1
|
Guillot SJ, Lang C, Simonot M, Beckett D, Lulé D, Balz LT, Knehr A, Stuart-Lopez G, Vercruysse P, Dieterlé S, Weydt P, Dorst J, Kandler K, Kassubek J, Wassermann L, Rouaux C, Arthaud S, Da Cruz S, Luppi PH, Roselli F, Ludolph AC, Dupuis L, Bolborea M. Early-onset sleep alterations found in patients with amyotrophic lateral sclerosis are ameliorated by orexin antagonist in mouse models. Sci Transl Med 2025; 17:eadm7580. [PMID: 39879320 DOI: 10.1126/scitranslmed.adm7580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 09/19/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025]
Abstract
Sleep alterations have been described in several neurodegenerative diseases yet are currently poorly characterized in amyotrophic lateral sclerosis (ALS). This study investigates sleep macroarchitecture and related hypothalamic signaling disruptions in ALS. Using polysomnography, we found that both patients with ALS as well as asymptomatic C9ORF72 and SOD1 mutation carriers exhibited increased wakefulness and reduced non-rapid eye movement sleep. Increased wakefulness correlated with diminished cognitive performance in both clinical cohorts. Similar changes in sleep macroarchitecture were observed in three ALS mouse models (Sod1G86R, FusΔNLS/+, and TDP43Q331K). A single oral administration of a dual-orexin receptor antagonist or intracerebroventricular delivery of melanin-concentrating hormone (MCH) through an osmotic pump over 15 days partially normalized sleep patterns in mouse models. MCH treatment did not extend the survival of Sod1G86R mice but did decrease the loss of lumbar motor neurons. These findings suggest MCH and orexin signaling as potential targets to treat sleep alterations that arise in early stages of the disease.
Collapse
Affiliation(s)
- Simon J Guillot
- University of Strasbourg, INSERM, Strasbourg Translational Neuroscience & Psychiatry STEP-CRBS, UMR-S 1329, 67000 Strasbourg, France
| | - Christina Lang
- Department of Neurology, University Hospital of Ulm, 89081 Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). 89081 Ulm, Germany
| | - Marie Simonot
- University of Strasbourg, INSERM, Strasbourg Translational Neuroscience & Psychiatry STEP-CRBS, UMR-S 1329, 67000 Strasbourg, France
| | - Daniel Beckett
- University of Strasbourg, INSERM, Strasbourg Translational Neuroscience & Psychiatry STEP-CRBS, UMR-S 1329, 67000 Strasbourg, France
| | - Dorothée Lulé
- Department of Neurology, University Hospital of Ulm, 89081 Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). 89081 Ulm, Germany
| | - Luisa T Balz
- Department of Neurology, University Hospital of Ulm, 89081 Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). 89081 Ulm, Germany
| | - Antje Knehr
- Department of Neurology, University Hospital of Ulm, 89081 Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). 89081 Ulm, Germany
| | - Geoffrey Stuart-Lopez
- University of Strasbourg, INSERM, Strasbourg Translational Neuroscience & Psychiatry STEP-CRBS, UMR-S 1329, 67000 Strasbourg, France
| | - Pauline Vercruysse
- University of Strasbourg, INSERM, Strasbourg Translational Neuroscience & Psychiatry STEP-CRBS, UMR-S 1329, 67000 Strasbourg, France
| | - Stéphane Dieterlé
- University of Strasbourg, INSERM, Strasbourg Translational Neuroscience & Psychiatry STEP-CRBS, UMR-S 1329, 67000 Strasbourg, France
| | - Patrick Weydt
- Department of Neurology, University Hospital of Ulm, 89081 Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 53127 Bonn, Germany
| | - Johannes Dorst
- Department of Neurology, University Hospital of Ulm, 89081 Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). 89081 Ulm, Germany
| | - Katharina Kandler
- Department of Neurology, University Hospital of Ulm, 89081 Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). 89081 Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, University Hospital of Ulm, 89081 Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). 89081 Ulm, Germany
| | - Laura Wassermann
- Department of Neurology, University Hospital of Ulm, 89081 Ulm, Germany
| | - Caroline Rouaux
- University of Strasbourg, INSERM, Strasbourg Translational Neuroscience & Psychiatry STEP-CRBS, UMR-S 1329, 67000 Strasbourg, France
| | - Sébastien Arthaud
- Centre of Neuroscience of Lyon, CNRS/INSERM, UMR 5292/UMR 1028, 69675 Lyon, France
| | - Sandrine Da Cruz
- VIB-KU Leuven Center for Brain and Disease Research and Department of Neurosciences, KU Leuven, 3001 Leuven, Belgium
| | - Pierre-Hervé Luppi
- Centre of Neuroscience of Lyon, CNRS/INSERM, UMR 5292/UMR 1028, 69675 Lyon, France
| | - Francesco Roselli
- Department of Neurology, University Hospital of Ulm, 89081 Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). 89081 Ulm, Germany
| | - Albert C Ludolph
- Department of Neurology, University Hospital of Ulm, 89081 Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). 89081 Ulm, Germany
| | - Luc Dupuis
- University of Strasbourg, INSERM, Strasbourg Translational Neuroscience & Psychiatry STEP-CRBS, UMR-S 1329, 67000 Strasbourg, France
| | - Matei Bolborea
- University of Strasbourg, INSERM, Strasbourg Translational Neuroscience & Psychiatry STEP-CRBS, UMR-S 1329, 67000 Strasbourg, France
| |
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
|
Xiong T, Tsuchida L, Inutsuka A, Onaka T, Yamada K, Orikasa C. Novel aspect of oxytocin neurons mediating parental behavior and aversive burying behavior under the control of melanin-concentrating hormone neurons. Front Behav Neurosci 2024; 18:1459957. [PMID: 39376641 PMCID: PMC11456465 DOI: 10.3389/fnbeh.2024.1459957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/12/2024] [Indexed: 10/09/2024] Open
Abstract
Parental behavior comprises a set of crucial actions essential for offspring survival. In this study, a double transgenic mouse model engineered to specifically express channelrhodopsin-2 (ChR2) in paraventricular hypothalamic nucleus (PVN)-oxytocin neurons and ablate lateral hypothalamic area (LHA)-melanin-concentrating hormone (MCH) neurons was used to determine the relationship between PVN-oxytocin neurons and LHA-MCH neurons associated with parental behavior. Optogenetic stimulation of ChR2-expressing PVN-oxytocin neurons induces typical parental behavior with intact LHA-MCH neurons. However, after the partial ablation of LHA-MCH neurons, even optogenetic stimulation of PVN-oxytocin neurons failed to induce parental behavior in virgin male mice, resulting in neglect rather than parental behavior. Furthermore, approximately half of the subjects exhibited burying behavior toward pups, suggesting that pups became aversive stimuli, and male mice actively performed burying behavior to avoid these aversive stimuli. This study emphasizes the novel aspect of oxytocin neurons that could result in neglect in the absence of LHA-MCH neurons regulation.
Collapse
Affiliation(s)
- Tingbi Xiong
- Laboratory for Psychology and Behavioral Neuroscience, University of Tsukuba, Tsukuba, Japan
| | - Lena Tsuchida
- Laboratory for Psychology and Behavioral Neuroscience, University of Tsukuba, Tsukuba, Japan
| | - Ayumu Inutsuka
- Department of Physiology, Jichi Medical University, Shimotsuke, Japan
| | - Tatsushi Onaka
- Department of Physiology, Jichi Medical University, Shimotsuke, Japan
| | - Kazuo Yamada
- Laboratory for Psychology and Behavioral Neuroscience, University of Tsukuba, Tsukuba, Japan
| | - Chitose Orikasa
- Laboratory for Morphological and Biomolecular Imaging, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
4
|
Spencer CD, Miller PA, Williams-Ikhenoba JG, Nikolova RG, Chee MJ. Regulation of the Mouse Ventral Tegmental Area by Melanin-Concentrating Hormone. J Neurosci 2024; 44:e0790232024. [PMID: 38806249 PMCID: PMC11223476 DOI: 10.1523/jneurosci.0790-23.2024] [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: 04/30/2023] [Revised: 05/02/2024] [Accepted: 05/15/2024] [Indexed: 05/30/2024] Open
Abstract
Melanin-concentrating hormone (MCH) acts via its sole receptor MCHR1 in rodents and is an important regulator of homeostatic behaviors like feeding, sleep, and mood to impact overall energy balance. The loss of MCH signaling by MCH or MCHR1 deletion produces hyperactive mice with increased energy expenditure, and these effects are consistently associated with a hyperdopaminergic state. We recently showed that MCH suppresses dopamine release in the nucleus accumbens, which principally receives dopaminergic projections from the ventral tegmental area (VTA), but the mechanisms underlying MCH-regulated dopamine release are not clearly defined. MCHR1 expression is widespread and includes dopaminergic VTA cells. However, as the VTA is a neurochemically diverse structure, we assessed Mchr1 gene expression at glutamatergic, GABAergic, and dopaminergic VTA cells and determined if MCH inhibited the activity of VTA cells and/or their local microcircuit. Mchr1 expression was robust in major VTA cell types, including most dopaminergic (78%) or glutamatergic cells (52%) and some GABAergic cells (38%). Interestingly, MCH directly inhibited dopaminergic and GABAergic cells but did not regulate the activity of glutamatergic cells. Rather, MCH produced a delayed increase in excitatory input to dopamine cells and a corresponding decrease in GABAergic input to glutamatergic VTA cells. Our findings suggested that MCH may acutely suppress dopamine release while disinhibiting local glutamatergic signaling to restore dopamine levels. This indicated that the VTA is a target of MCH action, which may provide bidirectional regulation of energy balance.
Collapse
Affiliation(s)
- Carl Duncan Spencer
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Persephone A Miller
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | | | - Ralitsa G Nikolova
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Melissa J Chee
- Department of Neuroscience, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| |
Collapse
|
5
|
Concetti C, Viskaitis P, Grujic N, Duss SN, Privitera M, Bohacek J, Peleg-Raibstein D, Burdakov D. Exploratory Rearing Is Governed by Hypothalamic Melanin-Concentrating Hormone Neurons According to Locus Ceruleus. J Neurosci 2024; 44:e0015242024. [PMID: 38575343 PMCID: PMC11112542 DOI: 10.1523/jneurosci.0015-24.2024] [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: 01/03/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
Information seeking, such as standing on tiptoes to look around in humans, is observed across animals and helps survival. Its rodent analog-unsupported rearing on hind legs-was a classic model in deciphering neural signals of cognition and is of intense renewed interest in preclinical modeling of neuropsychiatric states. Neural signals and circuits controlling this dedicated decision to seek information remain largely unknown. While studying subsecond timing of spontaneous behavioral acts and activity of melanin-concentrating hormone (MCH) neurons (MNs) in behaving male and female mice, we observed large MN activity spikes that aligned to unsupported rears. Complementary causal, loss and gain of function, analyses revealed specific control of rear frequency and duration by MNs and MCHR1 receptors. Activity in a key stress center of the brain-the locus ceruleus noradrenaline cells-rapidly inhibited MNs and required functional MCH receptors for its endogenous modulation of rearing. By defining a neural module that both tracks and controls rearing, these findings may facilitate further insights into biology of information seeking.
Collapse
Affiliation(s)
- Cristina Concetti
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Paulius Viskaitis
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Nikola Grujic
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Sian N Duss
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Mattia Privitera
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Johannes Bohacek
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Daria Peleg-Raibstein
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Denis Burdakov
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| |
Collapse
|
6
|
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
|
7
|
Calafate S, Özturan G, Thrupp N, Vanderlinden J, Santa-Marinha L, Morais-Ribeiro R, Ruggiero A, Bozic I, Rusterholz T, Lorente-Echeverría B, Dias M, Chen WT, Fiers M, Lu A, Vlaeminck I, Creemers E, Craessaerts K, Vandenbempt J, van Boekholdt L, Poovathingal S, Davie K, Thal DR, Wierda K, Oliveira TG, Slutsky I, Adamantidis A, De Strooper B, de Wit J. Early alterations in the MCH system link aberrant neuronal activity and sleep disturbances in a mouse model of Alzheimer's disease. Nat Neurosci 2023:10.1038/s41593-023-01325-4. [PMID: 37188873 DOI: 10.1038/s41593-023-01325-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 04/10/2023] [Indexed: 05/17/2023]
Abstract
Early Alzheimer's disease (AD) is associated with hippocampal hyperactivity and decreased sleep quality. Here we show that homeostatic mechanisms transiently counteract the increased excitatory drive to CA1 neurons in AppNL-G-F mice, but that this mechanism fails in older mice. Spatial transcriptomics analysis identifies Pmch as part of the adaptive response in AppNL-G-F mice. Pmch encodes melanin-concentrating hormone (MCH), which is produced in sleep-active lateral hypothalamic neurons that project to CA1 and modulate memory. We show that MCH downregulates synaptic transmission, modulates firing rate homeostasis in hippocampal neurons and reverses the increased excitatory drive to CA1 neurons in AppNL-G-F mice. AppNL-G-F mice spend less time in rapid eye movement (REM) sleep. AppNL-G-F mice and individuals with AD show progressive changes in morphology of CA1-projecting MCH axons. Our findings identify the MCH system as vulnerable in early AD and suggest that impaired MCH-system function contributes to aberrant excitatory drive and sleep defects, which can compromise hippocampus-dependent functions.
Collapse
Affiliation(s)
- Sara Calafate
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium.
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Gökhan Özturan
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Nicola Thrupp
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Jeroen Vanderlinden
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Luísa Santa-Marinha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rafaela Morais-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Antonella Ruggiero
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ivan Bozic
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| | - Thomas Rusterholz
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Blanca Lorente-Echeverría
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Marcelo Dias
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Wei-Ting Chen
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Mark Fiers
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Ashley Lu
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Ine Vlaeminck
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Eline Creemers
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Katleen Craessaerts
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Joris Vandenbempt
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Luuk van Boekholdt
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
- KU Leuven, Department of Otorhinolaryngology, Leuven, Belgium
| | - Suresh Poovathingal
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Kristofer Davie
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Dietmar Rudolf Thal
- Department of Imaging and Pathology, Laboratory of Neuropathology, and Leuven Brain Institute, KU-Leuven, O&N IV, Leuven, Belgium
- Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Keimpe Wierda
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Antoine Adamantidis
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Bart De Strooper
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium.
- UK Dementia Research Institute (UK DRI@UCL) at University College London, London, UK.
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium.
| |
Collapse
|
8
|
Rahman MM, Islam MR, Supti FA, Dhar PS, Shohag S, Ferdous J, Shuvo SK, Akter A, Hossain MS, Sharma R. Exploring the Therapeutic Effect of Neurotrophins and Neuropeptides in Neurodegenerative Diseases: at a Glance. Mol Neurobiol 2023:10.1007/s12035-023-03328-5. [PMID: 37052791 DOI: 10.1007/s12035-023-03328-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/22/2023] [Indexed: 04/14/2023]
Abstract
Neurotrophins and neuropeptides are the essential regulators of peripheral nociceptive nerves that help to induce, sensitize, and maintain pain. Neuropeptide has a neuroprotective impact as it increases trophic support, regulates calcium homeostasis, and reduces excitotoxicity and neuroinflammation. In contrast, neurotrophins target neurons afflicted by ischemia, epilepsy, depression, and eating disorders, among other neuropsychiatric conditions. Neurotrophins are reported to inhibit neuronal death. Strategies maintained for "brain-derived neurotrophic factor (BDNF) therapies" are to upregulate BDNF levels using the delivery of protein and genes or compounds that target BDNF production and boosting BDNF signals by expanding with BDNF mimetics. This review discusses the mechanisms of neurotrophins and neuropeptides against acute neural damage as well as highlighting neuropeptides as a potential therapeutic agent against Parkinson's disease (PD), Huntington's disease (HD), Alzheimer's disease (AD), and Machado-Joseph disease (MJD), the signaling pathways affected by neurotrophins and their receptors in both standard and diseased CNS systems, and future perspectives that can lead to the potent application of neurotrophins and neuropeptides in neurodegenerative diseases (NDs).
Collapse
Affiliation(s)
- Md Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Fatema Akter Supti
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Puja Sutro Dhar
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Sheikh Shohag
- Department of Genetic Engineering and Biotechnology, Faculty of Earth and Ocean Science, Bangabandhu Sheikh Mujibur Rahman Maritime University, Mirpur 12, Dhaka, 1216, Bangladesh
| | - Jannatul Ferdous
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Shakil Khan Shuvo
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Aklima Akter
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Md Sarowar Hossain
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Rohit Sharma
- Department of Rasa Shastra & Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
9
|
Stutz B, Waterson MJ, Šestan-Peša M, Dietrich MO, Škarica M, Sestan N, Racz B, Magyar A, Sotonyi P, Liu ZW, Gao XB, Matyas F, Stoiljkovic M, Horvath TL. AgRP neurons control structure and function of the medial prefrontal cortex. Mol Psychiatry 2022; 27:3951-3960. [PMID: 35906488 PMCID: PMC9891653 DOI: 10.1038/s41380-022-01691-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 02/07/2023]
Abstract
Hypothalamic agouti-related peptide and neuropeptide Y-expressing (AgRP) neurons have a critical role in both feeding and non-feeding behaviors of newborn, adolescent, and adult mice, suggesting their broad modulatory impact on brain functions. Here we show that constitutive impairment of AgRP neurons or their peripubertal chemogenetic inhibition resulted in both a numerical and functional reduction of neurons in the medial prefrontal cortex (mPFC) of mice. These changes were accompanied by alteration of oscillatory network activity in mPFC, impaired sensorimotor gating, and altered ambulatory behavior that could be reversed by the administration of clozapine, a non-selective dopamine receptor antagonist. The observed AgRP effects are transduced to mPFC in part via dopaminergic neurons in the ventral tegmental area and may also be conveyed by medial thalamic neurons. Our results unmasked a previously unsuspected role for hypothalamic AgRP neurons in control of neuronal pathways that regulate higher-order brain functions during development and in adulthood.
Collapse
Affiliation(s)
- Bernardo Stutz
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA
| | - Michael J Waterson
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA
| | - Matija Šestan-Peša
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA
| | - Marcelo O Dietrich
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Mario Škarica
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Nenad Sestan
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Bence Racz
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Aletta Magyar
- Institute of Cognitive Neuroscience and Psychology, Research Center for Natural Sciences, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Peter Sotonyi
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Zhong-Wu Liu
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA
| | - Xiao-Bing Gao
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA
| | - Ferenc Matyas
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
- Institute of Cognitive Neuroscience and Psychology, Research Center for Natural Sciences, Budapest, Hungary
- Institute of Experimental Medicine, Budapest, Hungary
| | - Milan Stoiljkovic
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA.
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary.
| |
Collapse
|
10
|
Shao YQ, Fan L, Wu WY, Zhu YJ, Xu HT. A developmental switch between electrical and neuropeptide communication in the ventromedial hypothalamus. Curr Biol 2022; 32:3137-3145.e3. [PMID: 35659861 DOI: 10.1016/j.cub.2022.05.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/15/2022] [Accepted: 05/11/2022] [Indexed: 12/29/2022]
Abstract
Dissecting neural connectivity patterns within local brain regions is an essential step to understanding the function of the brain.1 Neural microcircuits in brain regions, such as the neocortex and the hippocampus, have been extensively studied.2 By contrast, the microcircuit in the hypothalamus remains largely uncharacterized. The hypothalamus is crucial for animals' survival and reproduction.3 Knowledge of how different hypothalamic nuclei coordinate with each other and outside brain regions for hypothalamus-related functions has been significantly advanced.4-9 Although there are limited studies on the neural microcircuit in the lateral hypothalamus (LHA)10,11 and the suprachiasmatic nucleus (SCN),12,13 the patterns of neural microcircuits in most of the given hypothalamic nuclei remain largely unknown. This study applied combinatory approaches to address the local neural circuit pattern in the ventromedial hypothalamus (VMH) and other hypothalamic nuclei. We discovered a unique neural circuit design in the VMH. Neurons in the VMH were electrically coupled at the early postnatal stage like ones in the neocortex.14 However, unlike neocortical neurons,14,15 they developed very few chemical synapses after the disappearance of electrical synapses. Instead, VMH neurons communicated with neuropeptides. The similar scarceness of synaptic connectivity found in other hypothalamic nuclei further indicated that the lack of synaptic connections is a unique feature for local neural circuits in most adult hypothalamic nuclei. Thus, our findings provide a solid synaptic basis at the cellular level to understand hypothalamic functions better.
Collapse
Affiliation(s)
- Yin-Qi Shao
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liu Fan
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wen-Yan Wu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi-Jun Zhu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hua-Tai Xu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China.
| |
Collapse
|
11
|
Janković SM, Đešević M. Advancements in neuroactive peptides in seizures. Expert Rev Neurother 2022; 22:129-143. [DOI: 10.1080/14737175.2022.2031983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Slobodan M. Janković
- - University of Kragujevac, Faculty of Medical Sciences, Kragujevac, Serbia
- University Clinical Center, Kragujevac, Serbia
| | - Miralem Đešević
- - Private Policlinic Center Eurofarm Sarajevo, Cardiology Department, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
12
|
Adamantidis AR, Schmidt MH, Carter ME, Burdakov D, Peyron C, Scammell TE. A circuit perspective on narcolepsy. Sleep 2021; 43:5699663. [PMID: 31919524 PMCID: PMC7215265 DOI: 10.1093/sleep/zsz296] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/13/2019] [Indexed: 01/25/2023] Open
Abstract
The sleep disorder narcolepsy is associated with symptoms related to either boundary state control that include excessive daytime sleepiness and sleep fragmentation, or rapid eye movement (REM) sleep features including cataplexy, sleep paralysis, hallucinations, and sleep-onset REM sleep events (SOREMs). Although the loss of Hypocretin/Orexin (Hcrt/Ox) peptides or their receptors have been associated with the disease, here we propose a circuit perspective of the pathophysiological mechanisms of these narcolepsy symptoms that encompasses brain regions, neuronal circuits, cell types, and transmitters beyond the Hcrt/Ox system. We further discuss future experimental strategies to investigate brain-wide mechanisms of narcolepsy that will be essential for a better understanding and treatment of the disease.
Collapse
Affiliation(s)
- A R Adamantidis
- Department of Neurology, Centre for Experimental Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland.,Department of Biomedical Research, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| | - M H Schmidt
- Department of Neurology, Centre for Experimental Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland.,Ohio Sleep Medicine Institute, Dublin, OH
| | - M E Carter
- Department of Biology, Program in Neuroscience, Williams College, Williamstown, MA
| | - D Burdakov
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - C Peyron
- Center for Research in Neuroscience of Lyon, SLEEP team, CNRS UMR5292, INSERM U1028, University Lyon 1, Bron, France
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| |
Collapse
|
13
|
Concetti C, Burdakov D. Orexin/Hypocretin and MCH Neurons: Cognitive and Motor Roles Beyond Arousal. Front Neurosci 2021; 15:639313. [PMID: 33828450 PMCID: PMC8019792 DOI: 10.3389/fnins.2021.639313] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/01/2021] [Indexed: 02/01/2023] Open
Abstract
The lateral hypothalamus (LH) is classically implicated in sleep-wake control. It is the main source of orexin/hypocretin and melanin-concentrating hormone (MCH) neuropeptides in the brain, which have been both implicated in arousal state switching. These neuropeptides are produced by non-overlapping LH neurons, which both project widely throughout the brain, where release of orexin and MCH activates specific postsynaptic G-protein-coupled receptors. Optogenetic manipulations of orexin and MCH neurons during sleep indicate that they promote awakening and REM sleep, respectively. However, recordings from orexin and MCH neurons in awake, moving animals suggest that they also act outside sleep/wake switching. Here, we review recent studies showing that both orexin and MCH neurons can rapidly (sub-second-timescale) change their firing when awake animals experience external stimuli, or during self-paced exploration of objects and places. However, the sensory-behavioral correlates of orexin and MCH neural activation can be quite different. Orexin neurons are generally more dynamic, with about 2/3rds of them activated before and during self-initiated running, and most activated by sensory stimulation across sensory modalities. MCH neurons are activated in a more select manner, for example upon self-paced investigation of novel objects and by certain other novel stimuli. We discuss optogenetic and chemogenetic manipulations of orexin and MCH neurons, which combined with pharmacological blockade of orexin and MCH receptors, imply that these rapid LH dynamics shape fundamental cognitive and motor processes due to orexin and MCH neuropeptide actions in the awake brain. Finally, we contemplate whether the awake control of psychomotor brain functions by orexin and MCH are distinct from their “arousal” effects.
Collapse
Affiliation(s)
- Cristina Concetti
- Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Denis Burdakov
- Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
14
|
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
|
15
|
Diniz GB, Battagello DS, Klein MO, Bono BSM, Ferreira JGP, Motta‐Teixeira LC, Duarte JCG, Presse F, Nahon J, Adamantidis A, Chee MJ, Sita LV, Bittencourt JC. Ciliary melanin‐concentrating hormone receptor 1 (MCHR1) is widely distributed in the murine CNS in a sex‐independent manner. J Neurosci Res 2020; 98:2045-2071. [DOI: 10.1002/jnr.24651] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/24/2020] [Accepted: 05/07/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Giovanne B. Diniz
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
- Department of Neurosurgery Yale School of Medicine New Haven CT USA
| | - Daniella S. Battagello
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
| | - Marianne O. Klein
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
| | | | - Jozélia G. P. Ferreira
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
| | - Livia C. Motta‐Teixeira
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
| | - Jessica C. G. Duarte
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
| | - Françoise Presse
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC) Université Côte d’AzurCNRS Valbonne France
| | - Jean‐Louis Nahon
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC) Université Côte d’AzurCNRS Valbonne France
| | | | - Melissa J. Chee
- Department of Neuroscience Carleton University Ottawa ON Canada
| | - Luciane V. Sita
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
| | - Jackson C. Bittencourt
- Department of Anatomy Institute of Biomedical Sciences University of Sao Paulo Sao Paulo Brazil
- Center for Neuroscience and Behavior Institute of Psychology University of Sao Paulo Sao Paulo Brazil
| |
Collapse
|
16
|
Wang D, Zhang J, Bai Y, Zheng X, Alizamini MM, Shang W, Yang Q, Li M, Li Y, Sui N. Melanin-concentrating hormone in rat nucleus accumbens or lateral hypothalamus differentially impacts morphine and food seeking behaviors. J Psychopharmacol 2020; 34:478-489. [PMID: 31909693 DOI: 10.1177/0269881119895521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Identifying neural substrates that are differentially affected by drugs of abuse and natural rewards is key to finding a target for an efficacious treatment for substance abuse. Melanin-concentrating hormone is a polypeptide with an inhibitory effect on the mesolimbic dopamine system. Here we test the hypothesis that melanin-concentrating hormone in the lateral hypothalamus and nucleus accumbens shell is differentially involved in the regulation of morphine and food-rewarded behaviors. METHODS Male Sprague-Dawley rats were trained with morphine (5.0 mg/kg, subcutaneously) or food pellets (standard chow, 10-14 g) to induce a conditioned place preference, immediately followed by extinction training. Melanin-concentrating hormone (1.0 µg/side) or saline was infused into the nucleus accumbens shell or lateral hypothalamus before the reinstatement primed by morphine or food, and locomotor activity was simultaneously monitored. As the comparison, melanin-concentrating hormone was also microinjected into the nucleus accumbens shell or lateral hypothalamus before the expression of food or morphine-induced conditioned place preference. RESULTS Microinfusion of melanin-concentrating hormone into the nucleus accumbens shell (but not into the lateral hypothalamus) prevented the reinstatement of morphine conditioned place preference but had no effect on the reinstatement of food conditioned place preference. In contrast, microinfusion of melanin-concentrating hormone into the lateral hypothalamus (but not in the nucleus accumbens shell) inhibited the reinstatement of food conditioned place preference but had no effect on the reinstatement of morphine conditioned place preference. CONCLUSIONS These results suggest a clear double dissociation of melanin-concentrating hormone in morphine/food rewarding behaviors and melanin-concentrating hormone in the nucleus accumbens shell. Melanin-concentrating hormone could be a potential target for therapeutic intervention for morphine abuse without affecting natural rewards.
Collapse
Affiliation(s)
- Dongmei Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Jianjun Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Yunjing Bai
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Xigeng Zheng
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Mirmohammadali M Alizamini
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Wen Shang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Qingxiong Yang
- School of Karst Science, Guizhou Normal University/State Engineering Technology Institute for Karst Desertification Control, Guiyang, China
| | - Ming Li
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Yonghui Li
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Nan Sui
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
17
|
Chen Q, Qu K, Ma Z, Zhan J, Zhang F, Shen J, Ning Q, Jia P, Zhang J, Chen N, Chen H, Huang B, Lei C. Genome-Wide Association Study Identifies Genomic Loci Associated With Neurotransmitter Concentration in Cattle. Front Genet 2020; 11:139. [PMID: 32292413 PMCID: PMC7119242 DOI: 10.3389/fgene.2020.00139] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 02/06/2020] [Indexed: 11/30/2022] Open
Abstract
Abnormal neurotransmitter concentration is one of the factors that affect the health status, behavioral personality, and welfare level of animals, but the genetic basis of the abnormality is still largely unknown. The objective of this study is to identify putative genomic loci associated with neurotransmitter concentration in cattle. We measured serotonin (5HT), dopamine (DA), cortisol, glutamate (Glu), and ACTH concentrations in blood serum using double-antibody sandwich ELISA in 30 Brahman cattle and 127 Yunling cattle. Interestingly, we found that ACTH concentration was positively correlated with body weight, cannon circumference, and hip width (P < 0.05). Genome-wide association study (GWAS) was performed with mixed linear models using autosomal SNPs derived from the whole-genome sequence. We identified five, five, two, three, and five suggestive loci associated with 5HT, DA, cortisol, Glu, and ACTH concentration, respectively. These 20 associated loci implicated 18 candidate genes. For Glu concentration, the most significant association locus was assigned to MCHR1, a G-coupled receptor that could modulate glutamate release. For dopamine concentration, a very strong association locus was located in the intron of SLC18A2, which is a critical mediator of dopamine dynamics. However, for ACTH concentration, a very strong association locus was assigned to HTR1F, a G protein-coupled receptor that can influence the release of ACTH. Other candidate genes of interest identified for neurotransmitter concentration were PRMT6, GADD45A, PCCA, ANGPT1, ACCS, LOC100336971, TNR, GSDMA, CNTN3, CARMIL1, CDKAL1, RBFOX1, PCDH15, and LGALS12. Our findings will provide targets for the genetic improvement of neurotransmitter-related traits in domestic cattle and basic materials for studying the mechanism of neurotransmitter synthesis, release, and transport in human and animals.
Collapse
Affiliation(s)
- Qiuming Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Kaixing Qu
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Zhijie Ma
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Jingxi Zhan
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Fengwei Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jiafei Shen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Qingqing Ning
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Peng Jia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jicai Zhang
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Ningbo Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hong Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Bizhi Huang
- Yunnan Academy of Grassland and Animal Science, Kunming, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
18
|
Fakhoury M, Salman I, Najjar W, Merhej G, Lawand N. The Lateral Hypothalamus: An Uncharted Territory for Processing Peripheral Neurogenic Inflammation. Front Neurosci 2020; 14:101. [PMID: 32116534 PMCID: PMC7029733 DOI: 10.3389/fnins.2020.00101] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/24/2020] [Indexed: 12/20/2022] Open
Abstract
The roles of the hypothalamus and particularly the lateral hypothalamus (LH) in the regulation of inflammation and pain have been widely studied. The LH consists of a parasympathetic area that has connections with all the major parts of the brain. It controls the autonomic nervous system (ANS), regulates feeding behavior and wakeful cycles, and is a part of the reward system. In addition, it contains different types of neurons, most importantly the orexin neurons. These neurons, though few in number, perform critical functions such as inhibiting pain transmission and interfering with the reward system, feeding behavior and the hypothalamic pituitary axis (HPA). Recent evidence has identified a new role for orexin neurons in the modulation of pain transmission associated with several inflammatory diseases, including rheumatoid arthritis and ulcerative colitis. Here, we review recent findings on the various physiological functions of the LH with special emphasis on the orexin/receptor system and its role in mediating inflammatory pain.
Collapse
Affiliation(s)
- Marc Fakhoury
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Israa Salman
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Wassim Najjar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - George Merhej
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nada Lawand
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Neurology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
19
|
The role of co-neurotransmitters in sleep and wake regulation. Mol Psychiatry 2019; 24:1284-1295. [PMID: 30377299 PMCID: PMC6491268 DOI: 10.1038/s41380-018-0291-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 09/17/2018] [Accepted: 10/08/2018] [Indexed: 12/11/2022]
Abstract
Sleep and wakefulness control in the mammalian brain requires the coordination of various discrete interconnected neurons. According to the most conventional sleep model, wake-promoting neurons (WPNs) and sleep-promoting neurons (SPNs) compete for network dominance, creating a systematic "switch" that results in either the sleep or awake state. WPNs and SPNs are ubiquitous in the brainstem and diencephalon, areas that together contain <1% of the neurons in the human brain. Interestingly, many of these WPNs and SPNs co-express and co-release various types of the neurotransmitters that often have opposing modulatory effects on the network. Co-transmission is often beneficial to structures with limited numbers of neurons because it provides increasing computational capability and flexibility. Moreover, co-transmission allows subcortical structures to bi-directionally control postsynaptic neurons, thus helping to orchestrate several complex physiological functions such as sleep. Here, we present an in-depth review of co-transmission in hypothalamic WPNs and SPNs and discuss its functional significance in the sleep-wake network.
Collapse
|
20
|
Urbanavicius J, Fabius S, Roncalho A, Joca S, Torterolo P, Scorza C. Melanin-concentrating hormone in the Locus Coeruleus aggravates helpless behavior in stressed rats. Behav Brain Res 2019; 374:112120. [PMID: 31376444 DOI: 10.1016/j.bbr.2019.112120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/17/2019] [Accepted: 07/30/2019] [Indexed: 10/26/2022]
Abstract
Animal studies have shown that antagonists of receptor 1 of Melanin-Concentrating Hormone (MCH-R1) elicit antidepressive-like behavior, suggesting that MCH-R1 might be a novel target for the treatment of depression and supports the hypothesis that MCHergic signaling regulates depressive-like behaviors. Consistent with the evidence that MCHergic neurons send projections to dorsal and median raphe nuclei, we have previously demonstrated that MCH microinjections in both nuclei induced a depressive-like behavior. Even though MCH neurons also project to Locus Coeruleus (LC), only a few studies have reported the behavioral and neurochemical effect of MCH into the LC. We studied the effects of MCH (100 and 200 ng) into the LC on coping-stress related behaviors associated with depression, using two different behavioral tests: the forced swimming test (FST) and the learned helplessness (LH). To characterize the functional interaction between MCH and the noradrenergic LC system, we also evaluated the neurochemical effects of MCH (100 ng) on the extracellular levels of noradrenaline (NA) in the medial prefrontal cortex (mPFC), an important LC terminal region involved in emotional processing. MCH administration into the LC elicited a depressive-like behavior evidenced in both paradigms. Interestingly, in the LH, MCH (100) elicited a significant increase in escape failures only in stressed animals. A significant decrease in prefrontal levels of NA was observed after MCH microinjection into the LC. Our results demonstrate that increased MCH signaling into the LC triggers depressive-like behaviors, especially in stressed animals. These data further corroborate the important role of MCH in the neurobiology of depression.
Collapse
Affiliation(s)
- Jessika Urbanavicius
- Departament of Experimental Neuropharmacology, Instituto de Investigaciones Biológicas Clemente Estable, Uruguay
| | - Sara Fabius
- Departament of Experimental Neuropharmacology, Instituto de Investigaciones Biológicas Clemente Estable, Uruguay
| | - Aline Roncalho
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Samia Joca
- Department of Physics and Chemistry, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, Brazil
| | - Pablo Torterolo
- Department of Physiology, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Cecilia Scorza
- Departament of Experimental Neuropharmacology, Instituto de Investigaciones Biológicas Clemente Estable, Uruguay.
| |
Collapse
|
21
|
Bansal R, Engle SE, Antonellis PJ, Whitehouse LS, Baucum AJ, Cummins TR, Reiter JF, Berbari NF. Hedgehog Pathway Activation Alters Ciliary Signaling in Primary Hypothalamic Cultures. Front Cell Neurosci 2019; 13:266. [PMID: 31249512 PMCID: PMC6582312 DOI: 10.3389/fncel.2019.00266] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/29/2019] [Indexed: 11/13/2022] Open
Abstract
Primary cilia dysfunction has been associated with hyperphagia and obesity in both ciliopathy patients and mouse models of cilia perturbation. Neurons throughout the brain possess these solitary cellular appendages, including in the feeding centers of the hypothalamus. Several cell biology questions associated with primary neuronal cilia signaling are challenging to address in vivo. Here we utilize primary hypothalamic neuronal cultures to study ciliary signaling in relevant cell types. Importantly, these cultures contain neuronal populations critical for appetite and satiety such as pro-opiomelanocortin (POMC) and agouti related peptide (AgRP) expressing neurons and are thus useful for studying signaling involved in feeding behavior. Correspondingly, these cultured neurons also display electrophysiological activity and respond to both local and peripheral signals that act on the hypothalamus to influence feeding behaviors, such as leptin and melanin concentrating hormone (MCH). Interestingly, we found that cilia mediated hedgehog signaling, generally associated with developmental processes, can influence ciliary GPCR signaling (Mchr1) in terminally differentiated neurons. Specifically, pharmacological activation of the hedgehog-signaling pathway using the smoothened agonist, SAG, attenuated the ability of neurons to respond to ligands (MCH) of ciliary GPCRs. Understanding how the hedgehog pathway influences cilia GPCR signaling in terminally differentiated neurons could reveal the molecular mechanisms associated with clinical features of ciliopathies, such as hyperphagia-associated obesity.
Collapse
Affiliation(s)
- Ruchi Bansal
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, United States
| | - Staci E Engle
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, United States
| | - Patrick J Antonellis
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, United States
| | - Logan S Whitehouse
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, United States
| | - Anthony J Baucum
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, United States.,Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Theodore R Cummins
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, United States.,Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Nicolas F Berbari
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, United States.,Stark Neurosciences Research Institute, Indianapolis, IN, United States.,Center for Diabetes and Metabolic Disorders Research, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
22
|
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
|
23
|
Sabetghadam A, Grabowiecka-Nowak A, Kania A, Gugula A, Blasiak E, Blasiak T, Ma S, Gundlach AL, Blasiak A. Melanin-concentrating hormone and orexin systems in rat nucleus incertus: Dual innervation, bidirectional effects on neuron activity, and differential influences on arousal and feeding. Neuropharmacology 2018; 139:238-256. [DOI: 10.1016/j.neuropharm.2018.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/20/2018] [Accepted: 07/04/2018] [Indexed: 12/24/2022]
|
24
|
Li S, Yip A, Bird J, Seok BS, Chan A, Godden KE, Tam LD, Ghelardoni S, Balaban E, Martinez-Gonzalez D, Pompeiano M. Melanin-concentrating hormone (MCH) neurons in the developing chick brain. Brain Res 2018; 1700:19-30. [PMID: 30420052 DOI: 10.1016/j.brainres.2018.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/27/2018] [Accepted: 07/01/2018] [Indexed: 01/09/2023]
Abstract
The present study was undertaken because no previous developmental studies exist on MCH neurons in any avian species. After validating a commercially-available antibody for use in chickens, immunohistochemical examinations first detected MCH neurons around embryonic day (E) 8 in the posterior hypothalamus. This population increased thereafter, reaching a numerical maximum by E20. MCH-positive cell bodies were found only in the posterior hypothalamus at all ages examined, restricted to a region showing very little overlap with the locations of hypocretin/orexin (H/O) neurons. Chickens had fewer MCH than H/O neurons, and MCH neurons also first appeared later in development than H/O neurons (the opposite of what has been found in rodents). MCH neurons appeared to originate from territories within the hypothalamic periventricular organ that partially overlap with the source of diencephalic serotonergic neurons. Chicken MCH fibers developed exuberantly during the second half of embryonic development, and they became abundant in the same brain areas as in rodents, including the hypothalamus (by E12), locus coeruleus (by E12), dorsal raphe nucleus (by E20) and septum (by E20). These observations suggest that MCH cells may play different roles during development in chickens and rodents; but once they have developed, MCH neurons exhibit similar phenotypes in birds and rodents.
Collapse
Affiliation(s)
- SiHan Li
- Department of Psychology, McGill University, Montreal, QC H3A 1B1, Canada
| | - Alissa Yip
- Department of Psychology, McGill University, Montreal, QC H3A 1B1, Canada
| | - Jaimie Bird
- Department of Psychology, McGill University, Montreal, QC H3A 1B1, Canada
| | - Bong Soo Seok
- Department of Psychology, McGill University, Montreal, QC H3A 1B1, Canada
| | - Aimee Chan
- Department of Psychology, McGill University, Montreal, QC H3A 1B1, Canada
| | - Kyle E Godden
- Department of Psychology, McGill University, Montreal, QC H3A 1B1, Canada
| | - Laurel D Tam
- Department of Psychology, McGill University, Montreal, QC H3A 1B1, Canada
| | | | - Evan Balaban
- Department of Psychology, McGill University, Montreal, QC H3A 1B1, Canada
| | | | - Maria Pompeiano
- Department of Psychology, McGill University, Montreal, QC H3A 1B1, Canada.
| |
Collapse
|
25
|
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
|
26
|
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]
|
27
|
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]
|
28
|
Diniz GB, Bittencourt JC. The Melanin-Concentrating Hormone as an Integrative Peptide Driving Motivated Behaviors. Front Syst Neurosci 2017; 11:32. [PMID: 28611599 PMCID: PMC5447028 DOI: 10.3389/fnsys.2017.00032] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/04/2017] [Indexed: 12/14/2022] Open
Abstract
The melanin-concentrating hormone (MCH) is an important peptide implicated in the control of motivated behaviors. History, however, made this peptide first known for its participation in the control of skin pigmentation, from which its name derives. In addition to this peripheral role, MCH is strongly implicated in motivated behaviors, such as feeding, drinking, mating and, more recently, maternal behavior. It is suggested that MCH acts as an integrative peptide, converging sensory information and contributing to a general arousal of the organism. In this review, we will discuss the various aspects of energy homeostasis to which MCH has been associated to, focusing on the different inputs that feed the MCH peptidergic system with information regarding the homeostatic status of the organism and the exogenous sensory information that drives this system, as well as the outputs that allow MCH to act over a wide range of homeostatic and behavioral controls, highlighting the available morphological and hodological aspects that underlie these integrative actions. Besides the well-described role of MCH in feeding behavior, a prime example of hypothalamic-mediated integration, we will also examine those functions in which the participation of MCH has not yet been extensively characterized, including sexual, maternal, and defensive behaviors. We also evaluated the available data on the distribution of MCH and its function in the context of animals in their natural environment. Finally, we briefly comment on the evidence for MCH acting as a coordinator between different modalities of motivated behaviors, highlighting the most pressing open questions that are open for investigations and that could provide us with important insights about hypothalamic-dependent homeostatic integration.
Collapse
Affiliation(s)
- Giovanne B. Diniz
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São PauloSão Paulo, Brazil
| | - Jackson C. Bittencourt
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São PauloSão Paulo, Brazil
- Center for Neuroscience and Behavior, Institute of Psychology, University of São PauloSão Paulo, Brazil
| |
Collapse
|
29
|
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
|
30
|
Ono D, Yamanaka A. Hypothalamic regulation of the sleep/wake cycle. Neurosci Res 2017; 118:74-81. [PMID: 28526553 DOI: 10.1016/j.neures.2017.03.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/02/2017] [Accepted: 03/13/2017] [Indexed: 12/30/2022]
Abstract
Sleep is one of the most important physiological functions in mammals. It is regulated by not only homeostatic regulation but also circadian clock. Several neuropeptide-producing neurons located in the hypothalamus are implicated in the regulation of sleep/wakefulness. Among them, orexin/hypocretin-producing neurons (orexin neurons) are a crucial component for maintenance of wakefulness, because lack of orexin function results in narcolepsy, which is a sleep disorder. Recent findings have identified substances that excite or inhibit neural activity of orexin neurons. Furthermore neural projections of the neurons which release these substances have been revealed. In addition to orexin, melanin concentrating hormone (MCH)-producing neurons in the lateral hypothalamic area (LHA) are also implicated in the regulation of sleep/wakefulness. MCH neurons are active during sleep but become silent during wakefulness. Recently developed innovative methods including optogenetics and pharmacogenetics have provided substantial insights into the regulation of sleep/wakefulness. In vivo optical recordings and retrograde and anterograde tracing methods will allow us to understand additional details regarding important interactions between these two types of neurons in the LHA and other neurons in the brain. Finally we discuss the circadian clock and sleep/wake cycle. Understanding of the neural networks and its circadian modulation of sleep/wake cycles remain to be investigated.
Collapse
Affiliation(s)
- Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan.
| |
Collapse
|
31
|
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
|
32
|
Abstract
Many of the neurocircuits and hormones known to underlie the sensations of hunger and satiety also substantially alter the activity of the dopaminergic reward system. Much interest lies in the ways that hunger, satiety, and reward tie together, as the epidemic of obesity seems tied to the recent development and mass availability of highly palatable foods. In this review, we will first discuss the basic neurocircuitry of the midbrain and basal forebrain reward system. We will elaborate how several important mediators of hunger-the agouti-related protein neurons of the arcuate nucleus, the lateral hypothalamic nucleus, and ghrelin-enhance the sensitivity of the dopaminergic reward system. Then, we will elaborate how mediators of satiety-the nucleus tractus solitarius, pro-opiomelanocortin neurons of the arcuate nucleus, and its peripheral hormonal influences such as leptin-reduce the reward system sensitivity. We hope to provide a template by which future research may identify the ways in which highly rewarding foods bypass this balanced system to produce excessive food consumption.
Collapse
Affiliation(s)
- Ryan Michael Cassidy
- Brown Foundation of the Institute of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, Neuroscience Program MD Anderson Cancer Center and UTHealth Graduate School of Biological Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
- *Correspondence: Ryan Michael Cassidy,
| | - Qingchun Tong
- Brown Foundation of the Institute of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, Neuroscience Program MD Anderson Cancer Center and UTHealth Graduate School of Biological Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
33
|
Skrapits K, Kanti V, Savanyú Z, Maurnyi C, Szenci O, Horváth A, Borsay BÁ, Herczeg L, Liposits Z, Hrabovszky E. Lateral hypothalamic orexin and melanin-concentrating hormone neurons provide direct input to gonadotropin-releasing hormone neurons in the human. Front Cell Neurosci 2015; 9:348. [PMID: 26388735 PMCID: PMC4559643 DOI: 10.3389/fncel.2015.00348] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 08/20/2015] [Indexed: 12/03/2022] Open
Abstract
Hypophysiotropic projections of gonadotropin-releasing hormone (GnRH)-synthesizing neurons form the final common output way of the hypothalamus in the neuroendocrine control of reproduction. Several peptidergic neuronal systems of the medial hypothalamus innervate human GnRH cells and mediate crucially important hormonal and metabolic signals to the reproductive axis, whereas much less is known about the contribution of the lateral hypothalamic area to the afferent control of human GnRH neurons. Orexin (ORX)- and melanin-concentrating hormone (MCH)-synthesizing neurons of this region have been implicated in diverse behavioral and autonomic processes, including sleep and wakefulness, feeding and other functions. In the present immunohistochemical study, we addressed the anatomical connectivity of these neurons to human GnRH cells in post-mortem hypothalamic samples obtained from autopsies. We found that 38.9 ± 10.3% and 17.7 ± 3.3% of GnRH-immunoreactive (IR) perikarya in the infundibular nucleus of human male subjects received ORX-IR and MCH-IR contacts, respectively. On average, each 1 mm segment of GnRH dendrites received 7.3 ± 1.1 ORX-IR and 3.7 ± 0.5 MCH-IR axo-dendritic appositions. Overall, the axo-dendritic contacts dominated over the axo-somatic contacts and represented 80.5 ± 6.4% of ORX-IR and 76.7 ± 4.6% of MCH-IR inputs to GnRH cells. Based on functional evidence from studies of laboratory animals, the direct axo-somatic and axo-dendritic input from ORX and MCH neurons to the human GnRH neuronal system may convey critical metabolic and other homeostatic signals to the reproducive axis. In this study, we also report the generation and characterization of new antibodies for immunohistochemical detection of GnRH neurons in histological sections.
Collapse
Affiliation(s)
- Katalin Skrapits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences Budapest, Hungary
| | - Vivien Kanti
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences Budapest, Hungary
| | - Zsófia Savanyú
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences Budapest, Hungary
| | - Csilla Maurnyi
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences Budapest, Hungary
| | - Ottó Szenci
- Department and Clinic for Production Animals, Faculty of Veterinary Science, Szent István University Üllő, Hungary ; MTA-SZIE Large Animal Clinical Research Group, Dóra major Üllő, Hungary
| | - András Horváth
- Department and Clinic for Production Animals, Faculty of Veterinary Science, Szent István University Üllő, Hungary
| | - Beáta Á Borsay
- Department of Forensic Medicine, Faculty of Medicine of the University of Debrecen Debrecen, Hungary
| | - László Herczeg
- Department of Forensic Medicine, Faculty of Medicine of the University of Debrecen Debrecen, Hungary
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences Budapest, Hungary ; Department of Neuroscience, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University Budapest, Hungary
| | - Erik Hrabovszky
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences Budapest, Hungary
| |
Collapse
|
34
|
Devera A, Pascovich C, Lagos P, Falconi A, Sampogna S, Chase MH, Torterolo P. Melanin-concentrating hormone (MCH) modulates the activity of dorsal raphe neurons. Brain Res 2015; 1598:114-28. [DOI: 10.1016/j.brainres.2014.12.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 12/09/2014] [Accepted: 12/13/2014] [Indexed: 12/27/2022]
|
35
|
Hamamoto A, Kobayashi Y, Saito Y. Identification of amino acids that are selectively involved in Gi/o activation by rat melanin-concentrating hormone receptor 1. Cell Signal 2015; 27:818-27. [PMID: 25617691 DOI: 10.1016/j.cellsig.2015.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 12/30/2014] [Accepted: 01/14/2015] [Indexed: 02/01/2023]
Abstract
Many G-protein-coupled receptors (GPCRs) are known to functionally couple to multiple G-protein subfamily members. Although promiscuous G-protein coupling enables GPCRs to mediate diverse signals, only a few GPCRs have been identified with differential determinants for coupling to distinct Gα proteins. Mammalian melanin-concentrating hormone receptor 1 (MCHR1) couples to dual G-protein subfamilies. However, the selectivity mechanisms between MCHR1 and different subtypes of Gα proteins are unclear. Our previous studies demonstrated that mammalian MCHR1 couples to both Gi/o and Gq, whereas goldfish MCHR1 exclusively couples to Gq. In this study, we analyzed multiple sequence alignments between rat and goldfish MCHR1s, and designed three multisubstituted mutants of rat MCHR1 by replacing corresponding residues with those in goldfish MCHR1, focusing on regions around the cytosolic intracellular loops. By measurement of intracellular Ca(2+) mobilization, we found that two MCHR1 mutants, i2_6sub and i3_6sub, which contained six simultaneously substituted residues in the second intracellular loop or a combination of substituted residues in the third intracellular loop and fifth transmembrane domain, respectively, significantly reduced Gi/o-sensitive pertussis toxin responsiveness without altering Gq-mediated activity. Analyses of 10 other substitutions revealed that the multiple substitutions in i2_6sub and i3_6sub were necessary for Gi/o-selective responses. As judged by Gi/o-dependent GTPγS binding and cyclic AMP assays, i2_6sub and i3_6sub elicited phenotypes for impaired Gi/o-mediated signaling. We also monitored the dynamic mass redistribution (DMR) in living cells, which reveals receptor activity as an optical trace containing activation of all GPCR coupling classes. Cells transfected with i2_6sub or i3_6sub exhibited reduced Gi/o-mediated DMR responses compared with those transfected with MCHR1. These data suggest that two different regions independently affect the Gi/o-protein preference, and that multiple residues comprise a conformation favoring Gi/o-protein coupling and subsequently result in Gi/o-selective signaling.
Collapse
Affiliation(s)
- Akie Hamamoto
- Graduate School of Integrated Arts and Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuki Kobayashi
- Graduate School of Integrated Arts and Sciences, Hiroshima University, Hiroshima, Japan
| | - Yumiko Saito
- Graduate School of Integrated Arts and Sciences, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
36
|
Haemmerle CAS, Campos AMP, Bittencourt JC. Melanin-concentrating hormone inputs to the nucleus accumbens originate from distinct hypothalamic sources and are apposed to GABAergic and cholinergic cells in the Long-Evans rat brain. Neuroscience 2015; 289:392-405. [PMID: 25613687 DOI: 10.1016/j.neuroscience.2015.01.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/06/2015] [Accepted: 01/09/2015] [Indexed: 01/12/2023]
Abstract
Melanin-concentrating hormone [MCH] is a neuropeptide that modulates several behaviors, such as feeding and reward. Because the hedonic and rewarding features of a food also influence feeding behavior, the nucleus accumbens [Acb] has been highlighted as a key area integrating these roles. Functional data confirm that MCH acts on a subdivision of the Acb; however, considering the importance of finding anatomical and neurochemical data that correlate the previously demonstrated function of MCH, we delineated this investigation based on the following points: (1) Is there a pattern of innervation by MCH fibers regarding the subregions within the Acb? (2) Specifically, which hypothalamic nuclei synthesize MCH and innervate the Acb? (3) Finally, what are the neurochemical identities of the accumbal neurons innervated by MCH inputs? We examined the MCH immunoreactivity [MCH-ir] in the Acb in rat brains using the peroxidase technique. Additionally, after injecting retrograde neuronal tracer [Fluoro-Gold® - FG®] into subdivisions of the Acb [shell or core], we mapped single- or double-labeled cells. Moreover, using a double immunoperoxidase protocol, we investigated the MCH-ir fibers for gamma-aminobutyric acid [GABA]-ir and choline acetyltransferase [ChAT]-ir cells in the shell subdivision of the Acb [AcbSh]. We found that the MCH-ir fibers preferentially innervate the medial AcbSh, particularly the septal pole. This innervation originated from the incerto-hypothalamic area [IHy], internuclear area, lateral hypothalamic area, perifornical area, periventricular nucleus and posterior hypothalamus. Moreover, the IHy has the highest relationship between double/single retrogradely labeled cells [n=5.33±0.66/16±0.93, i.e. 33.33%] in the whole hypothalamus. Furthermore, our data suggest that MCH-ir fibers are in apposition to GABAergic and cholinergic cells in the AcbSh. Therefore, we provide anatomical support to the ongoing functional studies investigating the relation among the hypothalamus, MCH transmission into the Acb and the involvement of known neuronal phenotypes within the AcbSh.
Collapse
Affiliation(s)
- C A S Haemmerle
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, 05508-000 São Paulo, Brazil.
| | - A M P Campos
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, 05508-000 São Paulo, Brazil.
| | - J C Bittencourt
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, 05508-000 São Paulo, Brazil; Center for Neuroscience and Behavior, Institute of Psychology, University of São Paulo, 05508-000 São Paulo, Brazil.
| |
Collapse
|
37
|
Konadhode RR, Pelluru D, Shiromani PJ. Neurons containing orexin or melanin concentrating hormone reciprocally regulate wake and sleep. Front Syst Neurosci 2015; 8:244. [PMID: 25620917 PMCID: PMC4287014 DOI: 10.3389/fnsys.2014.00244] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 12/12/2014] [Indexed: 01/22/2023] Open
Abstract
Neurons containing orexin (hypocretin), or melanin concentrating hormone (MCH) are intermingled with each other in the perifornical and lateral hypothalamus. Each is a separate and distinct neuronal population, but they project to similar target areas in the brain. Orexin has been implicated in regulating arousal since loss of orexin neurons is associated with the sleep disorder narcolepsy. Microinjections of orexin into the brain or optogenetic stimulation of orexin neurons increase waking. Orexin neurons are active in waking and quiescent in sleep, which is consistent with their role in promoting waking. On the other hand, the MCH neurons are quiet in waking but active in sleep, suggesting that they could initiate sleep. Recently, for the first time the MCH neurons were stimulated optogenetically and it increased sleep. Indeed, optogenetic activation of MCH neurons induced sleep in both mice and rats at a circadian time when they should be awake, indicating the powerful effect that MCH neurons have in suppressing the wake-promoting effect of not only orexin but also of all of the other arousal neurotransmitters. Gamma-Aminobutyric acid (GABA) is coexpressed with MCH in the MCH neurons, although MCH is also inhibitory. The inhibitory tone of the MCH neurons is opposite to the excitatory tone of the orexin neurons. We hypothesize that strength in activity of each determines wake vs. sleep.
Collapse
Affiliation(s)
- Roda Rani Konadhode
- Departments of Psychiatry and Behavioral Sciences, Medical University of South Carolina Charleston, SC, USA
| | - Dheeraj Pelluru
- Departments of Psychiatry and Behavioral Sciences, Medical University of South Carolina Charleston, SC, USA
| | - Priyattam J Shiromani
- Departments of Psychiatry and Behavioral Sciences, Medical University of South Carolina Charleston, SC, USA ; Ralph H. Johnson VA Medical Center, Medical University of South Carolina Charleston, SC, USA
| |
Collapse
|
38
|
Monti JM, Lagos P, Jantos H, Torterolo P. Increased REM sleep after intra-locus coeruleus nucleus microinjection of melanin-concentrating hormone (MCH) in the rat. Prog Neuropsychopharmacol Biol Psychiatry 2015; 56:185-8. [PMID: 25257545 DOI: 10.1016/j.pnpbp.2014.09.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 08/30/2014] [Accepted: 09/15/2014] [Indexed: 12/01/2022]
Abstract
A study was carried out on the effects of unilateral microinjection of melanin-concentrating hormone (MCH) into the right locus coeruleus (LC) on the sleep-wake cycle in rats prepared for chronic sleep recordings. MCH 200 ng significantly augmented rapid-eye-movement sleep (REMS) time during the first, second and third 2-h of recording. Furthermore, MCH 100 ng induced a significant increase of REMS during the first 2-h period after treatment. The increment of the behavioral state was related to a greater number of REMS episodes. It is suggested that MCH deactivation of noradrenergic neurons located in the LC facilitates the occurrence of REMS.
Collapse
Affiliation(s)
- Jaime M Monti
- Department of Pharmacology and Therapeutics, School of Medicine Clinics Hospital, Montevideo 11600, Uruguay.
| | - Patricia Lagos
- Department of Physiology, School of Medicine, Montevideo 11300, Uruguay
| | - Héctor Jantos
- Department of Pharmacology and Therapeutics, School of Medicine Clinics Hospital, Montevideo 11600, Uruguay
| | - Pablo Torterolo
- Department of Physiology, School of Medicine, Montevideo 11300, Uruguay
| |
Collapse
|
39
|
Parks GS, Wang L, Wang Z, Civelli O. Identification of neuropeptide receptors expressed by melanin-concentrating hormone neurons. J Comp Neurol 2014; 522:3817-33. [PMID: 24978951 PMCID: PMC4167928 DOI: 10.1002/cne.23642] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 06/17/2014] [Accepted: 06/24/2014] [Indexed: 01/13/2023]
Abstract
Melanin-concentrating hormone (MCH) is a 19-amino-acid cyclic neuropeptide that acts in rodents via the MCH receptor 1 (MCHR1) to regulate a wide variety of physiological functions. MCH is produced by a distinct population of neurons located in the lateral hypothalamus (LH) and zona incerta (ZI), but MCHR1 mRNA is widely expressed throughout the brain. The physiological responses and behaviors regulated by the MCH system have been investigated, but less is known about how MCH neurons are regulated. The effects of most classical neurotransmitters on MCH neurons have been studied, but those of most neuropeptides are poorly understood. To gain insight into how neuropeptides regulate the MCH system, we investigated which neuropeptide receptors are expressed by MCH neurons by using double in situ hybridization. In all, 20 receptors, selected based on either a suspected interaction with the MCH system or demonstrated high expression levels in the LH and ZI, were tested to determine whether they are expressed by MCH neurons. Overall, 11 neuropeptide receptors were found to exhibit significant colocalization with MCH neurons: nociceptin/orphanin FQ opioid receptor (NOP), MCHR1, both orexin receptors (ORX), somatostatin receptors 1 and 2 (SSTR1, SSTR2), kisspeptin recepotor (KissR1), neurotensin receptor 1 (NTSR1), neuropeptide S receptor (NPSR), cholecystokinin receptor A (CCKAR), and the κ-opioid receptor (KOR). Among these receptors, six have never before been linked to the MCH system. Surprisingly, several receptors thought to regulate MCH neurons displayed minimal colocalization with MCH, suggesting that they may not directly regulate the MCH system.
Collapse
Affiliation(s)
- Gregory S. Parks
- Department of Pharmacology, University of California Irvine, Irvine, California 92697
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California, 92697
| | - Lien Wang
- Department of Pharmacology, University of California Irvine, Irvine, California 92697
| | - Zhiwei Wang
- Department of Pharmacology, University of California Irvine, Irvine, California 92697
| | - Olivier Civelli
- Department of Pharmacology, University of California Irvine, Irvine, California 92697
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California, 92697
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, California, 92697
| |
Collapse
|
40
|
Wollmann G, Paglino JC, Maloney PR, Ahmadi SA, van den Pol AN. Attenuation of vesicular stomatitis virus infection of brain using antiviral drugs and an adeno-associated virus-interferon vector. Virology 2014; 475:1-14. [PMID: 25462341 DOI: 10.1016/j.virol.2014.10.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 10/04/2014] [Accepted: 10/20/2014] [Indexed: 12/21/2022]
Abstract
Vesicular stomatitis virus (VSV) shows promise as a vaccine-vector and oncolytic virus. However, reports of neurotoxicity of VSV remain a concern. We compared 12 antiviral compounds to control infection of VSV-CT9-M51 and VSV-rp30 using murine and human brain cultures, and in vivo mouse models. Inhibition of replication, cytotoxicity and infectivity was strongest with ribavirin and IFN-α and to some extent with mycophenolic acid, chloroquine, and adenine 9-β-d-arabinofuranoside. To generate continuous IFN exposure, we made an adeno-associated virus vector expressing murine IFN; AAV-mIFN-β protected mouse brain cells from VSV, as did a combination of IFN, ribavirin and chloroquine. Intracranial AAV-mIFN-β protected the brain against VSV-CT9-M51. In SCID mice bearing human glioblastoma, AAV-mIFN-β moderately enhanced survival. VSV-CT9-M51 doubled median survival when administered after AAV-mIFN-β; some surviving mice showed complete tumor destruction. Together, these data suggest that AAV-IFN or IFN with ribavirin and chloroquine provide an optimal anti-virus combination against VSV in the brain.
Collapse
Affiliation(s)
- Guido Wollmann
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Justin C Paglino
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Patrick R Maloney
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Sebastian A Ahmadi
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States
| | - Anthony N van den Pol
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520, United States.
| |
Collapse
|
41
|
Benedetto L, Pereira M, Ferreira A, Torterolo P. Melanin-concentrating hormone in the medial preoptic area reduces active components of maternal behavior in rats. Peptides 2014; 58:20-5. [PMID: 24893251 DOI: 10.1016/j.peptides.2014.05.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 01/16/2023]
Abstract
Melanin-concentrating hormone (MCH) is an inhibitory neuropeptide mainly synthesized in neurons of the lateral hypothalamus and incerto-hypothalamic area of mammals that has been implicated in behavioral functions related to motivation. During lactation, this neuropeptide is also expressed in the medial preoptic area (mPOA), a key region of the maternal behavior circuitry. Notably, whereas MCH expression in the mPOA progressively increases during lactation, maternal behavior naturally declines, suggesting that elevated MCHergic activity in the mPOA inhibit maternal behavior in the late postpartum period. To explore this idea, we assessed the maternal behavior of early postpartum females following bilateral microinfusions of either MCH (50 and 100 ng/0.2 μl/side) or the same volume of vehicle into the mPOA. As expected, females receiving 100 ng MCH into the mPOA exhibited significant deficits in the active components of maternal behavior, including retrieving and nest building. In contrast, nursing, as well as other behaviors, including locomotor activity, exploration, and anxiety-like behavior, were not affected by intra-mPOA MCH infusion. The present results, together with previous findings showing elevated expression of this neuropeptide toward the end of the postpartum period, suggest that modulation of mPOA function by MCH may contribute to the weaning of maternal responsiveness characteristic of the late postpartum period.
Collapse
Affiliation(s)
- Luciana Benedetto
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Mariana Pereira
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ, USA
| | - Annabel Ferreira
- Sección de Fisiología y Nutrición, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay.
| | - Pablo Torterolo
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
42
|
Optogenetic manipulation of activity and temporally controlled cell-specific ablation reveal a role for MCH neurons in sleep/wake regulation. J Neurosci 2014; 34:6896-909. [PMID: 24828644 DOI: 10.1523/jneurosci.5344-13.2014] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Melanin-concentrating hormone (MCH) is a neuropeptide produced in neurons sparsely distributed in the lateral hypothalamic area. Recent studies have reported that MCH neurons are active during rapid eye movement (REM) sleep, but their physiological role in the regulation of sleep/wakefulness is not fully understood. To determine the physiological role of MCH neurons, newly developed transgenic mouse strains that enable manipulation of the activity and fate of MCH neurons in vivo were generated using the recently developed knockin-mediated enhanced gene expression by improved tetracycline-controlled gene induction system. The activity of these cells was controlled by optogenetics by expressing channelrhodopsin2 (E123T/T159C) or archaerhodopsin-T in MCH neurons. Acute optogenetic activation of MCH neurons at 10 Hz induced transitions from non-REM (NREM) to REM sleep and increased REM sleep time in conjunction with decreased NREM sleep. Activation of MCH neurons while mice were in NREM sleep induced REM sleep, but activation during wakefulness was ineffective. Acute optogenetic silencing of MCH neurons using archaerhodopsin-T had no effect on any vigilance states. Temporally controlled ablation of MCH neurons by cell-specific expression of diphtheria toxin A increased wakefulness and decreased NREM sleep duration without affecting REM sleep. Together, these results indicate that acute activation of MCH neurons is sufficient, but not necessary, to trigger the transition from NREM to REM sleep and that MCH neurons also play a role in the initiation and maintenance of NREM sleep.
Collapse
|
43
|
Luppi PH, Peyron C, Fort P. Role of MCH neurons in paradoxical (REM) sleep control. Sleep 2013; 36:1775-6. [PMID: 24293748 DOI: 10.5665/sleep.3192] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Pierre-Hervé Luppi
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veillesommeil," Lyon, France, University Lyon 1, Lyon, France
| | | | | |
Collapse
|
44
|
Jones BE, Hassani OK. The role of Hcrt/Orx and MCH neurons in sleep-wake state regulation. Sleep 2013; 36:1769-72. [PMID: 24293746 DOI: 10.5665/sleep.3188] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Barbara E Jones
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | | |
Collapse
|
45
|
Chee MJS, Pissios P, Maratos-Flier E. Neurochemical characterization of neurons expressing melanin-concentrating hormone receptor 1 in the mouse hypothalamus. J Comp Neurol 2013; 521:2208-34. [PMID: 23605441 DOI: 10.1002/cne.23273] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 11/09/2012] [Accepted: 11/20/2012] [Indexed: 01/16/2023]
Abstract
Melanin-concentrating hormone (MCH) is a hypothalamic neuropeptide that acts via MCH receptor 1 (MCHR1) in the mouse. It promotes positive energy balance; thus, mice lacking MCH or MCHR1 are lean, hyperactive, and resistant to diet-induced obesity. Identifying the cellular targets of MCH is an important step to understanding the mechanisms underlying MCH actions. We generated the Mchr1-cre mouse that expresses cre recombinase driven by the MCHR1 promoter and crossed it with a tdTomato reporter mouse. The resulting Mchr1-cre/tdTomato progeny expressed easily detectable tdTomato fluorescence in MCHR1 neurons, which were found throughout the olfactory system, striatum, and hypothalamus. To chemically identify MCH-targeted cell populations that play a role in energy balance, MCHR1 hypothalamic neurons were characterized by colabeling select hypothalamic neuropeptides with tdTomato fluorescence. TdTomato fluorescence colocalized with dynorphin, oxytocin, vasopressin, enkephalin, thyrothropin-releasing hormone, and corticotropin-releasing factor immunoreactive cells in the paraventricular nucleus. In the lateral hypothalamus, neurotensin, but neither orexin nor MCH neurons, expressed tdTomato. In the arcuate nucleus, both Neuropeptide Y and proopiomelanocortin cells expressed tdTomato. We further demonstrated that some of these arcuate neurons were also targets of leptin action. Interestingly, MCHR1 was expressed in the vast majority of leptin-sensitive proopiomelanocortin neurons, highlighting their importance for the orexigenic actions of MCH. Taken together, this study supports the use of the Mchr1-cre mouse for outlining the neuroanatomical distribution and neurochemical phenotype of MCHR1 neurons.
Collapse
Affiliation(s)
- Melissa J S Chee
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
46
|
Role of melanin-concentrating hormone in the nucleus accumbens shell in rats behaviourally sensitized to methamphetamine. Int J Neuropsychopharmacol 2013; 16:1767-80. [PMID: 23449013 DOI: 10.1017/s1461145713000072] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Melanin-concentrating hormone (MCH) is a neuropeptide and its receptor is extensively expressed throughout the brain. MCH has been suggested to regulate the rewarding and reinforcing effects of psychostimulants by potentiating the dopaminergic system within the midbrain. Moreover, MCH and its receptor can regulate ERK activity. The present study investigated the role of MCH in the nucleus accumbens (NAc) in rats behaviourally sensitized to methamphetamine (Meth). We found that the development of Meth-induced locomotor sensitization was attenuated by MCH infused into the NAc shell but not core. Moreover, the elevation of ERK phosphorylation in the NAc shell induced by Meth was inhibited by locally infused MCH. Infusion of the MCH receptor 1 (MCHR1) antagonist SNAP 94847 into the NAc shell but not core augmented the initiation of locomotor sensitization and amplitude of elevated phosphorylated ERK levels induced by Meth. The expression of Meth-induced locomotor sensitization and ERK alterations after 1 wk withdrawal were not affected by either MCH or SNAP 94847 infused into the NAc shell or core. These results indicate that MCH in the NAc shell plays a critical role in the development but not expression of Meth-induced locomotor sensitization in rats, which might be mediated by the ERK signalling pathway. Our study suggests that MCH might be a potential target for the treatment of Meth addiction.
Collapse
|
47
|
Abstract
Melanin concentrating hormone (MCH) is a cyclic neuropeptide present in the hypothalamus of all vertebrates. MCH is implicated in a number of behaviors but direct evidence is lacking. To selectively stimulate the MCH neurons the gene for the light-sensitive cation channel, channelrhodopsin-2, was inserted into the MCH neurons of wild-type mice. Three weeks later MCH neurons were stimulated for 1 min every 5 min for 24 h. A 10 Hz stimulation at the start of the night hastened sleep onset, reduced length of wake bouts by 50%, increased total time in non-REM and REM sleep at night, and increased sleep intensity during the day cycle. Sleep induction at a circadian time when all of the arousal neurons are active indicates that MCH stimulation can powerfully counteract the combined wake-promoting signal of the arousal neurons. This could be potentially useful in treatment of insomnia.
Collapse
|
48
|
Monti JM, Torterolo P, Lagos P. Melanin-concentrating hormone control of sleep–wake behavior. Sleep Med Rev 2013; 17:293-8. [DOI: 10.1016/j.smrv.2012.10.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2012] [Revised: 10/06/2012] [Accepted: 10/08/2012] [Indexed: 01/16/2023]
|
49
|
Lima FF, Sita LV, Oliveira AR, Costa HC, da Silva JM, Mortara RA, Haemmerle CA, Xavier GF, Canteras NS, Bittencourt JC. Hypothalamic melanin-concentrating hormone projections to the septo-hippocampal complex in the rat. J Chem Neuroanat 2013; 47:1-14. [DOI: 10.1016/j.jchemneu.2012.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 10/08/2012] [Accepted: 10/10/2012] [Indexed: 11/29/2022]
|
50
|
Abstract
Neuropeptides are found in many mammalian CNS neurons where they play key roles in modulating neuronal activity. In contrast to amino acid transmitter release at the synapse, neuropeptide release is not restricted to the synaptic specialization, and after release, a neuropeptide may diffuse some distance to exert its action through a G protein-coupled receptor. Some neuropeptides such as hypocretin/orexin are synthesized only in single regions of the brain, and the neurons releasing these peptides probably have similar functional roles. Other peptides such as neuropeptide Y (NPY) are synthesized throughout the brain, and neurons that synthesize the peptide in one region have no anatomical or functional connection with NPY neurons in other brain regions. Here, I review converging data revealing a complex interaction between slow-acting neuromodulator peptides and fast-acting amino acid transmitters in the control of energy homeostasis, drug addiction, mood and motivation, sleep-wake states, and neuroendocrine regulation.
Collapse
|