despite work over the past two decades on many neurotransmitter systems that regulate the SLD, gaps remain in our knowledge of the synaptic basis by which SLD REM neurons are regulated and in turn produce REM sleep atonia. Elucidating the anatomical, cellular and synaptic basis of REM sleep atonia control is a critical step for treating many sleep-related disorders including obstructive sleep apnoea (apnea), REM sleep behaviour disorder (RBD) and narcolepsy with cataplexy.
Collapse
Affiliation(s)
- Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02215, USA.
| | - Michael C Chen
- Department of Neurology, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Patrick M Fuller
- Department of Neurology, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
43
|
Oonk M, Krueger JM, Davis CJ. Voluntary Sleep Loss in Rats. Sleep 2016; 39:1467-79. [PMID: 27166236 PMCID: PMC4909628 DOI: 10.5665/sleep.5984] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/28/2016] [Indexed: 12/22/2022] Open
Abstract
STUDY OBJECTIVES Animal sleep deprivation (SDEP), in contrast to human SDEP, is involuntary and involves repeated exposure to aversive stimuli including the inability of the animal to control the waking stimulus. Therefore, we explored intracranial self-stimulation (ICSS), an operant behavior, as a method for voluntary SDEP in rodents. METHODS Male Sprague-Dawley rats were implanted with electroencephalography/electromyography (EEG/EMG) recording electrodes and a unilateral bipolar electrode into the lateral hypothalamus. Rats were allowed to self-stimulate, or underwent gentle handling-induced SDEP (GH-SDEP), during the first 6 h of the light phase, after which they were allowed to sleep. Other rats performed the 6 h ICSS and 1 w later were subjected to 6 h of noncontingent stimulation (NCS). During NCS the individual stimulation patterns recorded during ICSS were replayed. RESULTS After GH-SDEP, ICSS, or NCS, time in nonrapid eye movement (NREM) sleep and rapid eye movement (REM) sleep increased. Further, in the 24 h after SDEP, rats recovered all of the REM sleep lost during SDEP, but only 75% to 80% of the NREM sleep lost, regardless of the SDEP method. The magnitude of EEG slow wave responses occurring during NREM sleep also increased after SDEP treatments. However, NREM sleep EEG slow wave activity (SWA) responses were attenuated following ICSS, compared to GH-SDEP and NCS. CONCLUSIONS We conclude that ICSS and NCS can be used to sleep deprive rats. Changes in rebound NREM sleep EEG SWA occurring after ICSS, NCS, and GH-SDEP suggest that nonspecific effects of the SDEP procedure differentially affect recovery sleep phenotypes.
Collapse
Affiliation(s)
- Marcella Oonk
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA
| | - James M. Krueger
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA
| | - Christopher J. Davis
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA
- Sleep and Performance Research Center, Washington State University, Spokane, WA
| |
Collapse
|
44
|
Varin C, Arthaud S, Salvert D, Gay N, Libourel PA, Luppi PH, Léger L, Fort P. Sleep architecture and homeostasis in mice with partial ablation of melanin-concentrating hormone neurons. Behav Brain Res 2015; 298:100-10. [PMID: 26529469 DOI: 10.1016/j.bbr.2015.10.051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/23/2015] [Accepted: 10/25/2015] [Indexed: 12/23/2022]
Abstract
Recent reports support a key role of tuberal hypothalamic neurons secreting melanin concentrating-hormone (MCH) in the promotion of Paradoxical Sleep (PS). Controversies remain concerning their concomitant involvement in Slow-Wave Sleep (SWS). We studied the effects of their selective loss achieved by an Ataxin 3-mediated ablation strategy to decipher the contribution of MCH neurons to SWS and/or PS. Polysomnographic recordings were performed on male adult transgenic mice expressing Ataxin-3 transgene within MCH neurons (MCH(Atax)) and their wild-type littermates (MCH(WT)) bred on two genetic backgrounds (FVB/N and C57BL/6). Compared to MCH(WT) mice, MCH(Atax) mice were characterized by a significant drop in MCH mRNAs (-70%), a partial loss of MCH-immunoreactive neurons (-30%) and a marked reduction in brain density of MCH-immunoreactive fibers. Under basal condition, such MCH(Atax) mice exhibited higher PS amounts during the light period and a pronounced SWS fragmentation without any modification of SWS quantities. Moreover, SWS and PS rebounds following 4-h total sleep deprivation were quantitatively similar in MCH(Atax)vs. MCH(WT) mice. Additionally, MCH(Atax) mice were unable to consolidate SWS and increase slow-wave activity (SWA) in response to this homeostatic challenge as observed in MCH(WT) littermates. Here, we show that the partial loss of MCH neurons is sufficient to disturb the fine-tuning of sleep. Our data provided new insights into their contribution to subtle process managing SWS quality and its efficiency rather than SWS quantities, as evidenced by the deleterious impact on two powerful markers of sleep depth, i.e., SWS consolidation/fragmentation and SWA intensity under basal condition and under high sleep pressure.
Collapse
Affiliation(s)
- Christophe Varin
- Neuroscience Research Center of Lyon (CRNL), CNRS UMR 5292, INSERM U1028, SLEEP Team, Lyon, France; Université Claude Bernard, Lyon 1, Lyon, France
| | - Sébastien Arthaud
- Neuroscience Research Center of Lyon (CRNL), CNRS UMR 5292, INSERM U1028, SLEEP Team, Lyon, France; Université Claude Bernard, Lyon 1, Lyon, France
| | - Denise Salvert
- Neuroscience Research Center of Lyon (CRNL), CNRS UMR 5292, INSERM U1028, SLEEP Team, Lyon, France; Université Claude Bernard, Lyon 1, Lyon, France
| | - Nadine Gay
- Neuroscience Research Center of Lyon (CRNL), CNRS UMR 5292, INSERM U1028, SLEEP Team, Lyon, France; Université Claude Bernard, Lyon 1, Lyon, France
| | - Paul-Antoine Libourel
- Neuroscience Research Center of Lyon (CRNL), CNRS UMR 5292, INSERM U1028, SLEEP Team, Lyon, France; Université Claude Bernard, Lyon 1, Lyon, France
| | - Pierre-Hervé Luppi
- Neuroscience Research Center of Lyon (CRNL), CNRS UMR 5292, INSERM U1028, SLEEP Team, Lyon, France; Université Claude Bernard, Lyon 1, Lyon, France
| | - Lucienne Léger
- Neuroscience Research Center of Lyon (CRNL), CNRS UMR 5292, INSERM U1028, SLEEP Team, Lyon, France; Université Claude Bernard, Lyon 1, Lyon, France
| | - Patrice Fort
- Neuroscience Research Center of Lyon (CRNL), CNRS UMR 5292, INSERM U1028, SLEEP Team, Lyon, France; Université Claude Bernard, Lyon 1, Lyon, France.
| |
Collapse
|
45
|
Gao XB, Hermes G. Neural plasticity in hypocretin neurons: the basis of hypocretinergic regulation of physiological and behavioral functions in animals. Front Syst Neurosci 2015; 9:142. [PMID: 26539086 PMCID: PMC4612503 DOI: 10.3389/fnsys.2015.00142] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 10/02/2015] [Indexed: 12/22/2022] Open
Abstract
The neuronal system that resides in the perifornical and lateral hypothalamus (Pf/LH) and synthesizes the neuropeptide hypocretin/orexin participates in critical brain functions across species from fish to human. The hypocretin system regulates neural activity responsible for daily functions (such as sleep/wake homeostasis, energy balance, appetite, etc.) and long-term behavioral changes (such as reward seeking and addiction, stress response, etc.) in animals. The most recent evidence suggests that the hypocretin system undergoes substantial plastic changes in response to both daily fluctuations (such as food intake and sleep-wake regulation) and long-term changes (such as cocaine seeking) in neuronal activity in the brain. The understanding of these changes in the hypocretin system is essential in addressing the role of the hypocretin system in normal physiological functions and pathological conditions in animals and humans. In this review, the evidence demonstrating that neural plasticity occurs in hypocretin-containing neurons in the Pf/LH will be presented and possible physiological, behavioral, and mental health implications of these findings will be discussed.
Collapse
Affiliation(s)
- Xiao-Bing Gao
- Section of Comparative Medicine, Yale University School of Medicine New Haven, CT, USA ; Program on Integrative Cell Signaling and Neurobiology of Metabolism (ICSNM), Yale University School of Medicine New Haven, CT, USA
| | - Gretchen Hermes
- Department of Psychiatry, Yale University School of Medicine New Haven, CT, USA
| |
Collapse
|
46
|
Feltelius N, Persson I, Ahlqvist-Rastad J, Andersson M, Arnheim-Dahlström L, Bergman P, Granath F, Adori C, Hökfelt T, Kühlmann-Berenzon S, Liljeström P, Maeurer M, Olsson T, Örtqvist Å, Partinen M, Salmonson T, Zethelius B. A coordinated cross-disciplinary research initiative to address an increased incidence of narcolepsy following the 2009-2010 Pandemrix vaccination programme in Sweden. J Intern Med 2015; 278:335-53. [PMID: 26123389 DOI: 10.1111/joim.12391] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In response to the 2009-2010 influenza A(H1N1)pdm09 pandemic, a mass vaccination programme with the AS03-adjuvanted influenza A(H1N1) vaccine Pandemrix was initiated in Sweden. Unexpectedly, there were a number of narcolepsy cases amongst vaccinated children and adolescents reported. In this review, we summarize the results of a joint cross-disciplinary national research effort to investigate the adverse reaction signal from the spontaneous reporting system and to better understand possible causative mechanisms. A three- to fourfold increased risk of narcolepsy in vaccinated children and adolescents was verified by epidemiological studies. Of importance, no risk increase was observed for the other neurological and autoimmune diseases studied. Genetic studies confirmed the association with the allele HLA-DQB1*06:02, which is known to be related to sporadic narcolepsy. Furthermore, a number of studies using cellular and molecular experimental models investigated possible links between influenza vaccination and narcolepsy. Serum analysis, using a peptide microarray platform, showed that individuals who received Pandemrix exhibited a different epitope reactivity pattern to neuraminidase and haemagglutinin, as compared to individuals who were infected with H1N1. Patients with narcolepsy were also found to have increased levels of interferon-gamma production in response to streptococcus-associated antigens. The chain of patient-related events and the study results emerging over time were subjected to intense nationwide media attention. The importance of transparent communication and collaboration with patient representatives to maintain public trust in vaccination programmes is also discussed in the review. Organizational challenges due to this unexpected event delayed the initiation of some of the research projects, still the main objectives of this joint, cross-disciplinary research effort were reached, and important insights were acquired for future, similar situations in which a fast and effective task force may be required to evaluate vaccination-related adverse events.
Collapse
Affiliation(s)
| | - I Persson
- Medical Products Agency, Uppsala, Sweden
| | | | | | - L Arnheim-Dahlström
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - P Bergman
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet and Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - F Granath
- Clinical Epidemiology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - C Adori
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - T Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - P Liljeström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - M Maeurer
- Therapeutic Immunology Division, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - T Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Å Örtqvist
- Department of Communicable Disease Control and Prevention, Stockholm County Council, Stockholm, Sweden.,Department of Medicine, Unit of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - M Partinen
- Helsinki Sleep Clinic, Vitalmed Research Centre, Helsinki, Finland.,Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | | | | |
Collapse
|
47
|
Melanin-concentrating hormone neurons release glutamate for feedforward inhibition of the lateral septum. J Neurosci 2015; 35:3644-51. [PMID: 25716862 DOI: 10.1523/jneurosci.4187-14.2015] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Melanin-concentrating hormone (MCH) regulates vital physiological functions, including energy balance and sleep. MCH cells are thought to be GABAergic, releasing GABA to inhibit downstream targets. However, there is little experimental support for this paradigm. To better understand the synaptic mechanisms of mouse MCH neurons, we performed neuroanatomical mapping and characterization followed by optogenetics to test their functional connectivity at downstream targets. Synaptophysin-mediated projection mapping showed that the lateral septal nucleus (LS) contained the densest accumulation of MCH nerve terminals. We then expressed channel rhodopsin-2 in MCH neurons and photostimulated MCH projections to determine their effect on LS activity. Photostimulation of MCH projections evoked a monosynaptic glutamate release in the LS. Interestingly, this led to a feedforward inhibition that depressed LS firing by a robust secondary GABA release. This study presents a circuit analysis between MCH and LS neurons and confirms their functional connection via monosynaptic and polysynaptic pathways. Our findings indicate that MCH neurons are not exclusively GABAergic and reveal a glutamate-mediated, feedforward mechanism that inhibits LS cells.
Collapse
|
48
|
Linehan V, Trask RB, Briggs C, Rowe TM, Hirasawa M. Concentration-dependent activation of dopamine receptors differentially modulates GABA release onto orexin neurons. Eur J Neurosci 2015; 42:1976-83. [PMID: 26036709 DOI: 10.1111/ejn.12967] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 05/28/2015] [Indexed: 01/11/2023]
Abstract
Dopamine (DA) and orexin neurons play important roles in reward and food intake. There are anatomical and functional connections between these two cell groups: orexin peptides stimulate DA neurons in the ventral tegmental area and DA inhibits orexin neurons in the hypothalamus. However, the cellular mechanisms underlying the action of DA on orexin neurons remain incompletely understood. Therefore, the effect of DA on inhibitory transmission to orexin neurons was investigated in rat brain slices using the whole-cell patch-clamp technique. We found that DA modulated the frequency of spontaneous and miniature IPSCs (mIPSCs) in a concentration-dependent bidirectional manner. Low (1 μM) and high (100 μM) concentrations of DA decreased and increased IPSC frequency, respectively. These effects did not accompany a change in mIPSC amplitude and persisted in the presence of G-protein signaling inhibitor GDPβS in the pipette, suggesting that DA acts presynaptically. The decrease in mIPSC frequency was mediated by D2 receptors whereas the increase required co-activation of D1 and D2 receptors and subsequent activation of phospholipase C. In summary, our results suggest that DA has complex effects on GABAergic transmission to orexin neurons, involving cooperation of multiple receptor subtypes. The direction of dopaminergic influence on orexin neurons is dependent on the level of DA in the hypothalamus. At low levels DA disinhibits orexin neurons whereas at high levels it facilitates GABA release, which may act as negative feedback to curb the excitatory orexinergic output to DA neurons. These mechanisms may have implications for consummatory and motivated behaviours.
Collapse
Affiliation(s)
- Victoria Linehan
- Division of Biomedical Sciences, Memorial University, St. John's, Newfoundland, Canada, A1B3V6
| | - Robert B Trask
- Division of Biomedical Sciences, Memorial University, St. John's, Newfoundland, Canada, A1B3V6
| | - Chantalle Briggs
- Division of Biomedical Sciences, Memorial University, St. John's, Newfoundland, Canada, A1B3V6.,Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada, B3H4R2
| | - Todd M Rowe
- Division of Biomedical Sciences, Memorial University, St. John's, Newfoundland, Canada, A1B3V6
| | - Michiru Hirasawa
- Division of Biomedical Sciences, Memorial University, St. John's, Newfoundland, Canada, A1B3V6
| |
Collapse
|
49
|
Abstract
Sleep is expressed as a circadian rhythm and the two phenomena exist in a poorly understood relationship. Light affects each, simultaneously influencing rhythm phase and rapidly inducing sleep. Light has long been known to modulate sleep, but recent discoveries support its use as an effective nocturnal stimulus for eliciting sleep in certain rodents. “Photosomnolence” is mediated by classical and ganglion cell photoreceptors and occurs despite the ongoing high levels of locomotion at the time of stimulus onset. Brief photic stimuli trigger rapid locomotor suppression, sleep, and a large drop in core body temperature (Tc; Phase 1), followed by a relatively fixed duration interval of sleep (Phase 2) and recovery (Phase 3) to pre-sleep activity levels. Additional light can lengthen Phase 2. Potential retinal pathways through which the sleep system might be light-activated are described and the potential roles of orexin (hypocretin) and melanin-concentrating hormone are discussed. The visual input route is a practical avenue to follow in pursuit of the neural circuitry and mechanisms governing sleep and arousal in small nocturnal mammals and the organizational principles may be similar in diurnal humans. Photosomnolence studies are likely to be particularly advantageous because the timing of sleep is largely under experimenter control. Sleep can now be effectively studied using uncomplicated, nonintrusive methods with behavior evaluation software tools; surgery for EEG electrode placement is avoidable. The research protocol for light-induced sleep is easily implemented and useful for assessing the effects of experimental manipulations on the sleep induction pathway. Moreover, the experimental designs and associated results benefit from a substantial amount of existing neuroanatomical and pharmacological literature that provides a solid framework guiding the conduct and interpretation of future investigations.
Collapse
|
50
|
Süssmuth SD, Haider S, Landwehrmeyer GB, Farmer R, Frost C, Tripepi G, Andersen CA, Di Bacco M, Lamanna C, Diodato E, Massai L, Diamanti D, Mori E, Magnoni L, Dreyhaupt J, Schiefele K, Craufurd D, Saft C, Rudzinska M, Ryglewicz D, Orth M, Brzozy S, Baran A, Pollio G, Andre R, Tabrizi SJ, Darpo B, Westerberg G. An exploratory double-blind, randomized clinical trial with selisistat, a SirT1 inhibitor, in patients with Huntington's disease. Br J Clin Pharmacol 2015; 79:465-76. [PMID: 25223731 PMCID: PMC4345957 DOI: 10.1111/bcp.12512] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 09/06/2014] [Indexed: 12/22/2022] Open
Abstract
AIMS Selisistat, a selective SirT1 inhibitor is being developed as a potentially disease-modifying therapeutic for Huntington's disease (HD). This was the first study of selisistat in HD patients and was primarily aimed at development of pharmacodynamic biomarkers. METHODS This was a randomized, double-blind, placebo-controlled, multicentre exploratory study. Fifty-five male and female patients in early stage HD were randomized to receive 10 mg or 100 mg of selisistat or placebo once daily for 14 days. Blood sampling, clinical and safety assessments were conducted throughout the study. Candidate pharmacodynamic markers included circulating soluble huntingtin and innate immune markers. RESULTS Selisistat was found to be safe and well tolerated, and systemic exposure parameters showed that the average steady-state plasma concentration achieved at the 10 mg dose level (125 nm) was comparable with the IC50 for SirT1 inhibition. No adverse effects on motor, cognitive or functional readouts were recorded. While circulating levels of soluble huntingtin were not affected by selisistat in this study, the biological samples collected have allowed development of assay technology for use in future studies. No effects on innate immune markers were seen. CONCLUSIONS Selisistat was found to be safe and well tolerated in early stage HD patients at plasma concentrations within the anticipated therapeutic concentration range.
Collapse
Affiliation(s)
| | - Salman Haider
- Department of Neurodegenerative Disease, University College London Institute of NeurologyLondon, United Kingdom
| | | | - Ruth Farmer
- Department of Medical Statistics, London School of Hygiene and Tropical MedicineLondon, United Kingdom
| | - Chris Frost
- Department of Medical Statistics, London School of Hygiene and Tropical MedicineLondon, United Kingdom
| | | | | | | | - Claudia Lamanna
- Siena Biotech SpASiena, Italy
- European Huntington's Disease Network (EHDN)Chieti, Italy
| | | | | | | | | | | | - Jens Dreyhaupt
- Institute of Epidemiology and Medical Biometry, Ulm UniversityUlm, Germany
| | - Karin Schiefele
- Institute of Epidemiology and Medical Biometry, Ulm UniversityUlm, Germany
| | - David Craufurd
- Institute of Human Development, Faculty of Medical and Human Sciences, University of ManchesterManchester, United Kingdom
- Manchester Centre for Genomic Medicine, Central Manchester University Hospitals NHS Foundation Trust and Manchester Academic Health Science CentreManchester, United Kingdom
| | - Carsten Saft
- Department of Neurology, Huntington-Center NRW, St. Josef-Hospital, Ruhr-UniversityBochum, Germany
| | - Monika Rudzinska
- Department of Neurology, Medical University of SilesiaKatowice, Poland
| | - Danuta Ryglewicz
- Department of Neurology, Institute of Psychiatry and NeurologyWarsaw, Poland
| | - Michael Orth
- Department of Neurology, Ulm University HospitalUlm, Germany
| | | | | | | | - Ralph Andre
- Department of Neurodegenerative Disease, University College London Institute of NeurologyLondon, United Kingdom
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, University College London Institute of NeurologyLondon, United Kingdom
| | - Borje Darpo
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Karolinska Institutet, Danderyd HospitalStockholm, Sweden
- iCardiac TechnologiesRochester, NY, USA
| | - Goran Westerberg
- Siena Biotech SpASiena, Italy
- La Crocina Pharmaceutical Consultants D.I.San Giovanni d'Asso (S), Italy
| | - Paddington Consortium
- Department of Neurology, Ulm University HospitalUlm, Germany
- Department of Neurodegenerative Disease, University College London Institute of NeurologyLondon, United Kingdom
- Department of Medical Statistics, London School of Hygiene and Tropical MedicineLondon, United Kingdom
- Siena Biotech SpASiena, Italy
- European Huntington's Disease Network (EHDN)Chieti, Italy
- Institute of Epidemiology and Medical Biometry, Ulm UniversityUlm, Germany
- Institute of Human Development, Faculty of Medical and Human Sciences, University of ManchesterManchester, United Kingdom
- Manchester Centre for Genomic Medicine, Central Manchester University Hospitals NHS Foundation Trust and Manchester Academic Health Science CentreManchester, United Kingdom
- Department of Neurology, Huntington-Center NRW, St. Josef-Hospital, Ruhr-UniversityBochum, Germany
- Department of Neurology, Medical University of SilesiaKatowice, Poland
- Department of Neurology, Institute of Psychiatry and NeurologyWarsaw, Poland
- KCR.S.A.Warsaw, Poland
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Karolinska Institutet, Danderyd HospitalStockholm, Sweden
- iCardiac TechnologiesRochester, NY, USA
- La Crocina Pharmaceutical Consultants D.I.San Giovanni d'Asso (S), Italy
| |
Collapse
|