1
|
Luppi PH, Malcey J, Chancel A, Duval B, Cabrera S, Fort P. Neuronal network controlling REM sleep. J Sleep Res 2025; 34:e14266. [PMID: 38972672 DOI: 10.1111/jsr.14266] [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: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 07/09/2024]
Abstract
Rapid eye movement sleep is a state characterized by concomitant occurrence of rapid eye movements, electroencephalographic activation and muscle atonia. In this review, we provide up to date knowledge on the neuronal network controlling its onset and maintenance. It is now accepted that muscle atonia during rapid eye movement sleep is due to activation of glutamatergic neurons localized in the pontine sublaterodorsal tegmental nucleus. These neurons directly project and excite glycinergic/γ-aminobutyric acid-ergic pre-motoneurons localized in the ventromedial medulla. The sublaterodorsal tegmental nucleus rapid eye movement-on neurons are inactivated during wakefulness and non-rapid eye movement by rapid eye movement-off γ-aminobutyric acid-ergic neurons localized in the ventrolateral periaqueductal grey and the adjacent dorsal deep mesencephalic reticular nucleus. Melanin-concentrating hormone and γ-aminobutyric acid-ergic rapid eye movement sleep-on neurons localized in the lateral hypothalamus would inhibit these rapid eye movement sleep-off neurons initiating the state. Finally, the activation of a few limbic cortical structures during rapid eye movement sleep by the claustrum and the supramammillary nucleus as well as that of the basolateral amygdala would be involved in the function(s) of rapid eye movement sleep. In summary, rapid eye movement sleep is generated by a brainstem generator controlled by forebrain structures involved in autonomic control.
Collapse
Affiliation(s)
- Pierre-Hervé Luppi
- INSERM, U1028; CNRS, UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Lyon 1, Lyon, France
| | - Justin Malcey
- INSERM, U1028; CNRS, UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Lyon 1, Lyon, France
| | - Amarine Chancel
- INSERM, U1028; CNRS, UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Lyon 1, Lyon, France
| | - Blandine Duval
- INSERM, U1028; CNRS, UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
| | - Sébastien Cabrera
- INSERM, U1028; CNRS, UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Lyon 1, Lyon, France
| | - Patrice Fort
- INSERM, U1028; CNRS, UMR5292, Lyon Neuroscience Research Center, Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France
- University Lyon 1, Lyon, France
| |
Collapse
|
2
|
Naganuma F, Khanday M, Bandaru SS, Hasan W, Hirano K, Yoshikawa T, Vetrivelan R. Regulation of wakefulness by neurotensin neurons in the lateral hypothalamus. Exp Neurol 2024; 383:115035. [PMID: 39481513 DOI: 10.1016/j.expneurol.2024.115035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/04/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
The lateral hypothalamic region (LH) has been identified as a key region for arousal regulation, yet the specific cell types and underlying mechanisms are not fully understood. While neurons expressing orexins (OX) are considered the primary wake-promoting population in the LH, their loss does not reduce daily wake levels, suggesting the presence of additional wake-promoting populations. In this regard, we recently discovered that a non-OX cell group in the LH, marked by the expression of neurotensin (Nts), could powerfully drive wakefulness. Activation of these NtsLH neurons elicits rapid arousal from non-rapid eye movement (NREM) sleep and produces uninterrupted wakefulness for several hours in mice. However, it remains unknown if these neurons are necessary for spontaneous wakefulness and what their precise role is in the initiation and maintenance of this state. To address these questions, we first examined the activity dynamics of the NtsLH population across sleep-wake behavior using fiber photometry. We find that NtsLH neurons are more active during wakefulness, and their activity increases concurrently with, but does not precede, wake-onset. We then selectively destroyed the NtsLH neurons using a diphtheria-toxin-based conditional ablation method, which significantly reduced wake amounts and mean duration of wake bouts and increased the EEG delta power during wakefulness. These findings demonstrate a crucial role for NtsLH neurons in maintaining normal arousal levels, and their loss may be associated with chronic sleepiness in mice.
Collapse
Affiliation(s)
- Fumito Naganuma
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Mudasir Khanday
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America
| | - Sathyajit Sai Bandaru
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Whidul Hasan
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America
| | - Kyosuke Hirano
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Takeo Yoshikawa
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Ramalingam Vetrivelan
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
3
|
Luppi PH, Chancel A, Malcey J, Cabrera S, Fort P, Maciel RM. Which structure generates paradoxical (REM) sleep: The brainstem, the hypothalamus, the amygdala or the cortex? Sleep Med Rev 2024; 74:101907. [PMID: 38422648 DOI: 10.1016/j.smrv.2024.101907] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/31/2023] [Accepted: 01/19/2024] [Indexed: 03/02/2024]
Abstract
Paradoxical or Rapid eye movement (REM) sleep (PS) is a state characterized by REMs, EEG activation and muscle atonia. In this review, we discuss the contribution of brainstem, hypothalamic, amygdalar and cortical structures in PS genesis. We propose that muscle atonia during PS is due to activation of glutamatergic neurons localized in the pontine sublaterodorsal tegmental nucleus (SLD) projecting to glycinergic/GABAergic pre-motoneurons localized in the ventro-medial medulla (vmM). The SLD PS-on neurons are inactivated during wakefulness and slow-wave sleep by PS-off GABAergic neurons localized in the ventrolateral periaqueductal gray (vPAG) and the adjacent deep mesencephalic reticular nucleus. Melanin concentrating hormone (MCH) and GABAergic PS-on neurons localized in the posterior hypothalamus would inhibit these PS-off neurons to initiate the state. Finally, the activation of a few limbic cortical structures during PS by the claustrum and the supramammillary nucleus as well as that of the basolateral amygdala would also contribute to PS expression. Accumulating evidence indicates that the activation of these limbic structures plays a role in memory consolidation and would communicate to the PS-generating structures the need for PS to process memory. In summary, PS generation is controlled by structures distributed from the cortex to the medullary level of the brain.
Collapse
Affiliation(s)
- Pierre-Hervé Luppi
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, SLEEP Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France; University Claude Bernard, Lyon 1, Lyon, France.
| | - Amarine Chancel
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, SLEEP Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France; University Claude Bernard, Lyon 1, Lyon, France
| | - Justin Malcey
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, SLEEP Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France; University Claude Bernard, Lyon 1, Lyon, France
| | - Sébastien Cabrera
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, SLEEP Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France; University Claude Bernard, Lyon 1, Lyon, France
| | - Patrice Fort
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, SLEEP Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France; University Claude Bernard, Lyon 1, Lyon, France
| | - Renato M Maciel
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, SLEEP Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", Lyon, France; University Claude Bernard, Lyon 1, Lyon, France
| |
Collapse
|
4
|
Chancel A, Fort P, Luppi PH. The role of the hypothalamic Lhx6 GABAergic neurons in REM sleep control. Sleep 2024; 47:zsad331. [PMID: 38159085 PMCID: PMC10925945 DOI: 10.1093/sleep/zsad331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Indexed: 01/03/2024] Open
Affiliation(s)
- Amarine Chancel
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, SLEEP Team, Bron, France
| | - Patrice Fort
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, SLEEP Team, Bron, France
| | - Pierre-Hervé Luppi
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, SLEEP Team, Bron, France
| |
Collapse
|
5
|
Koyama Y. The role of orexinergic system in the regulation of cataplexy. Peptides 2023; 169:171080. [PMID: 37598758 DOI: 10.1016/j.peptides.2023.171080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/06/2023] [Accepted: 08/18/2023] [Indexed: 08/22/2023]
Abstract
Loss of orexin/hypocretin causes serious sleep disorder; narcolepsy. Cataplexy is the most striking symptom of narcolepsy, characterized by abrupt muscle paralysis induced by emotional stimuli, and has been considered pathological activation of REM sleep atonia system. Clinical treatments for cataplexy/narcolepsy and early pharmacological studies in narcoleptic dogs tell us about the involvement of monoaminergic and cholinergic systems in the control of cataplexy/narcolepsy. Muscle atonia may be induced by activation of REM sleep-atonia generating system in the brainstem. Emotional stimuli may be processed in the limbic systems including the amygdala, nucleus accumbens, and medial prefrontal cortex. It is now considered that orexin/hypocretin prevents cataplexy by modulating the activity of different points of cataplexy-inducing circuit, including monoaminergic/cholinergic systems, muscle atonia-generating systems, and emotion-related systems. This review will describe the recent advances in understanding the neural mechanisms controlling cataplexy, with a focus on the involvement of orexin/hypocretin system, and will discuss future experimental strategies that will lead to further understanding and treatment of this disease.
Collapse
Affiliation(s)
- Yoshimasa Koyama
- Faculty of Symbiotic Systems Science, Fukushima University, 1 Kanaya-gawa, Fukushima 960-1296, Japan..
| |
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
|
Adamantidis AR, de Lecea L. Sleep and the hypothalamus. Science 2023; 382:405-412. [PMID: 37883555 DOI: 10.1126/science.adh8285] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/08/2023] [Indexed: 10/28/2023]
Abstract
Neural substrates of wakefulness, rapid eye movement sleep (REMS), and non-REMS (NREMS) in the mammalian hypothalamus overlap both anatomically and functionally with cellular networks that support physiological and behavioral homeostasis. Here, we review the roles of sleep neurons of the hypothalamus in the homeostatic control of thermoregulation or goal-oriented behaviors during wakefulness. We address how hypothalamic circuits involved in opposing behaviors such as core body temperature and sleep compute conflicting information and provide a coherent vigilance state. Finally, we highlight some of the key unresolved questions and challenges, and the promise of a more granular view of the cellular and molecular diversity underlying the integrative role of the hypothalamus in physiological and behavioral homeostasis.
Collapse
Affiliation(s)
- Antoine R Adamantidis
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Luis de Lecea
- Department of Psychiatry and Behavioural Sciences, Stanford, CA, USA
- Wu Tsai Neurosciences Institute Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
8
|
Kalinowski D, Bogus-Nowakowska K, Kozłowska A, Równiak M. The Co-Expression Pattern of Calcium-Binding Proteins with γ-Aminobutyric Acid and Glutamate Transporters in the Amygdala of the Guinea Pig: Evidence for Glutamatergic Subpopulations. Int J Mol Sci 2023; 24:15025. [PMID: 37834473 PMCID: PMC10573686 DOI: 10.3390/ijms241915025] [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: 08/04/2023] [Revised: 09/27/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
The amygdala has large populations of neurons utilizing specific calcium-binding proteins such as parvalbumin (PV), calbindin (CB), or calretinin (CR). They are considered specialized subsets of γ-aminobutyric acid (GABA) interneurons; however, many of these cells are devoid of GABA or glutamate decarboxylase. The neurotransmitters used by GABA-immunonegative cells are still unknown, but it is suggested that a part may use glutamate. Thus, this study investigates in the amygdala of the guinea pig relationships between PV, CB, or CR-containing cells and GABA transporter (VGAT) or glutamate transporter type 2 (VGLUT2), markers of GABAergic and glutamatergic neurons, respectively. The results show that although most neurons using PV, CB, and CR co-expressed VGAT, each of these populations also had a fraction of VGLUT2 co-expressing cells. For almost all neurons using PV (~90%) co-expressed VGAT, while ~1.5% of them had VGLUT2. The proportion of neurons using CB and VGAT was smaller than that for PV (~80%), while the percentage of cells with VGLUT2 was larger (~4.5%). Finally, only half of the neurons using CR (~53%) co-expressed VGAT, while ~3.5% of them had VGLUT2. In conclusion, the populations of neurons co-expressing PV, CB, and CR are in the amygdala, primarily GABAergic. However, at least a fraction of neurons in each of them co-express VGLUT2, suggesting that these cells may use glutamate. Moreover, the number of PV-, CB-, and CR-containing neurons that may use glutamate is probably larger as they can utilize VGLUT1 or VGLUT3, which are also present in the amygdala.
Collapse
Affiliation(s)
- Daniel Kalinowski
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, pl. Łódzki 3, 10-727 Olsztyn, Poland; (K.B.-N.); (M.R.)
| | - Krystyna Bogus-Nowakowska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, pl. Łódzki 3, 10-727 Olsztyn, Poland; (K.B.-N.); (M.R.)
| | - Anna Kozłowska
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082 Olsztyn, Poland;
| | - Maciej Równiak
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, pl. Łódzki 3, 10-727 Olsztyn, Poland; (K.B.-N.); (M.R.)
| |
Collapse
|
9
|
Miao HT, Song RX, Xin Y, Wang LY, Lv JM, Liu NN, Wu ZY, Zhang W, Li Y, Zhang DX, Zhang LM. Spautin-1 Protects Against Mild TBI-Induced Anxiety-Like Behavior in Mice via Immunologically Silent Apoptosis. Neuromolecular Med 2023; 25:336-349. [PMID: 36745326 DOI: 10.1007/s12017-023-08737-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023]
Abstract
Anxiety is reportedly one of the most common mental changes after traumatic brain injury (TBI). Perineuronal nets (PNNs) produced by astrocytes in the lateral hypothalamus (LHA) that surround gamma-aminobutyric acid-ergic (GABAergic) neurons have been associated with anxiety. The potent anti-tumor effects of Spautin-1, a novel autophagy inhibitor, have been documented in malignant melanoma; moreover, the inhibition of autophagy is reported to mitigate anxiety disorders. However, little is known about the ability of spautin-1 to alleviate anxiety. In this study, we sought to investigate whether spautin-1 could alleviate anxiety-like behaviors post-TBI by reducing the loss of PNNs in the LHA. A mild TBI was established in mice through Feeney's weight-drop model. Then, Spautin-1 (20 mmol/2 μl) was immediately administered into the left lateral ventricle. Behavioral and pathological changes were assessed at 24 h, 7 days, 30 days, 31 days and 32 days after TBI by the neurological severity scores (NSS), open field test (OFT), elevated plus-maze (EPM) test, western blot, immunofluorescence assays and electron microscopy. Spautin-1 significantly reversed TBI-induced decreased time in the central zone during OFT and in the open-arm during the EPM test. Spautin-1 also increased PNNs around GABAergic neurons indicated by WFA- plus GAD2- positive A2-type astrocytes and attenuated M1-type microglia in the LHA 32 days after TBI compared to TBI alone. Moreover, compared to mice that only underwent TBI, spautin-1 downregulated autophagic vacuoles, abnormal organelles, the expression of Beclin 1, USP13, phospho-TBK1, and phospho-IRF3 and upregulated the levels of cleaved caspase-3, -7 and -9, but failed to increase TUNEL-positive cells in the LHA at 24 h. Spautin-1 alleviated anxiety-like behavior in mice exposed to mild TBI; this protective mechanism may be associated with decreased PNNs loss around GABAergic neurons via immunologically silent apoptosis induced by the caspase cascade.
Collapse
Affiliation(s)
- Hui-Tao Miao
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Rong-Xin Song
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Yue Xin
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Lu-Ying Wang
- Department of Anesthesia and Trauma Research, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Jin-Meng Lv
- Department of Anesthesia and Trauma Research, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Na-Na Liu
- Department of Pediatric, Cangzhou Central Hospital, Cangzhou, China
| | - Zhi-You Wu
- Department of Neurosurgery, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Wei Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Li
- Department of Anesthesiology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Li-Min Zhang
- Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China.
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China.
| |
Collapse
|
10
|
Sa M, Yoo ES, Koh W, Park MG, Jang HJ, Yang YR, Bhalla M, Lee JH, Lim J, Won W, Kwon J, Kwon JH, Seong Y, Kim B, An H, Lee SE, Park KD, Suh PG, Sohn JW, Lee CJ. Hypothalamic GABRA5-positive neurons control obesity via astrocytic GABA. Nat Metab 2023; 5:1506-1525. [PMID: 37653043 DOI: 10.1038/s42255-023-00877-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/25/2023] [Indexed: 09/02/2023]
Abstract
The lateral hypothalamic area (LHA) regulates food intake and energy balance. Although LHA neurons innervate adipose tissues, the identity of neurons that regulate fat is undefined. Here we show that GABRA5-positive neurons in LHA (GABRA5LHA) polysynaptically project to brown and white adipose tissues in the periphery. GABRA5LHA are a distinct subpopulation of GABAergic neurons and show decreased pacemaker firing in diet-induced obesity mouse models in males. Chemogenetic inhibition of GABRA5LHA suppresses fat thermogenesis and increases weight gain, whereas gene silencing of GABRA5 in LHA decreases weight gain. In the diet-induced obesity mouse model, GABRA5LHA are tonically inhibited by nearby reactive astrocytes releasing GABA, which is synthesized by monoamine oxidase B (Maob). Gene silencing of astrocytic Maob in LHA facilitates fat thermogenesis and reduces weight gain significantly without affecting food intake, which is recapitulated by administration of a Maob inhibitor, KDS2010. We propose that firing of GABRA5LHA suppresses fat accumulation and selective inhibition of astrocytic GABA is a molecular target for treating obesity.
Collapse
Affiliation(s)
- Moonsun Sa
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
| | - Eun-Seon Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Mingu Gordon Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Hyun-Jun Jang
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Yong Ryoul Yang
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Mridula Bhalla
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Jae-Hun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Woojin Won
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jea Kwon
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Joon-Ho Kwon
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Yejin Seong
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Byungeun Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Heeyoung An
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ki Duk Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Pann-Ghill Suh
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul, Republic of Korea.
- IBS School, University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
11
|
Blanco-Centurion C, Vidal-Ortiz A, Sato T, Shiromani PJ. Activity of GABA neurons in the zona incerta and ventral lateral periaqueductal grey is biased towards sleep. Sleep 2023; 46:6902001. [PMID: 36516419 DOI: 10.1093/sleep/zsac306] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/31/2022] [Indexed: 12/15/2022] Open
Abstract
STUDY OBJECTIVES As in various brain regions the activity of gamma-aminobutyric acid (GABA) neurons is largely unknown, we measured in vivo changes in calcium fluorescence in GABA neurons in the zona incerta (ZI) and the ventral lateral periaqueductal grey (vlPAG), two areas that have been implicated in regulating sleep. METHODS vGAT-Cre mice were implanted with sleep electrodes, microinjected with rAAV-DIO-GCaMP6 into the ZI (n = 6) or vlPAG (n = 5) (isoflurane anesthesia) and a GRIN (Gradient-Index) lens inserted atop the injection site. Twenty-one days later, fluorescence in individual vGAT neurons was recorded over multiple REM cycles. Regions of interest corresponding to individual vGAT somata were automatically extracted with PCA-ICA analysis. RESULTS In the ZI, 372 neurons were identified. Previously, we had recorded the activity of 310 vGAT neurons in the ZI and we combined the published dataset with the new dataset to create a comprehensive dataset of ZI vGAT neurons (total neurons = 682; mice = 11). In the vlPAG, 169 neurons (mice = 5) were identified. In both regions, most neurons were maximally active in REM sleep (R-Max; ZI = 51.0%, vlPAG = 60.9%). The second most abundant group was W-Max (ZI = 23.9%, vlPAG = 25.4%). In the ZI, but not in vlPAG, there were neurons that were NREMS-Max (11.7%). vlPAG had REMS-Off neurons (8.3%). In both areas, there were two minor classes: wake/REMS-Max and state indifferent. In the ZI, the NREMS-Max neurons fluoresced 30 s ahead of sleep onset. CONCLUSIONS These descriptive data show that the activity of GABA neurons is biased in favor of sleep in two brain regions implicated in sleep.
Collapse
Affiliation(s)
| | - Aurelio Vidal-Ortiz
- Laboratory of Sleep Medicine and Chronobiology, Ralph H. Johnson Veterans Healthcare System, Charleston, SC, USA
| | - Takashi Sato
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Priyattam J Shiromani
- Department of Psychiatry and Behavioral Sciences, Charleston, SC, USA
- Laboratory of Sleep Medicine and Chronobiology, Ralph H. Johnson Veterans Healthcare System, Charleston, SC, USA
| |
Collapse
|
12
|
Sulaman BA, Wang S, Tyan J, Eban-Rothschild A. Neuro-orchestration of sleep and wakefulness. Nat Neurosci 2023; 26:196-212. [PMID: 36581730 DOI: 10.1038/s41593-022-01236-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/16/2022] [Indexed: 12/31/2022]
Abstract
Although considered an inactive state for centuries, sleep entails many active processes occurring at the cellular, circuit and organismal levels. Over the last decade, several key technological advances, including calcium imaging and optogenetic and chemogenetic manipulations, have facilitated a detailed understanding of the functions of different neuronal populations and circuits in sleep-wake regulation. Here, we present recent progress and summarize our current understanding of the circuitry underlying the initiation, maintenance and coordination of wakefulness, rapid eye movement sleep (REMS) and non-REMS (NREMS). We propose a de-arousal model for sleep initiation, in which the neuromodulatory milieu necessary for sleep initiation is achieved by engaging in repetitive pre-sleep behaviors that gradually reduce vigilance to the external environment and wake-promoting neuromodulatory tone. We also discuss how brain processes related to thermoregulation, hunger and fear intersect with sleep-wake circuits to control arousal. Lastly, we discuss controversies and lingering questions in the sleep field.
Collapse
Affiliation(s)
- Bibi A Sulaman
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Su Wang
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Jean Tyan
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
13
|
Nollet M, Franks NP, Wisden W. Understanding Sleep Regulation in Normal and Pathological Conditions, and Why It Matters. J Huntingtons Dis 2023; 12:105-119. [PMID: 37302038 PMCID: PMC10473105 DOI: 10.3233/jhd-230564] [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] [Accepted: 05/22/2023] [Indexed: 06/12/2023]
Abstract
Sleep occupies a peculiar place in our lives and in science, being both eminently familiar and profoundly enigmatic. Historically, philosophers, scientists and artists questioned the meaning and purpose of sleep. If Shakespeare's verses from MacBeth depicting "Sleep that soothes away all our worries" and "relieves the weary laborer and heals hurt minds" perfectly epitomize the alleviating benefits of sleep, it is only during the last two decades that the growing understanding of the sophisticated sleep regulatory mechanisms allows us to glimpse putative biological functions of sleep. Sleep control brings into play various brain-wide processes occurring at the molecular, cellular, circuit, and system levels, some of them overlapping with a number of disease-signaling pathways. Pathogenic processes, including mood disorders (e.g., major depression) and neurodegenerative illnesses such Huntington's or Alzheimer's diseases, can therefore affect sleep-modulating networks which disrupt the sleep-wake architecture, whereas sleep disturbances may also trigger various brain disorders. In this review, we describe the mechanisms underlying sleep regulation and the main hypotheses drawn about its functions. Comprehending sleep physiological orchestration and functions could ultimately help deliver better treatments for people living with neurodegenerative diseases.
Collapse
Affiliation(s)
- Mathieu Nollet
- UK Dementia Research Institute and Department of Life Sciences, Imperial College London, London, UK
| | - Nicholas P. Franks
- UK Dementia Research Institute and Department of Life Sciences, Imperial College London, London, UK
| | - William Wisden
- UK Dementia Research Institute and Department of Life Sciences, Imperial College London, London, UK
| |
Collapse
|
14
|
Zhai X, Yuan Y, Xu L, Jun J, Li Y, Yan Y, Zhang L. Cerebrospinal fluid contacting nucleus and its 5-HT: A new insight into the regulation mechanism of general intravenous anesthesia. Brain Res 2022; 1798:148168. [DOI: 10.1016/j.brainres.2022.148168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
|
15
|
GABAergic and Glutamatergic Phenotypes of Neurons Expressing Calcium-Binding Proteins in the Preoptic Area of the Guinea Pig. Int J Mol Sci 2022; 23:ijms23147963. [PMID: 35887305 PMCID: PMC9320123 DOI: 10.3390/ijms23147963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
The mammalian preoptic area (POA) has large populations of calbindin (CB), calretinin (CR) and parvalbumin (PV) neurons, but phenotypes of these cells are unknown. Therefore, the question is whether neurons expressing CB, CR, and/or PV are GABAergic or glutamatergic. Double-immunofluorescence staining followed by epifluorescence and confocal microscopy was used to determine the coexpression patterns of CB, CR and PV expressing neurons with vesicular GABA transporters (VGAT) as specific markers of GABAergic neurons and vesicular glutamate transporters (VGLUT 2) as specific markers of glutamatergic neurons. The guinea pig was adopted as, like humans, it has a reproductive cycle with a true luteal phase and a long gestation period. The results demonstrated that in the guinea pig POA of both sexes, ~80% of CB+ and ~90% of CR+ neurons coexpress VGAT; however, one-fifth of CB+ neurons and one-third of CR+ cells coexpress VGLUT. About two-thirds of PV+ neurons express VGAT, and similar proportion of them coexpress VGLUT. Thus, many CB+, CR+ and PV+ neurons may be exclusively GABAergic (VGAT-expressing cells) or glutamatergic (VGLUT-expressing cells); however, at least a small fraction of CR+ cells and at least one-third of PV+ cells are likely neurons with a dual GABA/glutamate phenotype that may coexpress both transporters.
Collapse
|
16
|
Kostin A, Alam MA, Saevskiy A, McGinty D, Alam MN. Activation of the Ventrolateral Preoptic Neurons Projecting to the Perifornical-Hypothalamic Area Promotes Sleep: DREADD Activation in Wild-Type Rats. Cells 2022; 11:2140. [PMID: 35883583 PMCID: PMC9319714 DOI: 10.3390/cells11142140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/18/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
The ventrolateral preoptic area (VLPO) predominantly contains sleep-active neurons and is involved in sleep regulation. The perifornical-hypothalamic area (PF-HA) is a wake-regulatory region and predominantly contains wake-active neurons. VLPO GABAergic/galaninergic neurons project to the PF-HA. Previously, the specific contribution of VLPO neurons projecting to the PF-HA (VLPO > PF-HAPRJ) in sleep regulation in rats could not be investigated due to the lack of tools that could selectively target these neurons. We determined the contribution of VLPO > PF-HAPRJ neurons in sleep regulation by selectively activating them using designer receptors exclusively activated by designer drugs (DREADDs) in wild-type Fischer-344 rats. We used a combination of two viral vectors to retrogradely deliver the Cre-recombinase gene, specifically, in VLPO > PF-HA neurons, and further express hM3Dq in those neurons to selectively activate them for delineating their specific contributions to sleep−wake functions. Compared to the control, in DREADD rats, clozapine-N-oxide (CNO) significantly increased fos-expression, a marker of neuronal activation, in VLPO > PF-HAPRJ neurons (2% vs. 20%, p < 0.01) during the dark phase. CNO treatment also increased nonREM sleep (27% vs. 40%, p < 0.01) during the first 3 h of the dark phase, when rats are typically awake, and after exposure to the novel environment (55% vs. 65%; p < 0.01), which induces acute arousal during the light phase. These results support a hypothesis that VLPO > PF-HAPRJ neurons constitute a critical component of the hypothalamic sleep−wake regulatory circuitry and promote sleep by suppressing wake-active PF-HA neurons.
Collapse
Affiliation(s)
- Andrey Kostin
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, 16111 Plummer Street, Sepulveda, Los Angeles, CA 91343, USA; (A.K.); (M.A.A.); (D.M.)
| | - Md. Aftab Alam
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, 16111 Plummer Street, Sepulveda, Los Angeles, CA 91343, USA; (A.K.); (M.A.A.); (D.M.)
- Department of Psychiatry, University of California, Los Angeles, CA 90095, USA
| | - Anton Saevskiy
- Scientific Research and Technology Center for Neurotechnology, Southern Federal University, 344006 Rostov-on-Don, Russia;
| | - Dennis McGinty
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, 16111 Plummer Street, Sepulveda, Los Angeles, CA 91343, USA; (A.K.); (M.A.A.); (D.M.)
- Department of Psychology, University of California, Los Angeles, CA 90095, USA
| | - Md. Noor Alam
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, 16111 Plummer Street, Sepulveda, Los Angeles, CA 91343, USA; (A.K.); (M.A.A.); (D.M.)
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
17
|
Pandi-Perumal SR, Cardinali DP, Zaki NFW, Karthikeyan R, Spence DW, Reiter RJ, Brown GM. Timing is everything: Circadian rhythms and their role in the control of sleep. Front Neuroendocrinol 2022; 66:100978. [PMID: 35033557 DOI: 10.1016/j.yfrne.2022.100978] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/12/2021] [Accepted: 01/08/2022] [Indexed: 01/16/2023]
Abstract
Sleep and the circadian clock are intertwined and have persisted throughout history. The suprachiasmatic nucleus (SCN) orchestrates sleep by controlling circadian (Process C) and homeostatic (Process S) activities. As a "hand" on the endogenous circadian clock, melatonin is critical for sleep regulation. Light serves as a cue for sleep/wake control by activating retino-recipient cells in the SCN and subsequently suppressing melatonin. Clock genes are the molecular timekeepers that keep the 24 h cycle in place. Two main sleep and behavioural disorder diagnostic manuals have now officially recognised the importance of these processes for human health and well-being. The body's ability to respond to daily demands with the least amount of effort is maximised by carefully timing and integrating all components of sleep and waking. In the brain, the organization of timing is essential for optimal brain physiology.
Collapse
Affiliation(s)
- Seithikurippu R Pandi-Perumal
- Somnogen Canada Inc, College Street, Toronto, ON, Canada; Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Daniel P Cardinali
- Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, 1107 Buenos Aires, Argentina
| | - Nevin F W Zaki
- Department of Psychiatry, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | | | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Gregory M Brown
- Centre for Addiction and Mental Health, Molecular Brain Sciences, University of Toronto, 250 College St. Toronto, ON, Canada
| |
Collapse
|
18
|
Oesch LT, Adamantidis AR. How REM sleep shapes hypothalamic computations for feeding behavior. Trends Neurosci 2021; 44:990-1003. [PMID: 34663506 DOI: 10.1016/j.tins.2021.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/06/2021] [Accepted: 09/22/2021] [Indexed: 10/20/2022]
Abstract
The electrical activity of diverse brain cells is modulated across states of vigilance, namely wakefulness, non-rapid eye movement (NREM) sleep, and rapid eye movement (REM) sleep. Enhanced activity of neuronal circuits during NREM sleep impacts on subsequent awake behaviors, yet the significance of their activation, or lack thereof, during REM sleep remains unclear. This review focuses on feeding-promoting cells in the lateral hypothalamus (LH) that express the vesicular GABA and glycine transporter (vgat) as a model to further understand the impact of REM sleep on neural encoding of goal-directed behavior. It emphasizes both spatial and temporal aspects of hypothalamic cell dynamics across awake behaviors and REM sleep, and discusses a role for REM sleep in brain plasticity underlying energy homeostasis and behavioral optimization.
Collapse
Affiliation(s)
- Lukas T Oesch
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, Bern, Switzerland; Department of Biomedical Research, University of Bern, Bern, Switzerland; Department of Neurobiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Antoine R Adamantidis
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, Bern, Switzerland; Department of Biomedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
19
|
Blanco-Centurion C, Luo S, Vidal-Ortiz A, Swank C, Shiromani PJ. Activity of a subset of vesicular GABA-transporter neurons in the ventral zona incerta anticipates sleep onset. Sleep 2021; 44:6017820. [PMID: 33270105 DOI: 10.1093/sleep/zsaa268] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/01/2020] [Indexed: 01/03/2023] Open
Abstract
STUDY OBJECTIVES Sleep and wake are opposing behavioral states controlled by the activity of specific neurons that need to be located and mapped. To better understand how a waking brain falls asleep it is necessary to identify activity of individual phenotype-specific neurons, especially neurons that anticipate sleep onset. In freely behaving mice, we used microendoscopy to monitor calcium (Ca2+) fluorescence in individual hypothalamic neurons expressing the vesicular GABA transporter (vGAT), a validated marker of GABA neurons. METHODS vGAT-Cre mice (male = 3; female = 2) transfected with rAAV-FLEX-GCaMP6M in the lateral hypothalamus were imaged 30 days later during multiple episodes of waking (W), non-rapid-eye movement sleep (NREMS) or REMS (REMS). RESULTS 372 vGAT neurons were recorded in the zona incerta. 23.9% of the vGAT neurons showed maximal fluorescence during wake (classified as wake-max), 4% were NREM-max, 56.2% REM-max, 5.9% wake/REM max, while 9.9% were state-indifferent. In the NREM-max group, Ca2+ fluorescence began to increase before onset of NREM sleep, remained high throughout NREM sleep, and declined in REM sleep. CONCLUSIONS We found that 60.2% of the vGAT GABA neurons in the zona incerta had activity that was biased towards sleep (NREM and REMS). A subset of vGAT neurons (NREM-max) became active in advance of sleep onset and may induce sleep by inhibiting the activity of the arousal neurons. Abnormal activation of the NREM-max neurons may drive sleep attacks and hypersomnia.
Collapse
Affiliation(s)
- Carlos Blanco-Centurion
- Laboratory of Sleep Medicine and Chronobiology, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC
| | - SiWei Luo
- Laboratory of Sleep Medicine and Chronobiology, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC
| | | | - Colby Swank
- Laboratory of Sleep Medicine and Chronobiology, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC
| | - Priyattam J Shiromani
- Laboratory of Sleep Medicine and Chronobiology, Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC.,Ralph H. Johnson VA Medical Center, Charleston, SC
| |
Collapse
|
20
|
Gazea M, Furdan S, Sere P, Oesch L, Molnár B, Di Giovanni G, Fenno LE, Ramakrishnan C, Mattis J, Deisseroth K, Dymecki SM, Adamantidis AR, Lőrincz ML. Reciprocal Lateral Hypothalamic and Raphe GABAergic Projections Promote Wakefulness. J Neurosci 2021; 41:4840-4849. [PMID: 33888606 PMCID: PMC8260159 DOI: 10.1523/jneurosci.2850-20.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/15/2021] [Accepted: 02/28/2021] [Indexed: 01/06/2023] Open
Abstract
The lateral hypothalamus (LH), together with multiple neuromodulatory systems of the brain, such as the dorsal raphe nucleus (DR), is implicated in arousal, yet interactions between these systems are just beginning to be explored. Using a combination of viral tracing, circuit mapping, electrophysiological recordings from identified neurons, and combinatorial optogenetics in mice, we show that GABAergic neurons in the LH selectively inhibit GABAergic neurons in the DR, resulting in increased firing of a substantial fraction of its neurons that ultimately promotes arousal. These DRGABA neurons are wake active and project to multiple brain areas involved in the control of arousal, including the LH, where their specific activation potently influences local network activity leading to arousal from sleep. Our results show how mutual inhibitory projections between the LH and the DR promote wakefulness and suggest a complex arousal control by intimate interactions between long-range connections and local circuit dynamics.SIGNIFICANCE STATEMENT: Multiple brain systems including the lateral hypothalamus and raphe serotonergic system are involved in the regulation of the sleep/wake cycle, yet the interaction between these systems have remained elusive. Here we show that mutual disinhibition mediated by long range inhibitory projections between these brain areas can promote wakefulness. The main importance of this work relies in revealing the interaction between a brain area involved in autonomic regulation and another in controlling higher brain functions including reward, patience, mood and sensory coding.
Collapse
Affiliation(s)
- Mary Gazea
- Centre for Experimental Neurology, Department of Neurology, Inselspital, University Hospital Bern, University of Bern, Bern 3010, Switzerland
- Department of Biomedical Research, Inselspital University Hospital Bern, University of Bern, Bern 3010, Switzerland
| | - Szabina Furdan
- Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged 6726, Hungary
| | - Péter Sere
- Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged 6726, Hungary
- Department of Physiology, University of Szeged, Szeged 6720, Hungary
| | - Lukas Oesch
- Department of Biomedical Research, Inselspital University Hospital Bern, University of Bern, Bern 3010, Switzerland
- Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged 6726, Hungary
| | - Benedek Molnár
- Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged 6726, Hungary
- Department of Physiology, University of Szeged, Szeged 6720, Hungary
| | - Giuseppe Di Giovanni
- Neurosci ence Division, School of Bioscience, Cardiff University, Cardiff CF10 3AX, United Kingdom
- Department of Physiology and Biochemistry, University of Malta, MSD 2080, Malta
| | - Lief E Fenno
- Departments of Psychiatry & Behavioral Sciences and Bioengineering, Stanford University, Stanford 94305, California
| | | | - Joanna Mattis
- Departments of Psychiatry & Behavioral Sciences and Bioengineering, Stanford University, Stanford 94305, California
| | - Karl Deisseroth
- Departments of Psychiatry & Behavioral Sciences and Bioengineering, Stanford University, Stanford 94305, California
- Howard Hughes Medical Institute, Stanford University, Stanford 94305, California
| | - Susan M Dymecki
- Department of Genetics, Harvard Medical School, Boston 02115, Massachusetts
| | - Antoine R Adamantidis
- Centre for Experimental Neurology, Department of Neurology, Inselspital, University Hospital Bern, University of Bern, Bern 3010, Switzerland
- Department of Biomedical Research, Inselspital University Hospital Bern, University of Bern, Bern 3010, Switzerland
| | - Magor L Lőrincz
- Department of Physiology, Anatomy, and Neuroscience, University of Szeged, Szeged 6726, Hungary
- Department of Physiology, University of Szeged, Szeged 6720, Hungary
- Neurosci ence Division, School of Bioscience, Cardiff University, Cardiff CF10 3AX, United Kingdom
| |
Collapse
|
21
|
Kostin A, Alam MA, McGinty D, Alam MN. Adult hypothalamic neurogenesis and sleep-wake dysfunction in aging. Sleep 2021; 44:5986548. [PMID: 33202015 DOI: 10.1093/sleep/zsaa173] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/22/2020] [Indexed: 12/21/2022] Open
Abstract
In the mammalian brain, adult neurogenesis has been extensively studied in the hippocampal sub-granular zone and the sub-ventricular zone of the anterolateral ventricles. However, growing evidence suggests that new cells are not only "born" constitutively in the adult hypothalamus, but many of these cells also differentiate into neurons and glia and serve specific functions. The preoptic-hypothalamic area plays a central role in the regulation of many critical functions, including sleep-wakefulness and circadian rhythms. While a role for adult hippocampal neurogenesis in regulating hippocampus-dependent functions, including cognition, has been extensively studied, adult hypothalamic neurogenic process and its contributions to various hypothalamic functions, including sleep-wake regulation are just beginning to unravel. This review is aimed at providing the current understanding of the hypothalamic adult neurogenic processes and the extent to which it affects hypothalamic functions, including sleep-wake regulation. We propose that hypothalamic neurogenic processes are vital for maintaining the proper functioning of the hypothalamic sleep-wake and circadian systems in the face of regulatory challenges. Sleep-wake disturbance is a frequent and challenging problem of aging and age-related neurodegenerative diseases. Aging is also associated with a decline in the neurogenic process. We discuss a hypothesis that a decrease in the hypothalamic neurogenic process underlies the aging of its sleep-wake and circadian systems and associated sleep-wake disturbance. We further discuss whether neuro-regenerative approaches, including pharmacological and non-pharmacological stimulation of endogenous neural stem and progenitor cells in hypothalamic neurogenic niches, can be used for mitigating sleep-wake and other hypothalamic dysfunctions in aging.
Collapse
Affiliation(s)
- Andrey Kostin
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA
| | - Md Aftab Alam
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Psychiatry, University of California, Los Angeles, CA
| | - Dennis McGinty
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Psychology, University of California, Los Angeles, CA
| | - Md Noor Alam
- Research Service (151A3), Veterans Affairs Greater Los Angeles Healthcare System, Sepulveda, CA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| |
Collapse
|
22
|
Borniger JC, de Lecea L. Peripheral Lipopolyssacharide Rapidly Silences REM-Active LH GABA Neurons. Front Behav Neurosci 2021; 15:649428. [PMID: 33716686 PMCID: PMC7946974 DOI: 10.3389/fnbeh.2021.649428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/02/2021] [Indexed: 12/04/2022] Open
Abstract
Immune factors (e.g., cytokines, chemokines) can alter the activity of neuronal circuits to promote "sickness behavior," a suite of adaptive actions that organisms exhibit in response to infection/injury in order to maximize their chances of recovery (i.e., return to homeostasis). This includes drastic alterations in sleep/wake states, locomotor activity, and food intake, among other behaviors. Despite the ample evidence highlighting interactions between the brain and systemic immunity, studies on how immune challenges alter the activity of genetically defined cell populations controlling arousal states are scarce. As the lateral hypothalamus (LH) serves a major integrative function in behavioral arousal, food intake, and monitoring and responding to changes in systemic physiology, we investigated how GABAergic neurons within this brain region alter their activity across normal sleep/wake states and in response to a peripheral immune challenge with bacterial endotoxin [lipopolysaccharides (LPS)]. Using fiber photometry (GCaMP6s Ca2+ signal) in tandem with electroencephalogram (EEG)/EMG recordings to determine arousal states, we observed that population activity of GABAergic neurons in the lateral hypothalamus (LHGABA) is highest during rapid-eye-movement sleep (REM), and this activity changes drastically across spontaneous arousal state transitions, with the lowest activity observed during non-REM sleep. Upon intraperitoneal LPS challenge, LHGABA neurons rapidly decrease their activity in tandem with elimination of REM sleep behavior (characteristic of cytokine-induced sickness). Together, these data suggest that peripheral immune challenges can rapidly (in < 40 min) alter subcortical neuronal circuits controlling arousal states. Additionally, we demonstrate that fiber photometry offers a sensitive and cell-type specific tool that can be applied to study the neuronal substrates of sickness behavior.
Collapse
Affiliation(s)
- Jeremy C. Borniger
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
- Stanford University School of Medicine, Stanford, CA, United States
| | - Luis de Lecea
- Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
23
|
Oesch LT, Gazea M, Gent TC, Bandarabadi M, Gutierrez Herrera C, Adamantidis AR. REM sleep stabilizes hypothalamic representation of feeding behavior. Proc Natl Acad Sci U S A 2020; 117:19590-19598. [PMID: 32732431 PMCID: PMC7430996 DOI: 10.1073/pnas.1921909117] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
During rapid eye movement (REM) sleep, behavioral unresponsiveness contrasts strongly with intense brain-wide neural network dynamics. Yet, the physiological functions of this cellular activation remain unclear. Using in vivo calcium imaging in freely behaving mice, we found that inhibitory neurons in the lateral hypothalamus (LHvgat) show unique activity patterns during feeding that are reactivated during REM, but not non-REM, sleep. REM sleep-specific optogenetic silencing of LHvgat cells induced a reorganization of these activity patterns during subsequent feeding behaviors accompanied by decreased food intake. Our findings provide evidence for a role for REM sleep in the maintenance of cellular representations of feeding behavior.
Collapse
Affiliation(s)
- Lukas T Oesch
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, CH-3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, CH-3010 Bern, Switzerland
| | - Mary Gazea
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, CH-3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, CH-3010 Bern, Switzerland
| | - Thomas C Gent
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, CH-3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, CH-3010 Bern, Switzerland
| | - Mojtaba Bandarabadi
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, CH-3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, CH-3010 Bern, Switzerland
| | - Carolina Gutierrez Herrera
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, CH-3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, CH-3010 Bern, Switzerland
| | - Antoine R Adamantidis
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, CH-3010 Bern, Switzerland;
- Department of Biomedical Research, University of Bern, CH-3010 Bern, Switzerland
| |
Collapse
|
24
|
|
25
|
Li SB, de Lecea L. The hypocretin (orexin) system: from a neural circuitry perspective. Neuropharmacology 2020; 167:107993. [PMID: 32135427 DOI: 10.1016/j.neuropharm.2020.107993] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/23/2020] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
Hypocretin/orexin neurons are distributed restrictively in the hypothalamus, a brain region known to orchestrate diverse functions including sleep, reward processing, food intake, thermogenesis, and mood. Since the hypocretins/orexins were discovered more than two decades ago, extensive studies have accumulated concrete evidence showing the pivotal role of hypocretin/orexin in diverse neural modulation. New method of viral-mediated tracing system offers the possibility to map the monosynaptic inputs and detailed anatomical connectivity of Hcrt neurons. With the development of powerful research techniques including optogenetics, fiber-photometry, cell-type/pathway specific manipulation and neuronal activity monitoring, as well as single-cell RNA sequencing, the details of how hypocretinergic system execute functional modulation of various behaviors are coming to light. In this review, we focus on the function of neural pathways from hypocretin neurons to target brain regions. Anatomical and functional inputs to hypocretin neurons are also discussed. We further briefly summarize the development of pharmaceutical compounds targeting hypocretin signaling. This article is part of the special issue on Neuropeptides.
Collapse
Affiliation(s)
- Shi-Bin Li
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305, USA.
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305, USA.
| |
Collapse
|
26
|
Jones BE. Arousal and sleep circuits. Neuropsychopharmacology 2020; 45:6-20. [PMID: 31216564 PMCID: PMC6879642 DOI: 10.1038/s41386-019-0444-2] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/16/2019] [Accepted: 06/11/2019] [Indexed: 12/20/2022]
Abstract
The principal neurons of the arousal and sleep circuits are comprised by glutamate and GABA neurons, which are distributed within the reticular core of the brain and, through local and distant projections and interactions, regulate cortical activity and behavior across wake-sleep states. These are in turn modulated by the neuromodulatory systems that are comprised by acetylcholine, noradrenaline, dopamine, serotonin, histamine, orexin (hypocretin), and melanin-concentrating hormone (MCH) neurons. Glutamate and GABA neurons are heterogeneous in their profiles of discharge, forming distinct functional cell types by selective or maximal discharge during (1) waking and paradoxical (REM) sleep, (2) during slow wave sleep, (3) during waking, or (4) during paradoxical (REM) sleep. The neuromodulatory systems are each homogeneous in their profile of discharge, the majority discharging maximally during waking and paradoxical sleep or during waking. Only MCH neurons discharge maximally during sleep. They each exert their modulatory influence upon other neurons through excitatory and inhibitory receptors thus effecting a concerted differential change in the functionally different cell groups. Both arousal and sleep circuit neurons are homeostatically regulated as a function of their activity in part through changes in receptors. The major pharmacological agents used for the treatment of wake and sleep disorders act upon GABA and neuromodulatory transmission.
Collapse
Affiliation(s)
- Barbara E. Jones
- 0000 0004 1936 8649grid.14709.3bDepartment of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4 Canada
| |
Collapse
|
27
|
Venner A, De Luca R, Sohn LT, Bandaru SS, Verstegen AMJ, Arrigoni E, Fuller PM. An Inhibitory Lateral Hypothalamic-Preoptic Circuit Mediates Rapid Arousals from Sleep. Curr Biol 2019; 29:4155-4168.e5. [PMID: 31761703 DOI: 10.1016/j.cub.2019.10.026] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/10/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022]
Abstract
Among the neuronal populations implicated in sleep-wake control, the ventrolateral preoptic (VLPO) nucleus has emerged as a key sleep-promoting center. However, the synaptic drives that regulate the VLPO to control arousal levels in vivo have not to date been identified. Here, we show that sleep-promoting galaninergic neurons within the VLPO nucleus, defined pharmacologically and by single-cell transcript analysis, are postsynaptic targets of lateral hypothalamic GABAergic (LHGABA) neurons and that activation of this pathway in vivo rapidly drives wakefulness. Ca2+ imaging from LHGABA neurons indicate that they are both wake and rapid eye movement (REM)-sleep active. Consistent with the potent arousal-promoting property of the LHGABA → VLPO pathway, presynaptic inputs to LHGABA neurons originate from several canonical stress- and arousal-related network nodes. This work represents the first demonstration that direct synaptic inhibition of the VLPO area can suppress sleep-promoting neurons to rapidly promote arousal.
Collapse
Affiliation(s)
- Anne Venner
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Roberto De Luca
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Lauren T Sohn
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Sathyajit S Bandaru
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Anne M J Verstegen
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Patrick M Fuller
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
28
|
Yonemochi N, Ardianto C, Ueda D, Kamei J, Ikeda H. GABAergic function in the lateral hypothalamus regulates feeding behavior: Possible mediation via orexin. Neuropsychopharmacol Rep 2019; 39:289-296. [PMID: 31618533 PMCID: PMC7292314 DOI: 10.1002/npr2.12080] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/22/2019] [Accepted: 09/24/2019] [Indexed: 11/17/2022] Open
Abstract
Aim The lateral hypothalamus (LH) is known as the hunger center, but the mechanisms through which the LH regulates food intake are unclear. Since GABA neurons are reported to project to the LH, the present study investigated the role of GABAergic function in the LH in the regulation of feeding behavior. Methods GABA levels in the LH were measured by in vivo microdialysis. Food intake after drug injection into the LH was measured every 1 hour for 4 hours. The mRNA levels were measured using RT‐PCR. Results Food intake significantly increased GABA levels in the LH, suggesting that food intake stimulates GABAergic function in the LH. Injection of the GABAA receptor agonist muscimol into the LH significantly inhibited food intake, whereas injection of the GABAA receptor antagonist bicuculline into the LH did not significantly affect food intake. The inhibitory effect of muscimol injected into the LH was blocked by co‐administration of bicuculline. These results indicate that the stimulation of GABAA receptors in the LH inhibits food intake. We next examined whether the stimulation of GABAA receptors affects hypothalamic neuropeptides that are known to regulate feeding behavior. The injection of muscimol significantly decreased preproorexin mRNA in the hypothalamus. Conclusion These results indicate that food intake activates GABAergic function in the LH, which terminates feeding behavior by stimulating GABAA receptors. Moreover, it is suggested that the stimulation of GABAA receptors in the LH reduces food intake through inhibition of orexin neurons. We investigated whether GABAergic function in the lateral hypothalamus (LH) regulates feeding behavior. We showed that food intake increased GABA levels in the LH in in vivo microdialysis and that the GABAA receptor agonist muscimol injected into the LH decreased food intake. These results suggest that food intake activates GABA neurons projecting to the LH, and it terminates feeding behavior through GABAA receptors.![]()
Collapse
Affiliation(s)
- Naomi Yonemochi
- Department of Pathophysiology and Therapeutics, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | - Chrismawan Ardianto
- Department of Pathophysiology and Therapeutics, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | - Daiki Ueda
- Department of Pathophysiology and Therapeutics, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | - Junzo Kamei
- Department of Pathophysiology and Therapeutics, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan.,Department of Biomolecular Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | - Hiroko Ikeda
- Department of Pathophysiology and Therapeutics, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| |
Collapse
|
29
|
Shi Y, Xiao D, Dai L, Si Y, Fang Q, Wei X. The hypnotic effect of propofol involves inhibition of GABAergic neurons in the lateral hypothalamus. Neuroreport 2019; 30:927-932. [PMID: 31469720 DOI: 10.1097/wnr.0000000000001292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Propofol is widely used for induction and maintenance of anaesthesia, which causes a rapid loss of consciousness. So far the mechanisms underlying the effect of propofol are still largely unknown. Here, we found that microinjection of propofol in the lateral hypothalamus caused a significant decrease in wakefulness and an increase in the amount of non-rapid eye movement sleep and rapid eye movement sleep. Application of propofol in the lateral hypothalamus affected the electroencephalogram power spectra with a decrease in theta oscillations and an increase in the delta oscillations. Additionally, using whole-cell patch clamp recording, we found propofol inhibited the excitability of the GABAergic neurons in the lateral hypothalamus, which plays a critical role in controlling wakefulness. Altogether, these findings indicate that propofol targets lateral hypothalamus and generates a hypnotic state, which might involve the inhibition of GABAergic neurons. These results provide a novel mechanism to explain propofol-elicited anaesthesia.
Collapse
Affiliation(s)
- Yihua Shi
- Department of General Surgery, The First People's Hospital of WenLing, Wenling, Zhejiang
| | - Deshuang Xiao
- Department of General Surgery, The First People's Hospital of WenLing, Wenling, Zhejiang
| | - Lingbo Dai
- Department of General Surgery, The First People's Hospital of WenLing, Wenling, Zhejiang
| | - Yongyu Si
- Department of Anesthesiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Qian Fang
- Department of General Surgery, The First People's Hospital of WenLing, Wenling, Zhejiang
| | - Xing Wei
- Department of General Surgery, The First People's Hospital of WenLing, Wenling, Zhejiang
- Department of Anesthesiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
30
|
Abstract
Over the past decade, basic sleep research investigating the circuitry controlling sleep and wakefulness has been boosted by pharmacosynthetic approaches, including chemogenetic techniques using designed receptors exclusively activated by designer drugs (DREADD). DREADD offers a series of tools that selectively control neuronal activity as a way to probe causal relationship between neuronal sub-populations and the regulation of the sleep-wake cycle. Following the path opened by optogenetics, DREADD tools applied to discrete neuronal sub-populations in numerous brain areas quickly made their contribution to the discovery and the expansion of our understanding of critical brain structures involved in a wide variety of behaviors and in the control of vigilance state architecture.
Collapse
|
31
|
Molecular codes and in vitro generation of hypocretin and melanin concentrating hormone neurons. Proc Natl Acad Sci U S A 2019; 116:17061-17070. [PMID: 31375626 PMCID: PMC6708384 DOI: 10.1073/pnas.1902148116] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hypocretin/orexin (HCRT) and melanin concentrating hormone (MCH) neuropeptides are exclusively produced by the lateral hypothalamus and play important roles in sleep, metabolism, reward, and motivation. Loss of HCRT (ligands or receptors) causes the sleep disorder narcolepsy with cataplexy in humans and in animal models. How these neuropeptides are produced and involved in diverse functions remain unknown. Here, we developed methods to sort and purify HCRT and MCH neurons from the mouse late embryonic hypothalamus. RNA sequencing revealed key factors of fate determination for HCRT (Peg3, Ahr1, Six6, Nr2f2, and Prrx1) and MCH (Lmx1, Gbx2, and Peg3) neurons. Loss of Peg3 in mice significantly reduces HCRT and MCH cell numbers, while knock-down of a Peg3 ortholog in zebrafish completely abolishes their expression, resulting in a 2-fold increase in sleep amount. We also found that loss of HCRT neurons in Hcrt-ataxin-3 mice results in a specific 50% decrease in another orexigenic neuropeptide, QRFP, that might explain the metabolic syndrome in narcolepsy. The transcriptome results were used to develop protocols for the production of HCRT and MCH neurons from induced pluripotent stem cells and ascorbic acid was found necessary for HCRT and BMP7 for MCH cell differentiation. Our results provide a platform to understand the development and expression of HCRT and MCH and their multiple functions in health and disease.
Collapse
|
32
|
Cabanas M, Pistono C, Puygrenier L, Rakesh D, Jeantet Y, Garret M, Cho YH. Neurophysiological and Behavioral Effects of Anti-Orexinergic Treatments in a Mouse Model of Huntington's Disease. Neurotherapeutics 2019; 16:784-796. [PMID: 30915710 PMCID: PMC6694444 DOI: 10.1007/s13311-019-00726-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Huntington's disease (HD) is associated with sleep and circadian disturbances in addition to hallmark motor and cognitive impairments. Electrophysiological studies on HD mouse models have revealed an aberrant oscillatory activity at the beta frequency, during sleep, that is associated with HD pathology. Moreover, HD animal models display an abnormal sleep-wake cycle and sleep fragmentation. In this study, we investigated a potential involvement of the orexinergic system dysfunctioning in sleep-wake and circadian disturbances and abnormal network (i.e., beta) activity in the R6/1 mouse model. We found that the age at which orexin activity starts to deviate from normal activity pattern coincides with that of sleep disturbances as well as the beta activity. We also found that acute administration of Suvorexant, an orexin 1 and orexin 2 receptor antagonist, was sufficient to decrease the beta power significantly and to improve sleep in R6/1 mice. In addition, a 5-day treatment paradigm alleviated cognitive deficits and induced a gain of body weight in female HD mice. These results suggest that restoring normal activity of the orexinergic system could be an efficient therapeutic solution for sleep and behavioral disturbances in HD.
Collapse
Affiliation(s)
- Magali Cabanas
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France
| | - Cristiana Pistono
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France
| | - Laura Puygrenier
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France
| | - Divyangana Rakesh
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France
| | - Yannick Jeantet
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France
| | - Maurice Garret
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France
| | - Yoon H Cho
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, CNRS UMR 5287, Allee Geoffroy St Hilaire, CS 50023, 33615, Pessac Cedex, France.
- Institute of Cognitive and Integrative Neuroscience of Aquitaine, University of Bordeaux, Bordeaux, France.
| |
Collapse
|
33
|
Naganuma F, Kroeger D, Bandaru SS, Absi G, Madara JC, Vetrivelan R. Lateral hypothalamic neurotensin neurons promote arousal and hyperthermia. PLoS Biol 2019; 17:e3000172. [PMID: 30893297 PMCID: PMC6426208 DOI: 10.1371/journal.pbio.3000172] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 02/13/2019] [Indexed: 01/19/2023] Open
Abstract
Sleep and wakefulness are greatly influenced by various physiological and psychological factors, but the neuronal elements responsible for organizing sleep-wake behavior in response to these factors are largely unknown. In this study, we report that a subset of neurons in the lateral hypothalamic area (LH) expressing the neuropeptide neurotensin (Nts) is critical for orchestrating sleep-wake responses to acute psychological and physiological challenges or stressors. We show that selective activation of NtsLH neurons with chemogenetic or optogenetic methods elicits rapid transitions from non-rapid eye movement (NREM) sleep to wakefulness and produces sustained arousal, higher locomotor activity (LMA), and hyperthermia, which are commonly observed after acute stress exposure. On the other hand, selective chemogenetic inhibition of NtsLH neurons attenuates the arousal, LMA, and body temperature (Tb) responses to a psychological stress (a novel environment) and augments the responses to a physiological stress (fasting). A neurotensin-producing subset of neurons in the lateral hypothalamus promote arousal and thermogenesis; these neurons are necessary for appropriate sleep-wake and body temperature responses to various stressors. Adjusting sleep-wake behavior in response to environmental and physiological challenges may not only be of protective value, but can also be vital for the survival of the organism. For example, while it is crucial to increase wake to explore a novel environment to search for potential threats and food sources, it is also necessary to decrease wake and reduce energy expenditure during prolonged absence of food. In this study, we report that a subset of neurons in the lateral hypothalamic area (LH) expressing the neuropeptide neurotensin (Nts) is critical for orchestrating sleep-wake responses to such challenges. We show that brief activation of NtsLH neurons in mice evokes immediate arousals from sleep, while their sustained activation increases wake, locomotor activity, and body temperature for several hours. In contrast, when NtsLH neurons are inhibited, mice are neither able to sustain wake in a novel environment nor able to reduce wake during food deprivation. These data suggest that NtsLH neurons may be necessary for generating appropriate sleep-wake responses to a wide variety of environmental and physiological challenges.
Collapse
Affiliation(s)
- Fumito Naganuma
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Daniel Kroeger
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sathyajit S. Bandaru
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Gianna Absi
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Joseph C. Madara
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Ramalingam Vetrivelan
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
34
|
Mickelsen LE, Bolisetty M, Chimileski BR, Fujita A, Beltrami EJ, Costanzo JT, Naparstek JR, Robson P, Jackson AC. Single-cell transcriptomic analysis of the lateral hypothalamic area reveals molecularly distinct populations of inhibitory and excitatory neurons. Nat Neurosci 2019; 22:642-656. [PMID: 30858605 DOI: 10.1038/s41593-019-0349-8] [Citation(s) in RCA: 215] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 01/30/2019] [Indexed: 01/01/2023]
Abstract
The lateral hypothalamic area (LHA) coordinates an array of fundamental behaviors, including sleeping, waking, feeding, stress and motivated behavior. The wide spectrum of functions ascribed to the LHA may be explained by a heterogeneous population of neurons, the full diversity of which is poorly understood. We employed a droplet-based single-cell RNA-sequencing approach to develop a comprehensive census of molecularly distinct cell types in the mouse LHA. Neuronal populations were classified based on fast neurotransmitter phenotype and expression of neuropeptides, transcription factors and synaptic proteins, among other gene categories. We define 15 distinct populations of glutamatergic neurons and 15 of GABAergic neurons, including known and novel cell types. We further characterize a novel population of somatostatin-expressing neurons through anatomical and behavioral approaches, identifying a role for these neurons in specific forms of innate locomotor behavior. This study lays the groundwork for better understanding the circuit-level underpinnings of LHA function.
Collapse
Affiliation(s)
- Laura E Mickelsen
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA.,Connecticut Institute for the Brain and Cognitive Sciences, Storrs, CT, USA
| | - Mohan Bolisetty
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.,Bristol-Myers Squibb, Pennington, NJ, USA
| | - Brock R Chimileski
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Akie Fujita
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA.,Connecticut Institute for the Brain and Cognitive Sciences, Storrs, CT, USA.,Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Eric J Beltrami
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - James T Costanzo
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Jacob R Naparstek
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA. .,Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA. .,Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA.
| | - Alexander C Jackson
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA. .,Connecticut Institute for the Brain and Cognitive Sciences, Storrs, CT, USA. .,Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA.
| |
Collapse
|
35
|
Abstract
The functions of sleep remain a mystery. Yet they must be important since sleep is highly conserved, and its chronic disruption is associated with various metabolic, psychiatric, and neurodegenerative disorders. This review will cover our evolving understanding of the mechanisms by which sleep is controlled and the complex relationship between sleep and disease states.
Collapse
Affiliation(s)
- William J Joiner
- Department of Pharmacology, Biomedical Sciences Graduate Program, Neurosciences Graduate Program, and Center for Circadian Biology, University of California San Diego , La Jolla, California
| |
Collapse
|
36
|
Abstract
In the present chapter, hypotheses on the mechanisms responsible for the genesis of the three vigilance states, namely, waking, non-rapid eye movement (non-REM) also called slow-wave sleep (SWS), and REM sleep also called paradoxical sleep (PS), are presented. A huge number of studies first indicate that waking is induced by the activation of multiple waking systems, including the serotonergic, noradrenergic, cholinergic, and hypocretin systems. At the onset of sleep, the SWS-active neurons would be activated by the circadian clock localized in the suprachiasmatic nucleus and a hypnogenic factor, adenosine, which progressively accumulates in the brain during waking. A number of studies support the hypothesis that SWS results from the activation of GABAergic neurons localized in the ventrolateral preoptic nucleus (VLPO). However, new GABAergic systems recently described localized in the parafacial, accumbens, and reticular thalamic nuclei will be also presented. In addition, we will show that a large body of data strongly suggests that the switch from SWS to PS is due to the interaction of multiple populations of glutamatergic and GABAergic neurons localized in the posterior hypothalamus and the brainstem.
Collapse
Affiliation(s)
- Pierre-Hervé Luppi
- Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, Lyon, France.
- University Lyon 1, Lyon, France.
| | - Patrice Fort
- Team "Physiopathologie des réseaux neuronaux responsables du cycle veille-sommeil", INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, Lyon, France
- University Lyon 1, Lyon, France
| |
Collapse
|
37
|
Arrigoni E, Chee MJS, Fuller PM. To eat or to sleep: That is a lateral hypothalamic question. Neuropharmacology 2018; 154:34-49. [PMID: 30503993 DOI: 10.1016/j.neuropharm.2018.11.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 12/15/2022]
Abstract
The lateral hypothalamus (LH) is a functionally and anatomically complex brain region that is involved in the regulation of many behavioral and physiological processes including feeding, arousal, energy balance, stress, reward and motivated behaviors, pain perception, body temperature regulation, digestive functions and blood pressure. Despite noteworthy experimental efforts over the past decades, the circuit, cellular and synaptic bases by which these different processes are regulated by the LH remains incompletely understood. This knowledge gap links in large part to the high cellular heterogeneity of the LH. Fortunately, the rapid evolution of newer genetic and electrophysiological tools is now permitting the selective manipulation, typically genetically-driven, of discrete LH cell populations. This, in turn, permits not only assignment of function to discrete cell groups, but also reveals that considerable synergistic and antagonistic interactions exist between key LH cell populations that regulate feeding and arousal. For example, we now know that while LH melanin-concentrating hormone (MCH) and orexin/hypocretin neurons both function as sensors of the internal metabolic environment, their roles regulating sleep and arousal are actually opposing. Additional studies have uncovered similarly important roles for subpopulations of LH GABAergic cells in the regulation of both feeding and arousal. Herein we review the role of LH MCH, orexin/hypocretin and GABAergic cell populations in the regulation of energy homeostasis (including feeding) and sleep-wake and discuss how these three cell populations, and their subpopulations, may interact to optimize and coordinate metabolism, sleep and arousal. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.
Collapse
Affiliation(s)
- Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02215, USA.
| | - Melissa J S Chee
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | - Patrick M Fuller
- Department of Neurology, Beth Israel Deaconess Medical Center, Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02215, USA
| |
Collapse
|
38
|
Liu JJ, Mirabella VR, Pang ZP. Cell type- and pathway-specific synaptic regulation of orexin neurocircuitry. Brain Res 2018; 1731:145974. [PMID: 30296428 DOI: 10.1016/j.brainres.2018.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 12/19/2022]
Abstract
Orexin-expressing neurons are located exclusively in the lateral hypothalamic and perifornical areas and exhibit complex connectivity. The intricate wiring pattern is evident from a diverse function for orexin neurons in regulating many physiological processes and behaviors including sleep, metabolism, circadian cycles, anxiety, and reward. Nevertheless, the precise synaptic and circuitry-level mechanisms mediating these processes remain enigmatic, partially due to the wide spread connectivity of the orexin system, complex neurochemistry of orexin neurons, and previous lack of suitable tools to address its complexity. Here we summarize recent advances, focusing on synaptic regulatory mechanisms in the orexin neurocircuitry, including both the synaptic inputs to orexin neurons as well as their downstream targets in the brain. A clear and detailed elucidation of these mechanisms will likely provide novel insight into how dysfunction in orexin-mediated signaling leads to human disease and may ultimately be treated with more precise strategies.
Collapse
Affiliation(s)
- Jing-Jing Liu
- Child Health Institute of New Jersey, Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
| | - Vincent R Mirabella
- Child Health Institute of New Jersey, Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Zhiping P Pang
- Child Health Institute of New Jersey, Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| |
Collapse
|
39
|
Latifi B, Adamantidis A, Bassetti C, Schmidt MH. Sleep-Wake Cycling and Energy Conservation: Role of Hypocretin and the Lateral Hypothalamus in Dynamic State-Dependent Resource Optimization. Front Neurol 2018; 9:790. [PMID: 30344503 PMCID: PMC6183196 DOI: 10.3389/fneur.2018.00790] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/31/2018] [Indexed: 12/23/2022] Open
Abstract
The hypocretin (Hcrt) system has been implicated in a wide range of physiological functions from sleep-wake regulation to cardiovascular, behavioral, metabolic, and thermoregulagtory control. These wide-ranging physiological effects have challenged the identification of a parsimonious function for Hcrt. A compelling hypothesis suggests that Hcrt plays a role in the integration of sleep-wake neurophysiology with energy metabolism. For example, Hcrt neurons promote waking and feeding, but are also sensors of energy balance. Loss of Hcrt function leads to an increase in REM sleep propensity, but a potential role for Hcrt linking energy balance with REM sleep expression has not been addressed. Here we examine a potential role for Hcrt and the lateral hypothalamus (LH) in state-dependent resource allocation as a means of optimizing resource utilization and, as a result, energy conservation. We review the energy allocation hypothesis of sleep and how state-dependent metabolic partitioning may contribute toward energy conservation, but with additional examination of how the loss of thermoregulatory function during REM sleep may impact resource optimization. Optimization of energy expenditures at the whole organism level necessitates a top-down network responsible for coordinating metabolic operations in a state-dependent manner across organ systems. In this context, we then specifically examine the potential role of the LH in regulating this output control, including the contribution from both Hcrt and melanin concentrating hormone (MCH) neurons among a diverse LH cell population. We propose that this hypothalamic integration system is responsible for global shifts in state-dependent resource allocations, ultimately promoting resource optimization and an energy conservation function of sleep-wake cycling.
Collapse
Affiliation(s)
- Blerina Latifi
- Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Antoine Adamantidis
- Department of Neurology, Center for Experimental Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department of Biomedical Research, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Claudio Bassetti
- Department of Neurology, Center for Experimental Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Markus H Schmidt
- Department of Neurology, Center for Experimental Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Ohio Sleep Medicine Institute, Dublin, OH, United States
| |
Collapse
|
40
|
Discharge and Role of Acetylcholine Pontomesencephalic Neurons in Cortical Activity and Sleep-Wake States Examined by Optogenetics and Juxtacellular Recording in Mice. eNeuro 2018; 5:eN-CFN-0270-18. [PMID: 30225352 PMCID: PMC6140114 DOI: 10.1523/eneuro.0270-18.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/01/2018] [Accepted: 08/05/2018] [Indexed: 01/23/2023] Open
Abstract
Acetylcholine (ACh) neurons in the pontomesencephalic tegmentum (PMT) are thought to play an important role in promoting cortical activation with waking (W) and paradoxical sleep [PS; or rapid eye movement (REM)], but have yet to be proven to do so by selective stimulation and simultaneous recording of identified ACh neurons. Here, we employed optogenetics combined with juxtacellular recording and labeling of neurons in transgenic (TG) mice expressing ChR2 in choline acetyltransferase (ChAT)-synthesizing neurons. We established in vitro then in vivo in anesthetized (A) and unanesthetized (UA), head-fixed mice that photostimulation elicited a spike with short latency in neurons which could be identified by immunohistochemical staining as ACh neurons within the laterodorsal (LDT)/sublaterodorsal (SubLDT) and pedunculopontine tegmental (PPT) nuclei. Continuous light pulse stimulation during sleep evoked tonic spiking by ACh neurons that elicited a shift from irregular slow wave activity to rhythmic θ and enhanced γ activity on the cortex without behavioral arousal. With θ frequency rhythmic light pulse stimulation, ACh neurons discharged in bursts that occurred in synchrony with evoked cortical θ. During natural sleep-wake states, they were virtually silent during slow wave sleep (SWS), discharged in bursts during PS and discharged tonically during W. Yet, their bursting during PS was not rhythmic or synchronized with cortical θ but associated with phasic whisker movements. We conclude that ACh PMT neurons promote θ and γ cortical activity during W and PS by their tonic or phasic discharge through release of ACh onto local neurons within the PMT and/or more distant targets in the hypothalamus and thalamus.
Collapse
|
41
|
Abstract
A great pioneer in sleep research, Michel Jouvet applied rigorous scientific methods to the study of sleep-wake states and associated changes in consciousness which, with his vivid imagination and creative mind, he unveiled as the mysteries of sleep and waking such as to inspire a generation of researchers in the field. His initial discovery of a third state distinguished from waking (W) and slow wave sleep (SWS) by the paradoxical association of W-like cortical activity with sleep-like behavior and muscle atonia that he accordingly called "paradoxical sleep" (PS) began his investigation over some 50 years of the mechanisms of these three sleep-wake states. Using primarily lesion and pharmacological manipulations, he sought the systems which are necessary and sufficient, and he thereby provided an early blueprint of how the neuromodulatory systems could determine the sleep-wake states. With the application of increasingly more selective lesion and other advanced techniques including, notably, single unit recording combined with histochemical identification of recorded units, the monoamines and acetylcholine, together with peptidergic systems have been revealed to play modulatory, yet not essential, roles acting upon other intermingled glutamatergic and GABAergic neurons that are the effector neurons of the sleep-wake states and their cortical and behavioral correlates.
Collapse
Affiliation(s)
- Barbara E Jones
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC, H3A 2B4, Canada.
| |
Collapse
|
42
|
Sakai K. Behavioural state-specific neurons in the mouse medulla involved in sleep-wake switching. Eur J Neurosci 2018; 47:1482-1503. [PMID: 29791042 DOI: 10.1111/ejn.13963] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/12/2018] [Accepted: 04/30/2018] [Indexed: 11/28/2022]
Abstract
The medullary reticular formation (RF) is involved in the maintenance of several vital physiological functions and level of vigilance. In this study, in nonanesthetised, head-fixed mice, I examined the role of medullary RF neurons in the control of sleep-wake states, that is, wakefulness (W), slow-wave sleep (SWS) and paradoxical (or rapid eye movement) sleep (PS). I showed, for the first time, that the mouse medullary RF contains presumed SWS-promoting, SWS-on neurons that remain silent during W, display a sharp increase in discharge rate at sleep onset, and discharge tonically and selectively during SWS. In addition, I showed the presence in the medullary RF of both PS-on and PS-off neurons, which, respectively, commence discharging or cease firing selectively just prior to, and during, PS. PS-off neurons were located in the raphe nuclei and ventral medulla, while PS-on neurons were found in both the lateral part of the ventral gigantocellular reticular nucleus and the raphe nuclei, as were SWS-on neurons. PS-off and SWS-on neurons appear to play an important role in both the W-SWS and SWS-PS switches, while PS-on and PS-off neurons play an important role in the PS-W switch. The present findings on the trends in spike activity at the transitions from SWS to PS and from PS to W are in line with the reciprocal interaction hypothesis according to which PS occurs as a result of the cessation of discharge of PS-off neurons, while PS ends as a result of the start of discharge of PS-off neurons.
Collapse
Affiliation(s)
- Kazuya Sakai
- Integrative Physiology of the Brain Arousal System, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR5292, School of Medicine, Claude Bernard University, Lyon, France
| |
Collapse
|
43
|
Adamantidis A, Lüthi A. Optogenetic Dissection of Sleep-Wake States In Vitro and In Vivo. Handb Exp Pharmacol 2018; 253:125-151. [PMID: 29687163 DOI: 10.1007/164_2018_94] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Optogenetic tools have revolutionized insights into the fundamentals of brain function. This is particularly true for our current understanding of sleep-wake regulation and sleep rhythms. This is illustrated here through a comprehensive and step-by-step review over the major brain areas involved in transitions between sleep and wake states and in sleep rhythmogenesis.
Collapse
Affiliation(s)
- Antoine Adamantidis
- Department of Neurology, Inselspital University Hospital, University of Bern, Bern, Switzerland. .,Department of Clinical Research (DKF), University of Bern, Bern, Switzerland.
| | - Anita Lüthi
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
44
|
Regulation of Lateral Hypothalamic Orexin Activity by Local GABAergic Neurons. J Neurosci 2018; 38:1588-1599. [PMID: 29311142 DOI: 10.1523/jneurosci.1925-17.2017] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 12/04/2017] [Accepted: 12/27/2017] [Indexed: 12/31/2022] Open
Abstract
Orexin (also known as hypocretin) neurons are considered a key component of the ascending arousal system. They are active during wakefulness, at which time they drive and maintain arousal, and are silent during sleep. Their activity is controlled by long-range inputs from many sources, as well as by more short-range inputs, including from presumptive GABAergic neurons in the lateral hypothalamus/perifornical region (LH/PF). To characterize local GABAergic input to orexin neurons, we used channelrhodopsin-2-assisted circuit mapping in brain slices. We expressed channelrhodopsin-2 in GABAergic neurons (Vgat+) in the LH/PF and recorded from genetically identified surrounding orexin neurons (LH/PFVgat → Orx). We performed all experiments in mice of either sex. Photostimulation of LH/PF GABAergic neurons inhibited the firing of orexin neurons through the release of GABA, evoking GABAA-mediated IPSCs in orexin neurons. These photo-evoked IPSCs were maintained in the presence of TTX, indicating direct connectivity. Carbachol inhibited LH/PFVgat → Orx input through muscarinic receptors. By contrast, application of orexin was without effect on LH/PFVgat → Orx input, whereas dynorphin, another peptide produced by orexin neurons, inhibited LH/PFVgat → Orx input through κ-opioid receptors. Our results demonstrate that orexin neurons are under inhibitory control by local GABAergic neurons and that this input is depressed by cholinergic signaling, unaffected by orexin and inhibited by dynorphin. We propose that local release of dynorphin may, via collaterals, provides a positive feedback to orexin neurons and that, during wakefulness, orexin neurons may be disinhibited by acetylcholine and by their own release of dynorphin.SIGNIFICANCE STATEMENT The lateral hypothalamus contains important wake-promoting cell populations, including orexin-producing neurons. Intermingled with the orexin neurons, there are other cell populations that selectively discharge during nonrapid eye movement or rapid eye movement sleep. Some of these sleep-active neurons release GABA and are thought to inhibit wake-active neurons during rapid eye movement and nonrapid eye movement sleep. However, this hypothesis had not been tested. Here we show that orexin neurons are inhibited by a local GABAergic input. We propose that this local GABAergic input inhibits orexin neurons during sleep but that, during wakefulness, this input is depressed, possibly through cholinergically mediated disinhibition and/or by release of dynorphin from orexin neurons themselves.
Collapse
|
45
|
Homeostatic Changes in GABA and Acetylcholine Muscarinic Receptors on GABAergic Neurons in the Mesencephalic Reticular Formation following Sleep Deprivation. eNeuro 2018; 4:eN-NWR-0269-17. [PMID: 29302615 PMCID: PMC5752701 DOI: 10.1523/eneuro.0269-17.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/04/2017] [Accepted: 11/07/2017] [Indexed: 01/04/2023] Open
Abstract
We have examined whether GABAergic neurons in the mesencephalic reticular formation (RFMes), which are believed to inhibit the neurons in the pons that generate paradoxical sleep (PS or REMS), are submitted to homeostatic regulation under conditions of sleep deprivation (SD) by enforced waking during the day in mice. Using immunofluorescence, we investigated first, by staining for c-Fos, whether GABAergic RFMes neurons are active during SD and then, by staining for receptors, whether their activity is associated with homeostatic changes in GABAA or acetylcholine muscarinic type 2 (AChM2) receptors (Rs), which evoke inhibition. We found that a significantly greater proportion of the GABAergic neurons were positively stained for c-Fos after SD (∼27%) as compared to sleep control (SC; ∼1%) and sleep recovery (SR; ∼6%), suggesting that they were more active during waking with SD and less active or inactive during sleep with SC and SR. The density of GABAARs and AChM2Rs on the plasma membrane of the GABAergic neurons was significantly increased after SD and restored to control levels after SR. We conclude that the density of these receptors is increased on RFMes GABAergic neurons during presumed enhanced activity with SD and is restored to control levels during presumed lesser or inactivity with SR. Such increases in GABAAR and AChM2R with sleep deficits would be associated with increased susceptibility of the wake-active GABAergic neurons to inhibition from GABAergic and cholinergic sleep-active neurons and to thus permitting the onset of sleep and PS with muscle atonia.
Collapse
|
46
|
Siddiqi HM, Tabasum A, Qasim S, Akhtar MS, Kalsoom S, Ansari FL. Synthesis, Molecular Docking Studies, and Anticonvulsant Evaluation of Novel bis-Phenylhydrazones against Chemically induced Seizures in Mice. J CHIN CHEM SOC-TAIP 2017. [DOI: 10.1002/jccs.201700038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
| | - Aneela Tabasum
- Department of Chemistry; Quaid-i-Azam University; Islamabad 45320 Pakistan
| | - Sumera Qasim
- Faculty of Pharmacy; University of Sargodha; Sargodha Pakistan
| | | | - Saima Kalsoom
- Department of Chemistry; Quaid-i-Azam University; Islamabad 45320 Pakistan
| | | |
Collapse
|
47
|
Jones BE. Principal cell types of sleep-wake regulatory circuits. Curr Opin Neurobiol 2017; 44:101-109. [PMID: 28433001 DOI: 10.1016/j.conb.2017.03.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/13/2017] [Accepted: 03/31/2017] [Indexed: 10/19/2022]
Abstract
Electrophysiological recordings indicate that neurons which discharge maximally in association with distinct sleep-wake states are distributed through the brain, albeit in differing proportions. As studied using juxtacellular recording and labeling within the basal forebrain, four functional principal cell types are distinguished as: wake/paradoxical sleep (W/PS)-, slow wave sleep (SWS)-, W- and PS-max active. They are each comprised by both GABA and glutamate neurons, in addition to acetylcholine neurons belonging to the W/PS group. By their discharge profiles and interactions, the GABA and glutamate neurons of different groups are proposed to have the capacity to generate sleep-wake states with associated EEG and EMG activities, though to also be importantly regulated by neuromodulatory systems, each of which belong to one functional cell group.
Collapse
Affiliation(s)
- Barbara E Jones
- Department of Neurology and Neurosurgery, McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada.
| |
Collapse
|
48
|
Luppi PH, Peyron C, Fort P. Not a single but multiple populations of GABAergic neurons control sleep. Sleep Med Rev 2017; 32:85-94. [DOI: 10.1016/j.smrv.2016.03.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 03/04/2016] [Accepted: 03/04/2016] [Indexed: 12/15/2022]
|
49
|
Hall S, Deurveilher S, Ko KR, Burns J, Semba K. Region-specific increases in FosB/ΔFosB immunoreactivity in the rat brain in response to chronic sleep restriction. Behav Brain Res 2017; 322:9-17. [PMID: 28089853 DOI: 10.1016/j.bbr.2017.01.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/05/2017] [Accepted: 01/11/2017] [Indexed: 01/23/2023]
Abstract
Using a rat model of chronic sleep restriction (CSR) featuring periodic sleep deprivation with slowly rotating wheels (3h on/1h off), we previously observed that 99h of this protocol induced both homeostatic and allostatic (adaptive) changes in physiological and behavioural measures. Notably, the initial changes in sleep intensity and attention performance gradually adapted during CSR despite accumulating sleep loss. To identify brain regions involved in these responses, we used FosB/ΔFosB immunohistochemistry as a marker of chronic neuronal activation. Adult male rats were housed in motorized activity wheels and underwent the 3/1 CSR protocol for 99h, or 99h followed by 6 or 12days of recovery. Control rats were housed in home cages, locked activity wheels, or unlocked activity wheels that the animals could turn freely. Immunohistochemistry was conducted using an antibody that recognized both FosB and ΔFosB, and 24 brain regions involved in sleep/wake, autonomic, and limbic functions were examined. The number of darkly-stained FosB/ΔFosB-immunoreactive cells was increased immediately following 99h of CSR in 8/24 brain regions, including the medial preoptic and perifornical lateral hypothalamic areas, dorsomedial and paraventricular hypothalamic nuclei, and paraventricular thalamic nucleus. FosB/ΔFosB labeling was at control levels in all 8 brain areas following 6 or 12 recovery days, suggesting that most of the immunoreactivity immediately after CSR reflected FosB, the more transient marker of chronic neuronal activation. This region-specific induction of FosB/ΔFosB following CSR may be involved in the mechanisms underlying the allostatic changes in behavioural and physiological responses to CSR.
Collapse
Affiliation(s)
- Shannon Hall
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Samüel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kristin Robin Ko
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Joan Burns
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kazue Semba
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Psychology & Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
50
|
Herrera CG, Ponomarenko A, Korotkova T, Burdakov D, Adamantidis A. Sleep & metabolism: The multitasking ability of lateral hypothalamic inhibitory circuitries. Front Neuroendocrinol 2017; 44:27-34. [PMID: 27884682 DOI: 10.1016/j.yfrne.2016.11.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/18/2016] [Accepted: 11/19/2016] [Indexed: 02/01/2023]
Abstract
The anatomical and functional mapping of lateral hypothalamic circuits has been limited by the numerous cell types and complex, yet unclear, connectivity. Recent advances in functional dissection of input-output neurons in the lateral hypothalamus have identified subset of inhibitory cells as crucial modulators of both sleep-wake states and metabolism. Here, we summarize these recent studies and discuss the multi-tasking functions of hypothalamic circuitries in integrating sleep and metabolism in the mammalian brain.
Collapse
Affiliation(s)
- Carolina Gutierrez Herrera
- Department of Neurology and Department of Clinical Research, Inselspital University Hospital, University of Bern, Bern, Switzerland; Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Alexey Ponomarenko
- Leibniz Institute for Molecular Pharmacology (FMP)/NeuroCure Cluster of Excellence, Berlin, Germany
| | - Tatiana Korotkova
- Leibniz Institute for Molecular Pharmacology (FMP)/NeuroCure Cluster of Excellence, Berlin, Germany
| | - Denis Burdakov
- The Francis Crick Institute, Mill Hill Laboratory, London NW7 1AA, UK; Department of Developmental Neurobiology, King's College London, London WC2R 2LS, UK
| | - Antoine Adamantidis
- Department of Neurology and Department of Clinical Research, Inselspital University Hospital, University of Bern, Bern, Switzerland; Department of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|