1
|
Shi L, He Y, Lian Y, Luo J, Zhu X, Zhao H. Melanin-concentrating hormone: A promising target for antidepressant treatment. Pharmacol Biochem Behav 2025; 250:173999. [PMID: 40081601 DOI: 10.1016/j.pbb.2025.173999] [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: 12/08/2024] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Depression represents a complex neuropsychiatric disorder with an escalating global health burden, characterized by heterogeneous pathophysiology and profound impairments in cognitive-emotional functioning. Current treatment methods have limited efficacy in some individuals and may induce undesirable side effects, necessitating the exploration of novel therapeutic targets and techniques. Emerging research has identified neuropeptide systems as pivotal regulators of mood-related circuits, with melanin-concentrating hormone (MCH) signaling emerging as a particularly promising candidate for antidepressant development. The potential involvement of MCH in the pathophysiology of depression was first proposed over two decades ago. Since then, accumulating evidence from recent studies has progressively illuminated its multifaceted roles in modulating depressive behaviors and underlying neurobiological mechanisms. This review systematically analyzes the mechanistic interplay between MCH signaling and depression pathophenotypes, including its relationship with the hypothalamic-pituitary-adrenal (HPA) axis, neurotransmitter systems, synaptic plasticity, and the regulation of sleep-wakefulness. Particular emphasis is placed on advancing the therapeutic rationale for MCH receptor 1 (MCHR1) antagonists, which demonstrate rapid-onset antidepressant efficacy in preclinical studies compared to traditional agents. Nonetheless, the antidepressant mechanism of the MCH system still requires further elucidation to confirm its therapeutic potential.
Collapse
Affiliation(s)
- Lingchang Shi
- School of Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Ying He
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan 410208, China
| | - Yujun Lian
- School of Nursing, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jie Luo
- College of Acupuncture, Massage and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Xuan Zhu
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan 410208, China; Department of Science & Technology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| | - Hongqing Zhao
- Science & Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, Hunan 410208, China.
| |
Collapse
|
2
|
Wang YQ, Ma WX, Kong LX, Zhang H, Yuan PC, Qu WM, Liu CF, Huang ZL. Ambient chemical and physical approaches for the modulation of sleep and wakefulness. Sleep Med Rev 2025; 79:102015. [PMID: 39447526 DOI: 10.1016/j.smrv.2024.102015] [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: 09/20/2023] [Revised: 09/02/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024]
Abstract
Humans spend a third of their lives asleep. While the sleep-wake behaviors are primarily modulated by homeostasis and circadian rhythm, several ambient chemical and physical factors, including light, sound, odor, vibration, temperature, electromagnetic radiation, and ultrasound, also affect sleep and wakefulness. Light at different wavelengths has different effects on sleep and wakefulness. Sound not only promotes but also suppresses sleep; this effect is mediated by certain nuclei, including the pedunculopontine nucleus and inferior colliculus. Certain sleep-promoting odorants regulate sleep through the involvement of the olfactory bulb and olfactory tubercle. In addition, vibrations may induce sleep through the vestibular system. A modest increase in ambient temperature leads to an increase in sleep duration through the involvement of the preoptic area. Electromagnetic radiation has a dual effect on sleep-wake behaviors. The stimulation produced by the ambient chemical and physical factors activates the peripheral sensory system, which converts the chemical and physical stimuli into nerve impulses. This signal is then transmitted to the central nervous system, including several nuclei associated with the modulation of sleep-wake behaviors. This review summarizes the effects of ambient chemical and physical factors on the regulation of sleep and wakefulness, as well as the underlying neurobiological mechanisms.
Collapse
Affiliation(s)
- Yi-Qun Wang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Wei-Xiang Ma
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ling-Xi Kong
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hui Zhang
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, 241002, China
| | - Ping-Chuan Yuan
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, 241002, China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Joint International Research Laboratory of Sleep and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Bae R, Kim HK, Lu B, Ma J, Xing J, Kim HY. Role of Hypothalamus in Acupuncture's Effects. Brain Sci 2025; 15:72. [PMID: 39851439 PMCID: PMC11763592 DOI: 10.3390/brainsci15010072] [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: 12/15/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
The significant correlation between ancient medicinal practices and brain function marks a revolutionary frontier in the field of neuroscience. Acupuncture, a traditional oriental medicine, can affect brain regions, such as the hypothalamus, anterior cingulate, posterior cingulate, and hippocampus, and produces specific therapeutic effects, such as pain relief, suppression of hypertension, and alleviation of drug addiction. Among the brain regions, the hypothalamus, a small yet critical region in the brain, plays a pivotal role in maintaining homeostasis by regulating a wide array of physiological processes, including stress responses, energy balance, and pain modulation. Emerging evidence suggests that acupuncture may exert its therapeutic effects by modulating the activity of the hypothalamus and its associated neural circuits, particularly in relation to pain, stress, and metabolic regulation. Thus, we conducted a comprehensive review of past and current research on the role of the hypothalamus in mediating the therapeutic effects of acupuncture.
Collapse
Affiliation(s)
- Ryan Bae
- Morrissey College of Arts and Sciences, Boston College, Boston, MA 02467, USA
| | - Hyung Kyu Kim
- Department of Physiology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Baoji Lu
- Department of Physiology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Jing Ma
- Department of Physiology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Juping Xing
- Department of Physiology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Hee Young Kim
- Department of Physiology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
4
|
Naganuma F, Girgin B, Agu ABS, Hirano K, Nakamura T, Yanai K, Vetrivelan R, Mochizuki T, Yanagisawa M, Yoshikawa T. Pharmacological inhibition of histamine N-methyltransferase extends wakefulness and suppresses cataplexy in a mouse model of narcolepsy. Sleep 2025; 48:zsae244. [PMID: 39441998 DOI: 10.1093/sleep/zsae244] [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: 06/22/2024] [Revised: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
Histamine, a neurotransmitter, plays a predominant role in maintaining wakefulness. Furthermore, our previous studies showed that histamine N-methyltransferase (HNMT), a histamine-metabolizing enzyme, is important for regulating brain histamine concentration. However, the effects of pharmacological HNMT inhibition on mouse behavior, including the sleep-wake cycle and cataplexy, in a mouse model of narcolepsy have not yet been investigated. In the present study, we investigated the effects of metoprine, an HNMT inhibitor with high blood-brain barrier permeability, in wild-type (WT) and orexin-deficient (OxKO) narcoleptic mice. Metoprine increased brain histamine concentration in a time- and dose-dependent manner without affecting peripheral histamine concentrations. Behavioral tests showed that metoprine increased locomotor activity in both novel and familiar environments, but did not alter anxiety-like behavior. Sleep analysis showed that metoprine increased wakefulness and decreased non-rapid eye movement (NREM) sleep through the activation of the histamine H1 receptor (H1R) in WT mice. In contrast, the reduction of rapid eye movement (REM) sleep by metoprine occurred independent of H1R. In OxKO mice, metoprine was found to prolong wakefulness and robustly suppress cataplexy. In addition, metoprine has a greater therapeutic effect on cataplexy than pitolisant, which induces histamine release in the brain and has been approved for patients with narcolepsy. These data demonstrate that HNMT inhibition has a strong effect on wakefulness, demonstrating therapeutic potential against cataplexy in narcolepsy.
Collapse
Affiliation(s)
- Fumito Naganuma
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Miyagi, Japan
| | - Birkan Girgin
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Anne Bernadette S Agu
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Kyosuke Hirano
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Tadaho Nakamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Miyagi, Japan
- Division of Bioregulatory Pharmacology, Department of Pharmacology, Iwate Medical University, Iwate, Japan
| | - Kazuhiko Yanai
- Cyclotron and Radioisotope Center, Tohoku University, Miyagi, Japan
| | - Ramalingam Vetrivelan
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Takatoshi Mochizuki
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| | - Takeo Yoshikawa
- Department of Neuropharmacology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| |
Collapse
|
5
|
Kashiwagi M, Beck G, Kanuka M, Arai Y, Tanaka K, Tatsuzawa C, Koga Y, Saito YC, Takagi M, Oishi Y, Sakaguchi M, Baba K, Ikuno M, Yamakado H, Takahashi R, Yanagisawa M, Murayama S, Sakurai T, Sakai K, Nakagawa Y, Watanabe M, Mochizuki H, Hayashi Y. A pontine-medullary loop crucial for REM sleep and its deficit in Parkinson's disease. Cell 2024; 187:6272-6289.e21. [PMID: 39303715 DOI: 10.1016/j.cell.2024.08.046] [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: 03/01/2023] [Revised: 03/22/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024]
Abstract
Identifying the properties of the rapid eye movement (REM) sleep circuitry and its relation to diseases has been challenging due to the neuronal heterogeneity of the brainstem. Here, we show in mice that neurons in the pontine sublaterodorsal tegmentum (SubLDT) that express corticotropin-releasing hormone-binding protein (Crhbp+ neurons) and project to the medulla promote REM sleep. Within the medullary area receiving projections from Crhbp+ neurons, neurons expressing nitric oxide synthase 1 (Nos1+ neurons) project to the SubLDT and promote REM sleep, suggesting a positively interacting loop between the pons and the medulla operating as a core REM sleep circuit. Nos1+ neurons also project to areas that control wide forebrain activity. Ablating Crhbp+ neurons reduces sleep and impairs REM sleep atonia. In Parkinson's disease patients with REM sleep behavior disorders, CRHBP-immunoreactive neurons are largely reduced and contain pathologic α-synuclein, providing insight into the mechanisms underlying the sleep deficits characterizing this disease.
Collapse
Affiliation(s)
- Mitsuaki Kashiwagi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Goichi Beck
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mika Kanuka
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoshifumi Arai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kaeko Tanaka
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Chika Tatsuzawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yumiko Koga
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki C Saito
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Marina Takagi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yo Oishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Masanori Sakaguchi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kousuke Baba
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masashi Ikuno
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 605-8507, Japan
| | - Hodaka Yamakado
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 605-8507, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 605-8507, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Japan Life Science Center for Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shigeo Murayama
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Suita, Osaka 565-0871, Japan; Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Institute for Geriatrics and Gerontology, Itabashi-Ku, Tokyo 173-0015, Japan
| | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kazuya Sakai
- Integrative Physiology of the Brain Arousal System, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR5292, School of Medicine, Claude Bernard University Lyon 1, 69373 Lyon, France
| | - Yoshimi Nakagawa
- Division of Complex Biosystem Research Institute of Natural Medicine, University of Toyama, Toyama, Toyama 930-0194, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yu Hayashi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
6
|
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
|
7
|
Zhang XF, Li YD, Li Y, Li Y, Xu D, Bi LL, Xu HB. Ventral subiculum promotes wakefulness through several pathways in male mice. Neuropsychopharmacology 2024; 49:1468-1480. [PMID: 38734818 PMCID: PMC11251017 DOI: 10.1038/s41386-024-01875-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024]
Abstract
The ventral subiculum (vSUB), the major output structure of the hippocampal formation, regulates motivation, stress integration, and anxiety-like behaviors that rely on heightened arousal. However, the roles and underlying neural circuits of the vSUB in wakefulness are poorly known. Using in vivo fiber photometry and multichannel electrophysiological recordings in mice, we found that the vSUB glutamatergic neurons exhibited high activities during wakefulness. Moreover, activation of vSUB glutamatergic neurons caused an increase in wakefulness and anxiety-like behaviors and induced a rapid transition from sleep to wakefulness. In addition, optogenetic stimulation of vSUB glutamatergic terminals and retrograde-targeted chemogenetic activation of vSUB glutamatergic neurons revealed that vSUB promoted arousal by innervating the lateral hypothalamus (LH), nucleus accumbens (NAc) shell, and prefrontal cortex (PFC). Nevertheless, local microinjection of dopamine D1 or D2/D3 receptor antagonist blocked the wake-promoting effect induced by chemogenetic activation of vSUB pathways. Finally, chemogenetic inhibition of vSUB glutamatergic neurons decreased arousal. Altogether, our findings reveal a prominent contribution of vSUB glutamatergic neurons to the control of wakefulness through several pathways.
Collapse
Affiliation(s)
- Xue-Fen Zhang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yi-Dan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yue Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Ying Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Dan Xu
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Lin-Lin Bi
- Department of Pathology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China.
- Center for Pathology and Molecular Diagnostics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
| | - Hai-Bo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
8
|
Satpati A, Pereira FL, Soloviev AV, Mladinov M, Larsen E, Hua SL, Tu CL, Leite REP, Suemoto CK, Rodriguez RD, Paes VR, Walsh C, Spina S, Seeley WW, Pasqualucci CA, Filho WJ, Chang W, Neylan TC, Grinberg LT. The wake- and sleep-modulating neurons of the lateral hypothalamic area demonstrate a differential pattern of degeneration in Alzheimers disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.06.583765. [PMID: 38559184 PMCID: PMC10979907 DOI: 10.1101/2024.03.06.583765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
BACKGROUND Sleep-wake dysfunction is an early and common event in Alzheimer's disease (AD). The lateral hypothalamic area (LHA) regulates the sleep and wake cycle through wake-promoting orexinergic neurons (OrxN) and sleep-promoting melanin-concentrating hormone or MCHergic neurons (MCHN). These neurons share close anatomical proximity with functional reciprocity. This study investigated LHA OrxN and MCHN loss patterns in AD individuals. Understanding the degeneration pattern of these neurons will be instrumental in designing potential therapeutics to slow down the disease progression and remediate the sleep-wake dysfunction in AD. METHODS Postmortem human brain tissue from donors with AD (across progressive stages) and controls were examined using unbiased stereology. Formalin-fixed, celloidin-embedded hypothalamic sections were stained with Orx-A/MCH, p-tau (CP13), and counterstained with gallocyanin. Orx or MCH-positive neurons with or without CP13 inclusions and gallocyanin-stained neurons were considered for stereology counting. Additionally, we extracted RNA from the LHA using conventional techniques. We used customized Neuropathology and Glia nCounter (Nanostring) panels to study gene expression. Wald statistical test was used to compare the groups, and the genes were considered differentially expressed when the p-value was <.05. RESULTS We observed a progressive decline in OrxN alongside a relative preservation of MCHN. OrxN decreased by 58% (p=0.03) by Braak stages (BB) 1-2 and further declined to 81% (p=0.03) by BB 5-6. Conversely, MCHN demonstrated a non-statistical significant decline (27%, p=0.1088) by BB 6. We observed a progressive increase in differentially expressed genes (DEGs), starting with glial profile changes in BB2. While OrxN loss was observed, Orx-related genes showed upregulation in BB 3-4 compared to BB 0-1. GO and KEGG terms related to neuroinflammatory pathways were mainly enriched. CONCLUSIONS To date, OrxN loss in the LHA represents the first neuronal population to die preceding the loss of LC neurons. Conversely, MCHN shows resilience to AD p-tau accumulation across Braak stages. The initial loss of OrxN correlates with specific neuroinflammation, glial profile changes, and an overexpression of HCRT, possibly due to hyperexcitation following compensation mechanisms. Interventions preventing OrxN loss and inhibiting p-tau accumulation in the LHA could prevent neuronal loss in AD and, perhaps, the progression of the disease.
Collapse
|
9
|
Vetrivelan R, Bandaru SS. Neural Control of REM Sleep and Motor Atonia: Current Perspectives. Curr Neurol Neurosci Rep 2023; 23:907-923. [PMID: 38060134 PMCID: PMC11891935 DOI: 10.1007/s11910-023-01322-x] [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] [Accepted: 11/02/2023] [Indexed: 12/08/2023]
Abstract
PURPOSE OF REVIEW Since the formal discovery of rapid eye movement (REM) sleep in 1953, we have gained a vast amount of knowledge regarding the specific populations of neurons, their connections, and synaptic mechanisms regulating this stage of sleep and its accompanying features. This article discusses REM sleep circuits and their dysfunction, specifically emphasizing recent studies using conditional genetic tools. RECENT FINDINGS Sublaterodorsal nucleus (SLD) in the dorsolateral pons, especially the glutamatergic subpopulation in this region (SLDGlut), are shown to be indispensable for REM sleep. These neurons appear to be single REM generators in the rodent brain and may initiate and orchestrate all REM sleep events, including cortical and hippocampal activation and muscle atonia through distinct pathways. However, several cell groups in the brainstem and hypothalamus may influence SLDGlut neuron activity, thereby modulating REM sleep timing, amounts, and architecture. Damage to SLDGlut neurons or their projections involved in muscle atonia leads to REM behavior disorder, whereas the abnormal activation of this pathway during wakefulness may underlie cataplexy in narcolepsy. Despite some opposing views, it has become evident that SLDGlut neurons are the sole generators of REM sleep and its associated characteristics. Further research should prioritize a deeper understanding of their cellular, synaptic, and molecular properties, as well as the mechanisms that trigger their activation during cataplexy and make them susceptible in RBD.
Collapse
Affiliation(s)
- Ramalingam Vetrivelan
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA.
| | - Sathyajit Sai Bandaru
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| |
Collapse
|
10
|
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
|
11
|
Bouâouda H, Jha PK. Orexin and MCH neurons: regulators of sleep and metabolism. Front Neurosci 2023; 17:1230428. [PMID: 37674517 PMCID: PMC10478345 DOI: 10.3389/fnins.2023.1230428] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023] Open
Abstract
Sleep-wake and fasting-feeding are tightly coupled behavioral states that require coordination between several brain regions. The mammalian lateral hypothalamus (LH) is a functionally and anatomically complex brain region harboring heterogeneous cell populations that regulate sleep, feeding, and energy metabolism. Significant attempts were made to understand the cellular and circuit bases of LH actions. Rapid advancements in genetic and electrophysiological manipulation help to understand the role of discrete LH cell populations. The opposing action of LH orexin/hypocretin and melanin-concentrating hormone (MCH) neurons on metabolic sensing and sleep-wake regulation make them the candidate to explore in detail. This review surveys the molecular, genetic, and neuronal components of orexin and MCH signaling in the regulation of sleep and metabolism.
Collapse
Affiliation(s)
- Hanan Bouâouda
- Pharmacology Institute, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Pawan Kumar Jha
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
12
|
Fang LZ, Linehan V, Licursi M, Alberto CO, Power JL, Parsons MP, Hirasawa M. Prostaglandin E 2 activates melanin-concentrating hormone neurons to drive diet-induced obesity. Proc Natl Acad Sci U S A 2023; 120:e2302809120. [PMID: 37467285 PMCID: PMC10401019 DOI: 10.1073/pnas.2302809120] [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: 03/07/2023] [Accepted: 05/09/2023] [Indexed: 07/21/2023] Open
Abstract
Hypothalamic inflammation reduces appetite and body weight during inflammatory diseases, while promoting weight gain when induced by high-fat diet (HFD). How hypothalamic inflammation can induce opposite energy balance outcomes remains unclear. We found that prostaglandin E2 (PGE2), a key hypothalamic inflammatory mediator of sickness, also mediates diet-induced obesity (DIO) by activating appetite-promoting melanin-concentrating hormone (MCH) neurons in the hypothalamus in rats and mice. The effect of PGE2 on MCH neurons is excitatory at low concentrations while inhibitory at high concentrations, indicating that these neurons can bidirectionally respond to varying levels of inflammation. During prolonged HFD, endogenous PGE2 depolarizes MCH neurons through an EP2 receptor-mediated inhibition of the electrogenic Na+/K+-ATPase. Disrupting this mechanism by genetic deletion of EP2 receptors on MCH neurons is protective against DIO and liver steatosis in male and female mice. Thus, an inflammatory mediator can directly stimulate appetite-promoting neurons to exacerbate DIO and fatty liver.
Collapse
Affiliation(s)
- Lisa Z. Fang
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’sA1B 3V6, Canada
| | - Victoria Linehan
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’sA1B 3V6, Canada
| | - Maria Licursi
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’sA1B 3V6, Canada
| | - Christian O. Alberto
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’sA1B 3V6, Canada
| | - Jacob L. Power
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’sA1B 3V6, Canada
| | - Matthew P. Parsons
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’sA1B 3V6, Canada
| | - Michiru Hirasawa
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John’sA1B 3V6, Canada
| |
Collapse
|
13
|
Prokofeva K, Saito YC, Niwa Y, Mizuno S, Takahashi S, Hirano A, Sakurai T. Structure and Function of Neuronal Circuits Linking Ventrolateral Preoptic Nucleus and Lateral Hypothalamic Area. J Neurosci 2023; 43:4075-4092. [PMID: 37117013 PMCID: PMC10255079 DOI: 10.1523/jneurosci.1913-22.2023] [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: 10/04/2022] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 04/30/2023] Open
Abstract
To understand how sleep-wakefulness cycles are regulated, it is essential to disentangle structural and functional relationships between the preoptic area (POA) and lateral hypothalamic area (LHA), since these regions play important yet opposing roles in the sleep-wakefulness regulation. GABA- and galanin (GAL)-producing neurons in the ventrolateral preoptic nucleus (VLPO) of the POA (VLPOGABA and VLPOGAL neurons) are responsible for the maintenance of sleep, while the LHA contains orexin-producing neurons (orexin neurons) that are crucial for maintenance of wakefulness. Through the use of rabies virus-mediated neural tracing combined with in situ hybridization (ISH) in male and female orexin-iCre mice, we revealed that the vesicular GABA transporter (Vgat, Slc32a1)- and galanin (Gal)-expressing neurons in the VLPO directly synapse with orexin neurons in the LHA. A majority (56.3 ± 8.1%) of all VLPO input neurons connecting to orexin neurons were double-positive for Vgat and Gal Using projection-specific rabies virus-mediated tracing in male and female Vgat-ires-Cre and Gal-Cre mice, we discovered that VLPOGABA and VLPOGAL neurons that send projections to the LHA received innervations from similarly distributed input neurons in many brain regions, with the POA and LHA being among the main upstream areas. Additionally, we found that acute optogenetic excitation of axons of VLPOGABA neurons, but not VLPOGAL neurons, in the LHA of male Vgat-ires-Cre mice induced wakefulness. This study deciphers the connectivity between the VLPO and LHA, provides a large-scale map of upstream neuronal populations of VLPO→LHA neurons, and reveals a previously uncovered function of the VLPOGABA→LHA pathway in the regulation of sleep and wakefulness.SIGNIFICANCE STATEMENT We identified neurons in the ventrolateral preoptic nucleus (VLPO) that are positive for vesicular GABA transporter (Vgat) and/or galanin (Gal) and serve as presynaptic partners of orexin-producing neurons in the lateral hypothalamic area (LHA). We depicted monosynaptic input neurons of GABA- and galanin-producing neurons in the VLPO that send projections to the LHA throughout the entire brain. Their input neurons largely overlap, suggesting that they comprise a common neuronal population. However, acute excitatory optogenetic manipulation of the VLPOGABA→LHA pathway, but not the VLPOGAL→LHA pathway, evoked wakefulness. This study shows the connectivity of major components of the sleep/wake circuitry in the hypothalamus and unveils a previously unrecognized function of the VLPOGABA→LHA pathway in sleep-wakefulness regulation. Furthermore, we suggest the existence of subpopulations of VLPOGABA neurons that innervate LHA.
Collapse
Affiliation(s)
- Kseniia Prokofeva
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki C Saito
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yasutaka Niwa
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Seiya Mizuno
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Satoru Takahashi
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Arisa Hirano
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Life Science Center for Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
14
|
Calafate S, Özturan G, Thrupp N, Vanderlinden J, Santa-Marinha L, Morais-Ribeiro R, Ruggiero A, Bozic I, Rusterholz T, Lorente-Echeverría B, Dias M, Chen WT, Fiers M, Lu A, Vlaeminck I, Creemers E, Craessaerts K, Vandenbempt J, van Boekholdt L, Poovathingal S, Davie K, Thal DR, Wierda K, Oliveira TG, Slutsky I, Adamantidis A, De Strooper B, de Wit J. Early alterations in the MCH system link aberrant neuronal activity and sleep disturbances in a mouse model of Alzheimer's disease. Nat Neurosci 2023:10.1038/s41593-023-01325-4. [PMID: 37188873 DOI: 10.1038/s41593-023-01325-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 04/10/2023] [Indexed: 05/17/2023]
Abstract
Early Alzheimer's disease (AD) is associated with hippocampal hyperactivity and decreased sleep quality. Here we show that homeostatic mechanisms transiently counteract the increased excitatory drive to CA1 neurons in AppNL-G-F mice, but that this mechanism fails in older mice. Spatial transcriptomics analysis identifies Pmch as part of the adaptive response in AppNL-G-F mice. Pmch encodes melanin-concentrating hormone (MCH), which is produced in sleep-active lateral hypothalamic neurons that project to CA1 and modulate memory. We show that MCH downregulates synaptic transmission, modulates firing rate homeostasis in hippocampal neurons and reverses the increased excitatory drive to CA1 neurons in AppNL-G-F mice. AppNL-G-F mice spend less time in rapid eye movement (REM) sleep. AppNL-G-F mice and individuals with AD show progressive changes in morphology of CA1-projecting MCH axons. Our findings identify the MCH system as vulnerable in early AD and suggest that impaired MCH-system function contributes to aberrant excitatory drive and sleep defects, which can compromise hippocampus-dependent functions.
Collapse
Affiliation(s)
- Sara Calafate
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium.
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Gökhan Özturan
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Nicola Thrupp
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Jeroen Vanderlinden
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Luísa Santa-Marinha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rafaela Morais-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Antonella Ruggiero
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ivan Bozic
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| | - Thomas Rusterholz
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Blanca Lorente-Echeverría
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Marcelo Dias
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Wei-Ting Chen
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Mark Fiers
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Ashley Lu
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Ine Vlaeminck
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Eline Creemers
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Katleen Craessaerts
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Joris Vandenbempt
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Luuk van Boekholdt
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
- KU Leuven, Department of Otorhinolaryngology, Leuven, Belgium
| | - Suresh Poovathingal
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Kristofer Davie
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Dietmar Rudolf Thal
- Department of Imaging and Pathology, Laboratory of Neuropathology, and Leuven Brain Institute, KU-Leuven, O&N IV, Leuven, Belgium
- Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Keimpe Wierda
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Antoine Adamantidis
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Bart De Strooper
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium.
- UK Dementia Research Institute (UK DRI@UCL) at University College London, London, UK.
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium.
| |
Collapse
|
15
|
Ruiz-Viroga V, de Ceglia M, Morelli L, Castaño EM, Calvo EB, Suárez J, Rodríguez de Fonseca F, Galeano P, Lagos P. Acute intrahippocampal administration of melanin-concentrating hormone impairs memory consolidation and decreases the expression of MCHR-1 and TrkB receptors. Prog Neuropsychopharmacol Biol Psychiatry 2023; 123:110703. [PMID: 36565982 DOI: 10.1016/j.pnpbp.2022.110703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 11/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Interest in the role of melanin-concentrating hormone (MCH) in memory processes has increased in recent years, with some studies reporting memory-enhancing effects, while others report deleterious effects. Due to these discrepancies, this study seeks to provide new evidence about the role of MCH in memory consolidation and its relation with BDNF/TrkB system. To this end, in the first experiment, increased doses of MCH were acutely administered in both hippocampi to groups of male rats (25, 50, 200, and 500 ng). Microinjections were carried out immediately after finishing the sample trial of two hippocampal-dependent behavioral tasks: the Novel Object Recognition Test (NORT) and the modified Elevated Plus Maze (mEPM) test. Results indicated that a dose of 200 ng of MCH or higher impaired memory consolidation in both tasks. A second experiment was performed in which a dose of 200 ng of MCH was administered alone or co-administered with the MCHR-1 antagonist ATC-0175 at the end of the sample trial in the NORT. Results showed that MCH impaired memory consolidation, while the co-administration with ATC-0175 reverted this detrimental effect. Moreover, MCH induced a significant decrease in hippocampal MCHR-1 and TrkB expression with no modification in the expression of BDNF and NMDA receptor subunits NR1, NR2A, and NR2B. These results suggest that MCH in vivo elicits pro-amnesic effects in the rat hippocampus by decreasing the availability of its receptor and TrkB receptors, thus linking both endogenous systems to memory processes.
Collapse
Affiliation(s)
- Vicente Ruiz-Viroga
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, Montevideo ZP11800, Uruguay
| | - Marialuisa de Ceglia
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Av. Carlos Haya 82, Málaga 29010, Spain.
| | - Laura Morelli
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina.
| | - Eduardo M Castaño
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina.
| | - Eduardo Blanco Calvo
- Instituto de Investigación Biomédica de Málaga (IBIMA), Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Campus de Teatinos S/N, Málaga 29071, Spain.
| | - Juan Suárez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Universidad de Málaga, Málaga 29071, Spain.
| | - Fernando Rodríguez de Fonseca
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Av. Carlos Haya 82, Málaga 29010, Spain.
| | - Pablo Galeano
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina.
| | - Patricia Lagos
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, Montevideo ZP11800, Uruguay.
| |
Collapse
|
16
|
Kroeger D, Thundercliffe J, Phung A, De Luca R, Geraci C, Bragg S, McCafferty KJ, Bandaru SS, Arrigoni E, Scammell TE. Glutamatergic pedunculopontine tegmental neurons control wakefulness and locomotion via distinct axonal projections. Sleep 2022; 45:zsac242. [PMID: 36170177 PMCID: PMC9742893 DOI: 10.1093/sleep/zsac242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/02/2022] [Indexed: 12/15/2022] Open
Abstract
STUDY OBJECTIVES The pedunculopontine tegmental (PPT) nucleus is implicated in many brain functions, ranging from sleep/wake control and locomotion, to reward mechanisms and learning. The PPT contains cholinergic, GABAergic, and glutamatergic neurons with extensive ascending and descending axonal projections. Glutamatergic PPT (PPTvGlut2) neurons are thought to promote wakefulness, but the mechanisms through which this occurs are unknown. In addition, some researchers propose that PPTvGlut2 neurons promote locomotion, yet even though the PPT is a target for deep brain stimulation in Parkinson's disease, the role of the PPT in locomotion is debated. We hypothesized that PPTvGluT2 neurons drive arousal and specific waking behaviors via certain projections and modulate locomotion via others. METHODS We mapped the axonal projections of PPTvGlut2 neurons using conditional anterograde tracing and then photostimulated PPTvGlut2 soma or their axon terminal fields across sleep/wake states and analyzed sleep/wake behavior, muscle activity, and locomotion in transgenic mice. RESULTS We found that stimulation of PPTvGlut2 soma and their axon terminals rapidly triggered arousals from non-rapid eye movement sleep, especially with activation of terminals in the basal forebrain (BF) and lateral hypothalamus (LH). With photoactivation of PPTvGlut2 terminals in the BF and LH, this wakefulness was accompanied by locomotion and other active behaviors, but stimulation of PPTvGlut2 soma and terminals in the substantia nigra triggered only quiet wakefulness without locomotion. CONCLUSIONS These findings demonstrate the importance of the PPTvGluT2 neurons in driving various aspects of arousal and show that heterogeneous brain nuclei, such as the PPT, can promote a variety of behaviors via distinct axonal projections.
Collapse
Affiliation(s)
- Daniel Kroeger
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Department of Anatomy, Physiology, and Pharmacology, Auburn University, Auburn, AL, USA
| | - Jack Thundercliffe
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Alex Phung
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Roberto De Luca
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Carolyn Geraci
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Samuel Bragg
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Kayleen J McCafferty
- Department of Anatomy, Physiology, and Pharmacology, Auburn University, Auburn, AL, USA
| | - Sathyajit S Bandaru
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Matei M, Bergel A, Pezet S, Tanter M. Global dissociation of the posterior amygdala from the rest of the brain during REM sleep. Commun Biol 2022; 5:1306. [PMID: 36443640 PMCID: PMC9705305 DOI: 10.1038/s42003-022-04257-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 11/14/2022] [Indexed: 11/29/2022] Open
Abstract
Rapid-eye-movement sleep (REMS) or paradoxical sleep is associated with intense neuronal activity, fluctuations in autonomic control, body paralysis and brain-wide hyperemia. The mechanisms and functions of these energy-demanding patterns remain elusive and a global picture of brain activation during REMS is currently missing. In the present work, we performed functional ultrasound imaging on rats over multiple coronal and sagittal brain sections during hundreds of spontaneous REMS episodes to provide the spatiotemporal dynamics of vascular activity in 259 brain regions spanning more than 2/3 of the total brain volume. We first demonstrate a dissociation between basal/midbrain and cortical structures, the first ones sustaining tonic activation during REMS while the others are activated in phasic bouts. Second, we isolated the vascular compartment in our recordings and identified arteries in the anterior part of the brain as strongly involved in the blood supply during REMS episodes. Finally, we report a peculiar activation pattern in the posterior amygdala, which is strikingly disconnected from the rest of the brain during most REMS episodes. This last finding suggests that the amygdala undergoes specific processing during REMS and may be linked to the regulation of emotions and the creation of dream content during this very state.
Collapse
Affiliation(s)
- Marta Matei
- grid.15736.360000 0001 1882 0021Physics for Medicine Paris, Inserm U1273, ESPCI Paris, CNRS UMR 8063, Paris Sciences et Lettres research University, Paris, France
| | - Antoine Bergel
- grid.15736.360000 0001 1882 0021Physics for Medicine Paris, Inserm U1273, ESPCI Paris, CNRS UMR 8063, Paris Sciences et Lettres research University, Paris, France
| | - Sophie Pezet
- grid.15736.360000 0001 1882 0021Physics for Medicine Paris, Inserm U1273, ESPCI Paris, CNRS UMR 8063, Paris Sciences et Lettres research University, Paris, France
| | - Mickaël Tanter
- grid.15736.360000 0001 1882 0021Physics for Medicine Paris, Inserm U1273, ESPCI Paris, CNRS UMR 8063, Paris Sciences et Lettres research University, Paris, France
| |
Collapse
|
18
|
Potter LE, Burgess CR. The melanin-concentrating hormone system as a target for the treatment of sleep disorders. Front Neurosci 2022; 16:952275. [PMID: 36177357 PMCID: PMC9513178 DOI: 10.3389/fnins.2022.952275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Given the widespread prevalence of sleep disorders and their impacts on health, it is critical that researchers continue to identify and evaluate novel avenues of treatment. Recently the melanin-concentrating hormone (MCH) system has attracted commercial and scientific interest as a potential target of pharmacotherapy for sleep disorders. This interest emerges from basic scientific research demonstrating a role for MCH in regulating sleep, and particularly REM sleep. In addition to this role in sleep regulation, the MCH system and the MCH receptor 1 (MCHR1) have been implicated in a wide variety of other physiological functions and behaviors, including feeding/metabolism, reward, anxiety, depression, and learning. The basic research literature on sleep and the MCH system, and the history of MCH drug development, provide cause for both skepticism and cautious optimism about the prospects of MCH-targeting drugs in sleep disorders. Extensive efforts have focused on developing MCHR1 antagonists for use in obesity, however, few of these drugs have advanced to clinical trials, and none have gained regulatory approval. Additional basic research will be needed to fully characterize the MCH system’s role in sleep regulation, for example, to fully differentiate between MCH-neuron and peptide/receptor-mediated functions. Additionally, a number of issues relating to drug design will continue to pose a practical challenge for novel pharmacotherapies targeting the MCH system.
Collapse
Affiliation(s)
- Liam E. Potter
- Department of Molecular and Integrative Physiology, Michigan Medicine, Ann Arbor, MI, United States
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Liam E. Potter,
| | - Christian R. Burgess
- Department of Molecular and Integrative Physiology, Michigan Medicine, Ann Arbor, MI, United States
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
- Christian R. Burgess,
| |
Collapse
|
19
|
Rodrigues LTC, Patrone LGA, Gargaglioni LH, Dias MB. Melanin-concentrating hormone regulates the hypercapnic chemoreflex by acting in the lateral hypothalamic area. Exp Physiol 2022; 107:1298-1311. [PMID: 35930596 DOI: 10.1113/ep090318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 07/22/2022] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? MCH suppresses the hypercapnic chemoreflex but the mechanism by which this effect is produced has not been previously explored. What is the main finding and its importance? MCH acting in the lateral hypothalamic area but not in the locus coeruleus in rats, in the light period, attenuates the hypercapnic chemoreflex. Our data provide new insight regarding the role of MCH in the modulation of the hypercapnic ventilatory response. ABSTRACT Melanin-concentrating hormone (MCH) is a hypothalamic neuropeptide involved in a broad range of homeostatic functions including regulation of the hypercapnic chemoreflex. We evaluated whether MCH modulates the hypercapnic ventilatory response by acting in the lateral hypothalamic area (LHA) and/or in the locus coeruleus (LC). Here, we measured pulmonary ventilation (VE ), body temperature, electroencephalogram (EEG) and electromyogram (EMG) of unanesthetized adult male Wistar rats before and after microinjection of MCH [0.4 mM] or MCH1-R antagonist (SNAP-94847 [63 mM]) into the LHA and LC, in room air and 7% CO2 conditions during wakefulness and sleep, in the dark and light periods. MCH intra-LHA caused a decreased CO2 ventilatory response during wakefulness and sleep in the light period, while SNAP-94847 intra-LHA increased this response, during wakefulness in the light period. In the LC, MCH or the MCH1-R antagonist caused no change in the hypercapnic ventilatory response. Our results suggest that MCH, in the LHA, exerts an inhibitory modulation of the hypercapnic ventilatory response during the light-inactive period in rats. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Laísa T C Rodrigues
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University-UNESP, Botucatu, SP, Brazil
| | - Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, Sao Paulo State University-FCAV, Jaboticabal, SP, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University-FCAV, Jaboticabal, SP, Brazil
| | - Mirela B Dias
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University-UNESP, Botucatu, SP, Brazil
| |
Collapse
|
20
|
Translational Approaches to Influence Sleep and Arousal. Brain Res Bull 2022; 185:140-161. [PMID: 35550156 PMCID: PMC9554922 DOI: 10.1016/j.brainresbull.2022.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 12/16/2022]
Abstract
Sleep disorders are widespread in society and are prevalent in military personnel and in Veterans. Disturbances of sleep and arousal mechanisms are common in neuropsychiatric disorders such as schizophrenia, post-traumatic stress disorder, anxiety and affective disorders, traumatic brain injury, dementia, and substance use disorders. Sleep disturbances exacerbate suicidal ideation, a major concern for Veterans and in the general population. These disturbances impair quality of life, affect interpersonal relationships, reduce work productivity, exacerbate clinical features of other disorders, and impair recovery. Thus, approaches to improve sleep and modulate arousal are needed. Basic science research on the brain circuitry controlling sleep and arousal led to the recent approval of new drugs targeting the orexin/hypocretin and histamine systems, complementing existing drugs which affect GABAA receptors and monoaminergic systems. Non-invasive brain stimulation techniques to modulate sleep and arousal are safe and show potential but require further development to be widely applicable. Invasive viral vector and deep brain stimulation approaches are also in their infancy but may be used to modulate sleep and arousal in severe neurological and psychiatric conditions. Behavioral, pharmacological, non-invasive brain stimulation and cell-specific invasive approaches covered here suggest the potential to selectively influence arousal, sleep initiation, sleep maintenance or sleep-stage specific phenomena such as sleep spindles or slow wave activity. These manipulations can positively impact the treatment of a wide range of neurological and psychiatric disorders by promoting the restorative effects of sleep on memory consolidation, clearance of toxic metabolites, metabolism, and immune function and by decreasing hyperarousal.
Collapse
|
21
|
The Sleep-Promoting Ventrolateral Preoptic Nucleus: What Have We Learned over the Past 25 Years? Int J Mol Sci 2022; 23:ijms23062905. [PMID: 35328326 PMCID: PMC8954377 DOI: 10.3390/ijms23062905] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 01/09/2023] Open
Abstract
For over a century, the role of the preoptic hypothalamus and adjacent basal forebrain in sleep-wake regulation has been recognized. However, for years, the identity and location of sleep- and wake-promoting neurons in this region remained largely unresolved. Twenty-five years ago, Saper and colleagues uncovered a small collection of sleep-active neurons in the ventrolateral preoptic nucleus (VLPO) of the preoptic hypothalamus, and since this seminal discovery the VLPO has been intensively investigated by labs around the world, including our own. Herein, we first review the history of the preoptic area, with an emphasis on the VLPO in sleep-wake control. We then attempt to synthesize our current understanding of the circuit, cellular and synaptic bases by which the VLPO both regulates and is itself regulated, in order to exert a powerful control over behavioral state, as well as examining data suggesting an involvement of the VLPO in other physiological processes.
Collapse
|
22
|
Characterization of Hypothalamic MCH Neuron Development in a 3D Differentiation System of Mouse Embryonic Stem Cells. eNeuro 2022; 9:ENEURO.0442-21.2022. [PMID: 35437265 PMCID: PMC9047030 DOI: 10.1523/eneuro.0442-21.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 01/20/2023] Open
Abstract
Hypothalamic melanin-concentrating hormone (MCH) neurons are important regulators of multiple physiological processes, such as sleep, feeding, and memory. Despite the increasing interest in their neuronal functions, the molecular mechanism underlying MCH neuron development remains poorly understood. We report that a three-dimensional culture of mouse embryonic stem cells (mESCs) can generate hypothalamic-like tissues containing MCH-positive neurons, which reproduce morphologic maturation, neuronal connectivity, and neuropeptide/neurotransmitter phenotype of native MCH neurons. Using this in vitro system, we demonstrate that Hedgehog (Hh) signaling serves to produce major neurochemical subtypes of MCH neurons characterized by the presence or absence of cocaine- and amphetamine-regulated transcript (CART). Without exogenous Hh signals, mESCs initially differentiated into dorsal hypothalamic/prethalamic progenitors and finally into MCH+CART+ neurons through a specific intermediate progenitor state. Conversely, activation of the Hh pathway specified ventral hypothalamic progenitors that generate both MCH+CART− and MCH+CART+ neurons. These results suggest that in vivo MCH neurons may originate from multiple cell lineages that arise through early dorsoventral patterning of the hypothalamus. Additionally, we found that Hh signaling supports the differentiation of mESCs into orexin/hypocretin neurons, a well-defined cell group intermingled with MCH neurons in the lateral hypothalamic area (LHA). The present study highlights and improves the utility of mESC culture in the analysis of the developmental programs of specific hypothalamic cell types.
Collapse
|
23
|
Hypothalamic melanin-concentrating hormone regulates hippocampus-dorsolateral septum activity. Nat Neurosci 2022; 25:61-71. [PMID: 34980924 PMCID: PMC8741735 DOI: 10.1038/s41593-021-00984-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/10/2021] [Indexed: 12/15/2022]
Abstract
Hypothalamic melanin-concentrating hormone (MCH) polypeptide contributes to regulating energy homeostasis, sleep, and memory, though the mechanistic bases of its effects are unknown. Here, in mice, we uncover the physiological mechanism underlying the functional role of MCH signaling in projections to the dorsolateral septum (dLS), a region involved in routing hippocampal firing rhythms and encoding spatial memory based on such rhythms. Firing activity within the dLS in response to dorsal CA3 (dCA3) excitation is limited by strong feed-forward inhibition (FFI). We find that MCH synchronizes dLS neuronal firing with its dCA3 inputs by enhancing GABA release, which subsequently reduces the FFI and augments dCA3 excitatory input strength, both via presynaptic mechanisms. At the functional level, our data reveal a role for MCH signaling in the dLS in facilitating spatial memory. These findings support a model in which peptidergic signaling within the dLS modulates dorsal hippocampal output and supports memory encoding.
Collapse
|
24
|
Kubin L. Breathing during sleep. HANDBOOK OF CLINICAL NEUROLOGY 2022; 188:179-199. [PMID: 35965026 DOI: 10.1016/b978-0-323-91534-2.00005-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The depth, rate, and regularity of breathing change following transition from wakefulness to sleep. Interactions between sleep and breathing involve direct effects of the central mechanisms that generate sleep states exerted at multiple respiratory regulatory sites, such as the central respiratory pattern generator, respiratory premotor pathways, and motoneurons that innervate the respiratory pump and upper airway muscles, as well as effects secondary to sleep-related changes in metabolism. This chapter discusses respiratory effects of sleep as they occur under physiologic conditions. Breathing and central respiratory neuronal activities during nonrapid eye movement (NREM) sleep and REM sleep are characterized in relation to activity of central wake-active and sleep-active neurons. Consideration is given to the obstructive sleep apnea syndrome because in this common disorder, state-dependent control of upper airway patency by upper airway muscles attains high significance and recurrent arousals from sleep are triggered by hypercapnic and hypoxic episodes. Selected clinical trials are discussed in which pharmacological interventions targeted transmission in noradrenergic, serotonergic, cholinergic, and other state-dependent pathways identified as mediators of ventilatory changes during sleep. Central pathways for arousals elicited by chemical stimulation of breathing are given special attention for their important role in sleep loss and fragmentation in sleep-related respiratory disorders.
Collapse
Affiliation(s)
- Leszek Kubin
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
25
|
Raitiere MN. The Elusive "Switch Process" in Bipolar Disorder and Photoperiodism: A Hypothesis Centering on NADPH Oxidase-Generated Reactive Oxygen Species Within the Bed Nucleus of the Stria Terminalis. Front Psychiatry 2022; 13:847584. [PMID: 35782417 PMCID: PMC9243387 DOI: 10.3389/fpsyt.2022.847584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
One of the most striking and least understood aspects of mood disorders involves the "switch process" which drives the dramatic state changes characteristic of bipolar disorder. In this paper we explore the bipolar switch mechanism as deeply grounded in forms of seasonal switching (for example, from summer to winter phenotypes) displayed by many mammalian species. Thus we develop a new and unifying hypothesis that involves four specific claims, all converging to demonstrate a deeper affinity between the bipolar switch process and the light-sensitive (photoperiodic) nonhuman switch sequence than has been appreciated. First, we suggest that rapid eye movement (REM) sleep in both human and nonhuman plays a key role in probing for those seasonal changes in length of day that trigger the organism's characteristic involutional response (in certain animals, hibernation) to shorter days. Second, we claim that this general mammalian response requires the integrity of a neural circuit centering on the anterior bed nucleus of the stria terminalis. Third, we propose that a key molecular mediator of the switch process in both nonhumans and seasonal humans involves reactive oxygen species (ROS) of a particular provenance, namely those created by the enzyme NADPH oxidase (NOX). This position diverges from one currently prominent among students of bipolar disorder. In that tradition, the fact that patients afflicted with bipolar-spectrum disorders display indices of oxidative damage is marshaled to support the conclusion that ROS, escaping adventitiously from mitochondria, have a near-exclusive pathological role. Instead, we believe that ROS, originating instead in membrane-affiliated NOX enzymes upstream from mitochondria, take part in an eminently physiological signaling process at work to some degree in all mammals. Fourth and finally, we speculate that the diversion of ROS from that purposeful, genetically rooted seasonal switching task into the domain of human pathology represents a surprisingly recent phenomenon. It is one instigated mainly by anthropogenic modifications of the environment, especially "light pollution."
Collapse
Affiliation(s)
- Martin N Raitiere
- Department of Psychiatry, Providence St. Vincent Medical Center, Portland, OR, United States
| |
Collapse
|
26
|
Wu J, Liu D, Li J, Sun J, Huang Y, Zhang S, Gao S, Mei W. Central Neural Circuits Orchestrating Thermogenesis, Sleep-Wakefulness States and General Anesthesia States. Curr Neuropharmacol 2022; 20:223-253. [PMID: 33632102 PMCID: PMC9199556 DOI: 10.2174/1570159x19666210225152728] [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: 07/24/2020] [Revised: 02/01/2021] [Accepted: 02/24/2021] [Indexed: 11/22/2022] Open
Abstract
Great progress has been made in specifically identifying the central neural circuits (CNCs) of the core body temperature (Tcore), sleep-wakefulness states (SWs), and general anesthesia states (GAs), mainly utilizing optogenetic or chemogenetic manipulations. We summarize the neuronal populations and neural pathways of these three CNCs, which gives evidence for the orchestration within these three CNCs, and the integrative regulation of these three CNCs by different environmental light signals. We also outline some transient receptor potential (TRP) channels that function in the CNCs-Tcore and are modulated by some general anesthetics, which makes TRP channels possible targets for addressing the general-anestheticsinduced- hypothermia (GAIH). We suggest this review will provide new orientations for further consummating these CNCs and elucidating the central mechanisms of GAIH.
Collapse
Affiliation(s)
- Jiayi Wu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Daiqiang Liu
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiayan Li
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia Sun
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yujie Huang
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuang Zhang
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shaojie Gao
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Mei
- Department of Anesthesiology and Pain Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
27
|
Iacovides S, Kamerman P, Baker FC, Mitchell D. Why It Is Important to Consider the Effects of Analgesics on Sleep: A Critical Review. Compr Physiol 2021; 11:2589-2619. [PMID: 34558668 DOI: 10.1002/cphy.c210006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We review the known physiological mechanisms underpinning all of pain processing, sleep regulation, and pharmacology of analgesics prescribed for chronic pain. In particular, we describe how commonly prescribed analgesics act in sleep-wake neural pathways, with potential unintended impact on sleep and/or wake function. Sleep disruption, whether pain- or drug-induced, negatively impacts quality of life, mental and physical health. In the context of chronic pain, poor sleep quality heightens pain sensitivity and may affect analgesic function, potentially resulting in further analgesic need. Clinicians already have to consider factors including efficacy, abuse potential, and likely side effects when making analgesic prescribing choices. We propose that analgesic-related sleep disruption should also be considered. The neurochemical mechanisms underlying the reciprocal relationship between pain and sleep are poorly understood, and studies investigating sleep in those with specific chronic pain conditions (including those with comorbidities) are lacking. We emphasize the importance of further work to clarify the effects (intended and unintended) of each analgesic class to inform personalized treatment decisions in patients with chronic pain. © 2021 American Physiological Society. Compr Physiol 11:1-31, 2021.
Collapse
Affiliation(s)
- Stella Iacovides
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Peter Kamerman
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Fiona C Baker
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Human Sleep Research Program, SRI International, Menlo Park, California, USA
| | - Duncan Mitchell
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
28
|
Deurveilher S, Antonchuk M, Saumure BSC, Baldin A, Semba K. No loss of orexin/hypocretin, melanin-concentrating hormone or locus coeruleus noradrenergic neurons in a rat model of chronic sleep restriction. Eur J Neurosci 2021; 54:6027-6043. [PMID: 34355453 DOI: 10.1111/ejn.15412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022]
Abstract
Chronic sleep restriction (CSR) is common in modern society, adversely affecting cognitive performance and health. Yet how it impacts neurons regulating sleep remains unclear. Several studies using mice reported substantial losses of wake-active orexin/hypocretin and locus coeruleus (LC) noradrenergic neurons, but not rapid eye movement sleep-active melanin-concentrating hormone (MCH) neurons, following CSR. Here, we used immunohistochemistry and stereology to examine orexin, MCH and LC noradrenergic neurons in a rat model of CSR that uses programmed wheel rotation (3 h on/1 h off; '3/1' protocol). Adult male Wistar rats underwent one or four cycles of the 4-day 3/1 CSR protocol, with 2-day recovery between cycles in home cages. Time-matched control rats were housed in locked wheels/home cages. We found no significant differences in the numbers of orexin, MCH and LC noradrenergic neurons following either one- or four-cycle CSR protocol compared to respective controls. Similarly, the four-cycle CSR protocol had no effect on the densities of orexin axon terminals in the LC, noradrenergic dendrites in the LC and noradrenergic axon terminals in the frontal cortex. Body weights, however, decreased after one cycle of CSR and then increased with diminishing slope over the next three cycles. Thus, we found no evidence for loss of orexin or LC noradrenergic neurons following one and four cycles of the 4-day 3/1 CSR protocol in rats. Differences in CSR protocols and/or possible species differences in neuronal vulnerability to sleep loss may account for the discrepancy between the current results in rats and previous findings in mice.
Collapse
Affiliation(s)
- Samuel Deurveilher
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michael Antonchuk
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Brock St C Saumure
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andrew Baldin
- 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 and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
29
|
Pho H, Berger S, Freire C, Kim LJ, Shin MK, Streeter SR, Hosamane N, Cabassa ME, Anokye-Danso F, Dergacheva O, Amorim MR, Fleury-Curado T, Jun JC, Schwartz AR, Ahima RS, Mendelowitz D, Polotsky VY. Leptin receptor expression in the dorsomedial hypothalamus stimulates breathing during NREM sleep in db/db mice. Sleep 2021; 44:6149135. [PMID: 33624805 PMCID: PMC8193564 DOI: 10.1093/sleep/zsab046] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/06/2021] [Indexed: 12/12/2022] Open
Abstract
STUDY OBJECTIVES Obesity leads to obstructive sleep apnea (OSA), which is recurrent upper airway obstruction during sleep, and obesity hypoventilation syndrome (OHS), hypoventilation during sleep resulting in daytime hypercapnia. Impaired leptin signaling in the brain was implicated in both conditions, but mechanisms are unknown. We have previously shown that leptin stimulates breathing and treats OSA and OHS in leptin-deficient ob/ob mice and leptin-resistant diet-induced obese mice and that leptin's respiratory effects may occur in the dorsomedial hypothalamus (DMH). We hypothesized that leptin receptor LepRb-deficient db/db mice have obesity hypoventilation and that restoration of leptin signaling in the DMH will increase ventilation during sleep in these animals. METHODS We measured arterial blood gas in unanesthetized awake db/db mice. We subsequently infected these animals with Ad-LepRb or control Ad-mCherry virus into the DMH and measured ventilation during sleep as well as CO2 production after intracerebroventricular (ICV) infusions of phosphate-buffered saline or leptin. RESULTS Awake db/db mice had elevated CO2 levels in the arterial blood. Ad-LepRb infection resulted in LepRb expression in the DMH neurons in a similar fashion to wildtype mice. In LepRb-DMH db/db mice, ICV leptin shortened REM sleep and increased inspiratory flow, tidal volume, and minute ventilation during NREM sleep without any effect on the quality of NREM sleep or CO2 production. Leptin had no effect on upper airway obstruction in these animals. CONCLUSION Leptin stimulates breathing and treats obesity hypoventilation acting on LepRb-positive neurons in the DMH.
Collapse
Affiliation(s)
- Huy Pho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Slava Berger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carla Freire
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lenise J Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mi-Kyung Shin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stone R Streeter
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nishitha Hosamane
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Meaghan E Cabassa
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frederick Anokye-Danso
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Olga Dergacheva
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Mateus R Amorim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomaz Fleury-Curado
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan C Jun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alan R Schwartz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rexford S Ahima
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Corresponding author. Vsevolod (Seva) Y. Polotsky, Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Johns Hopkins Asthma and Allergy Center, Rm 4B65, Baltimore, MD 21224.
| |
Collapse
|
30
|
Izawa S, Yoneshiro T, Kondoh K, Nakagiri S, Okamatsu-Ogura Y, Terao A, Minokoshi Y, Yamanaka A, Kimura K. Melanin-concentrating hormone-producing neurons in the hypothalamus regulate brown adipose tissue and thus contribute to energy expenditure. J Physiol 2021; 600:815-827. [PMID: 33899241 DOI: 10.1113/jp281241] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/20/2021] [Indexed: 12/17/2022] Open
Abstract
KEY POINTS Melanin-concentrating hormone (MCH) neuron-ablated mice exhibit increased energy expenditure and reduced fat weight. Increased brown adipose tissue (BAT) activity and locomotor activity-independent energy expenditure contributed to body weight reduction in MCH neuron-ablated mice. MCH neurons send inhibitory input to the medullary raphe nucleus to modulate BAT activity. ABSTRACT Hypothalamic melanin-concentrating hormone (MCH) peptide robustly affects energy homeostasis. However, it is unclear whether and how MCH-producing neurons, which contain and release a variety of neuropeptides/transmitters, regulate energy expenditure in the central nervous system and peripheral tissues. We thus examined the regulation of energy expenditure by MCH neurons, focusing on interscapular brown adipose tissue (BAT) activity. MCH neuron-ablated mice exhibited reduced body weight, increased oxygen consumption, and increased BAT activity, which improved locomotor activity-independent energy expenditure. Trans-neuronal retrograde tracing with the recombinant pseudorabies virus revealed that MCH neurons innervate BAT via the sympathetic premotor region in the medullary raphe nucleus (MRN). MRN neurons were activated by MCH neuron ablation. Therefore, endogenous MCH neuron activity negatively modulates energy expenditure via BAT inhibition. MRN neurons might receive inhibitory input from MCH neurons to suppress BAT activity.
Collapse
Affiliation(s)
- Shuntaro Izawa
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan.,Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.,JSPS Research Fellowship for Young Scientists, Tokyo, 102-0083, Japan
| | - Takeshi Yoneshiro
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan.,Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, 153-8904, Japan
| | - Kunio Kondoh
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8585, Japan.,Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8585, Japan
| | - Shohei Nakagiri
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Yuko Okamatsu-Ogura
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Akira Terao
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan.,Department of Biology, School of Biological Sciences, Tokai University, Sapporo, 005-8601, Japan
| | - Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8585, Japan.,Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8585, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan.,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Kazuhiro Kimura
- Laboratory of Biochemistry, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| |
Collapse
|
31
|
Adamantidis AR, Schmidt MH, Carter ME, Burdakov D, Peyron C, Scammell TE. A circuit perspective on narcolepsy. Sleep 2021; 43:5699663. [PMID: 31919524 PMCID: PMC7215265 DOI: 10.1093/sleep/zsz296] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/13/2019] [Indexed: 01/25/2023] Open
Abstract
The sleep disorder narcolepsy is associated with symptoms related to either boundary state control that include excessive daytime sleepiness and sleep fragmentation, or rapid eye movement (REM) sleep features including cataplexy, sleep paralysis, hallucinations, and sleep-onset REM sleep events (SOREMs). Although the loss of Hypocretin/Orexin (Hcrt/Ox) peptides or their receptors have been associated with the disease, here we propose a circuit perspective of the pathophysiological mechanisms of these narcolepsy symptoms that encompasses brain regions, neuronal circuits, cell types, and transmitters beyond the Hcrt/Ox system. We further discuss future experimental strategies to investigate brain-wide mechanisms of narcolepsy that will be essential for a better understanding and treatment of the disease.
Collapse
Affiliation(s)
- A R Adamantidis
- Department of Neurology, Centre for Experimental Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland.,Department of Biomedical Research, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| | - M H Schmidt
- Department of Neurology, Centre for Experimental Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland.,Ohio Sleep Medicine Institute, Dublin, OH
| | - M E Carter
- Department of Biology, Program in Neuroscience, Williams College, Williamstown, MA
| | - D Burdakov
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - C Peyron
- Center for Research in Neuroscience of Lyon, SLEEP team, CNRS UMR5292, INSERM U1028, University Lyon 1, Bron, France
| | - Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| |
Collapse
|
32
|
Abstract
Sleep and wakefulness are complex, tightly regulated behaviors that occur in virtually all animals. With recent exciting developments in neuroscience methodologies such as optogenetics, chemogenetics, and cell-specific calcium imaging technology, researchers can advance our understanding of how discrete neuronal groups precisely modulate states of sleep and wakefulness. In this chapter, we provide an overview of key neurotransmitter systems, neurons, and circuits that regulate states of sleep and wakefulness. We also describe long-standing models for the regulation of sleep/wake and non-rapid eye movement/rapid eye movement cycling. We contrast previous knowledge derived from classic approaches such as brain stimulation, lesions, cFos expression, and single-unit recordings, with emerging data using the newest technologies. Our understanding of neural circuits underlying the regulation of sleep and wakefulness is rapidly evolving, and this knowledge is critical for our field to elucidate the enigmatic function(s) of sleep.
Collapse
|
33
|
Pascovich C, Niño S, Mondino A, Lopez-Hill X, Urbanavicius J, Monti J, Lagos P, Torterolo P. Microinjection of melanin-concentrating hormone (MCH) into the median raphe nucleus promotes REM sleep in rats. Sleep Sci 2021; 14:229-235. [PMID: 35186201 PMCID: PMC8848522 DOI: 10.5935/1984-0063.20200075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/30/2020] [Indexed: 11/20/2022] Open
Abstract
Melanin concentrating hormone (MCH) is a sleep-promoting neuromodulator synthesized by neurons located in the postero-lateral hypothalamus and incerto-hypothalamic area. MCHergic neurons have widespread projections including the serotonergic dorsal (DR) and median (MnR) raphe nuclei, both involved in the control of wakefulness and sleep. In the present study, we explored in rats the presence of the MCH receptor type 1 (MCHR-1) in serotonergic neurons of the MnR by double immunofluorescence. Additionally, we analyzed the effect on sleep of MCH microinjections into the MnR. We found that MCHR-1 protein was present in MnR serotonergic and non-serotonergic neurons. In this respect, the receptor was localized in the primary cilia of these neurons. Compared with saline, microinjections of MCH into the MnR induced a dose-related increase in REM sleep time, which was related to a rise in the number of REM sleep episodes, associated with a reduction in the time spent in W. No significant changes were observed in non-REM (NREM) sleep time. Our data strongly suggest that MCH projections towards the MnR, acting through the MCHR-1 located in the primary cilia, promote REM sleep.
Collapse
Affiliation(s)
- Claudia Pascovich
- Facultad de Medicina, Universidad de la República, Fisiología, Montevideo - Uruguay
| | - Sofia Niño
- Facultad de Medicina, Universidad de la República, Fisiología, Montevideo - Uruguay
| | - Alejandra Mondino
- Facultad de Medicina, Universidad de la República, Fisiología, Montevideo - Uruguay
| | - Ximena Lopez-Hill
- Instituto de Investigaciones Biológicas Clemente Estable, Neurofarmacología Experimental, Montevideo - Uruguay
| | - Jessika Urbanavicius
- Instituto de Investigaciones Biológicas Clemente Estable, Neurofarmacología Experimental, Montevideo - Uruguay
| | - Jaime Monti
- Hospital de Clínicas, Farmacología y Terapéutica, Montevideo - Uruguay
| | - Patricia Lagos
- Facultad de Medicina, Universidad de la República, Fisiología, Montevideo - Uruguay
| | - Pablo Torterolo
- Facultad de Medicina, Universidad de la República, Fisiología, Montevideo - Uruguay. ,Corresponding author: Pablo Torterolo. E-mail: /
| |
Collapse
|
34
|
Optogenetic sleep enhancement improves fear-associated memory processing following trauma exposure in rats. Sci Rep 2020; 10:18025. [PMID: 33093538 PMCID: PMC7581760 DOI: 10.1038/s41598-020-75237-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/12/2020] [Indexed: 01/08/2023] Open
Abstract
Sleep disturbances are commonly found in trauma-exposed populations. Additionally, trauma exposure results in fear-associated memory impairments. Given the interactions of sleep with learning and memory, we hypothesized that increasing sleep duration following trauma exposure would restore overall function and improve trauma-induced fear-associated memory dysfunction. Here, we utilized single prolonged stress, a validated rodent model of post-traumatic stress disorder, in combination with optogenetic activation of hypothalamic melanin-concentrating hormone containing cells to increase sleep duration. The goal of this work was to ascertain if post-trauma sleep increases are sufficient to improve fear-associated memory function. In our laboratory, optogenetic stimulation after trauma exposure was sufficient to increase REM sleep duration during both the Light and Dark Phase, whereas NREM sleep duration was only increased during the Dark Phase of the circadian day. Interestingly though, animals that received optogenetic stimulation showed significantly improved fear-associated memory processing compared to non-stimulated controls. These results suggest that sleep therapeutics immediately following trauma exposure may be beneficial and that post-trauma sleep needs to be further examined in the context of the development of post-traumatic stress disorder.
Collapse
|
35
|
Bandaru SS, Khanday MA, Ibrahim N, Naganuma F, Vetrivelan R. Sleep-Wake Control by Melanin-Concentrating Hormone (MCH) Neurons: a Review of Recent Findings. Curr Neurol Neurosci Rep 2020; 20:55. [PMID: 33006677 PMCID: PMC11891936 DOI: 10.1007/s11910-020-01075-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE OF THE REVIEW Melanin-concentrating hormone (MCH)-expressing neurons located in the lateral hypothalamus are considered as an integral component of sleep-wake circuitry. However, the precise role of MCH neurons in sleep-wake regulation has remained unclear, despite several years of research employing a wide range of techniques. We review recent data on this aspect, which are mostly inconsistent, and propose a novel role for MCH neurons in sleep regulation. RECENT FINDINGS While almost all studies using "gain-of-function" approaches show an increase in rapid eye movement sleep (or paradoxical sleep; PS), loss-of-function approaches have not shown reductions in PS. Similarly, the reported changes in wakefulness or non-rapid eye movement sleep (slow-wave sleep; SWS) with manipulation of the MCH system using conditional genetic methods are inconsistent. Currently available data do not support a role for MCH neurons in spontaneous sleep-wake but imply a crucial role for them in orchestrating sleep-wake responses to changes in external and internal environments.
Collapse
Affiliation(s)
- Sathyajit S Bandaru
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA
| | - Mudasir A Khanday
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Nazifa Ibrahim
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA
- Department of Public Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Fumito Naganuma
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ramalingam Vetrivelan
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, Center for Life Science # 711, Boston, MA, USA.
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Wen Y, Lv Y, Niu J, Xin C, Cui L, Vetrivelan R, Lu J. Roles of motor and cortical activity in sleep rebound in rat. Eur J Neurosci 2020; 52:4100-4114. [PMID: 32588491 DOI: 10.1111/ejn.14881] [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: 10/29/2019] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 11/29/2022]
Abstract
Sleep pressure that builds up gradually during the extended wakefulness results in sleep rebound. Several lines of evidence, however, suggest that wake per se may not be sufficient to drive sleep rebound and that rapid eye movement (REM) and non-rapid eye movement (NREM) sleep rebound may be differentially regulated. In this study, we investigated the relative contribution of brain versus physical activities in REM and NREM sleep rebound by four sets of experiments. First, we forced locomotion in rats in a rotating wheel for 4 hr and examined subsequent sleep rebound. Second, we exposed the rats lacking homeostatic sleep response after prolonged quiet wakefulness and arousal brain activity induced by chemoactivation of parabrachial nucleus to the same rotating wheel paradigm and tested if physical activity could rescue the sleep homeostasis. Third, we varied motor activity levels while concurrently inhibiting the cortical activity by administering ketamine or xylazine (motor inhibitor), or ketamine + xylazine mixture and investigated if motor activity in the absence of activated cortex can cause NREM sleep rebound. Fourth and finally, we manipulated cortical activity by administering ketamine (that induced active wakefulness and waking brain) alone or in combination with atropine (that selectively inhibits the cortex) and studied if cortical inhibition irrespective of motor activity levels can block REM sleep rebound. Our results demonstrate that motor activity but not cortical activity determines NREM sleep rebound whereas cortical activity but not motor activity determines REM sleep rebound.
Collapse
Affiliation(s)
- Yujun Wen
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Ningxia Key Laboratory of Craniocerebral Diseases, Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Yudan Lv
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Department of Neurology, Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jianguo Niu
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Ningxia Key Laboratory of Craniocerebral Diseases, Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Christopher Xin
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Li Cui
- Department of Neurology, Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ramalingam Vetrivelan
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jun Lu
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
37
|
Park SH, Weber F. Neural and Homeostatic Regulation of REM Sleep. Front Psychol 2020; 11:1662. [PMID: 32793050 PMCID: PMC7385183 DOI: 10.3389/fpsyg.2020.01662] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022] Open
Abstract
Rapid eye movement (REM) sleep is a distinct, homeostatically controlled brain state characterized by an activated electroencephalogram (EEG) in combination with paralysis of skeletal muscles and is associated with vivid dreaming. Understanding how REM sleep is controlled requires identification of the neural circuits underlying its initiation and maintenance, and delineation of the homeostatic processes regulating its expression on multiple timescales. Soon after its discovery in humans in 1953, the pons was demonstrated to be necessary and sufficient for the generation of REM sleep. But, especially within the last decade, researchers have identified further neural populations in the hypothalamus, midbrain, and medulla that regulate REM sleep by either promoting or suppressing this brain state. The discovery of these populations was greatly facilitated by the availability of novel technologies for the dissection of neural circuits. Recent quantitative models integrate findings about the activity and connectivity of key neurons and knowledge about homeostatic mechanisms to explain the dynamics underlying the recurrence of REM sleep. For the future, combining quantitative with experimental approaches to directly test model predictions and to refine existing models will greatly advance our understanding of the neural and homeostatic processes governing the regulation of REM sleep.
Collapse
Affiliation(s)
| | - Franz Weber
- Department of Neuroscience, Perelman School of Medicine, Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
38
|
Horne J. REM sleep vs exploratory wakefulness: Alternatives within adult ‘sleep debt’? Sleep Med Rev 2020; 50:101252. [DOI: 10.1016/j.smrv.2019.101252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/06/2019] [Accepted: 12/06/2019] [Indexed: 10/25/2022]
|
39
|
Regan MD, Flynn-Evans EE, Griko YV, Kilduff TS, Rittenberger JC, Ruskin KJ, Buck CL. Shallow metabolic depression and human spaceflight: a feasible first step. J Appl Physiol (1985) 2020; 128:637-647. [PMID: 31999524 DOI: 10.1152/japplphysiol.00725.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Synthetic torpor is an induced state of deep metabolic depression (MD) in an organism that does not naturally employ regulated and reversible MD. If applied to spaceflight crewmembers, this metabolic state may theoretically mitigate numerous biological and logistical challenges of human spaceflight. These benefits have been the focus of numerous recent articles where, invariably, they are discussed in the context of hypothetical deep MD states in which the metabolism of crewmembers is profoundly depressed relative to basal rates. However, inducing these deep MD states in humans, particularly humans aboard spacecraft, is currently impossible. Here, we discuss shallow MD as a feasible first step toward synthetic torpor during spaceflight and summarize perspectives following a recent NASA-hosted workshop. We discuss methods to safely induce shallow MD (e.g., sleep and slow wave enhancement via acoustic and photoperiod stimulation; moderate sedation via dexmedetomidine), which we define as an ~20% depression of metabolic rate relative to basal levels. We also discuss different modes of shallow MD application (e.g., habitual versus targeted, whereby shallow MD is induced routinely throughout a mission or only under certain circumstances, respectively) and different spaceflight scenarios that would benefit from its use. Finally, we propose a multistep development plan toward the application of synthetic torpor to human spaceflight, highlighting shallow MD's role. As space agencies develop missions to send humans further into space than ever before, shallow MD has the potential to confer health benefits for crewmembers, reduce demands on spacecraft capacities, and serve as a testbed for deeper MD technologies.
Collapse
Affiliation(s)
- Matthew D Regan
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Erin E Flynn-Evans
- Fatigue Countermeasures Laboratory, Human Systems Integration Division, NASA Ames Research Center, Moffett Field, California
| | - Yuri V Griko
- Countermeasure Development Laboratory, Space Biosciences Division, NASA Ames Research Center, Moffett Field, California
| | - Thomas S Kilduff
- Biosciences Division, Center for Neuroscience, SRI International, Menlo Park, California
| | - Jon C Rittenberger
- Guthrie Robert Packer Hospital Emergency Medicine Program, Geisinger Commonwealth School of Medicine, Scranton, Pennsylvania
| | - Keith J Ruskin
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, Illinois
| | - C Loren Buck
- Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona
| |
Collapse
|
40
|
Scammell TE, Jackson AC, Franks NP, Wisden W, Dauvilliers Y. Histamine: neural circuits and new medications. Sleep 2019; 42:5099478. [PMID: 30239935 PMCID: PMC6335869 DOI: 10.1093/sleep/zsy183] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Indexed: 12/12/2022] Open
Abstract
Histamine was first identified in the brain about 50 years ago, but only in the last few years have researchers gained an understanding of how it regulates sleep/wake behavior. We provide a translational overview of the histamine system, from basic research to new clinical trials demonstrating the usefulness of drugs that enhance histamine signaling. The tuberomammillary nucleus is the sole neuronal source of histamine in the brain, and like many of the arousal systems, histamine neurons diffusely innervate the cortex, thalamus, and other wake-promoting brain regions. Histamine has generally excitatory effects on target neurons, but paradoxically, histamine neurons may also release the inhibitory neurotransmitter GABA. New research demonstrates that activity in histamine neurons is essential for normal wakefulness, especially at specific circadian phases, and reducing activity in these neurons can produce sedation. The number of histamine neurons is increased in narcolepsy, but whether this affects brain levels of histamine is controversial. Of clinical importance, new compounds are becoming available that enhance histamine signaling, and clinical trials show that these medications reduce sleepiness and cataplexy in narcolepsy.
Collapse
Affiliation(s)
- Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Alexander C Jackson
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT
| | - Nicholas P Franks
- Department of Life Sciences and UK Dementia Research Institute, Imperial College London, UK
| | - William Wisden
- Department of Life Sciences and UK Dementia Research Institute, Imperial College London, UK
| | - Yves Dauvilliers
- Centre National de Référence Narcolepsie Hypersomnies, Unité des Troubles du Sommeil, Service de Neurologie, Hôpital Gui-de-Chauliac, Université Montpellier, INSERM, Montpellier, France
| |
Collapse
|
41
|
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
|
42
|
Thorpy MJ, Bogan RK. Update on the pharmacologic management of narcolepsy: mechanisms of action and clinical implications. Sleep Med 2019; 68:97-109. [PMID: 32032921 DOI: 10.1016/j.sleep.2019.09.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/21/2022]
Abstract
Narcolepsy is a chronic, debilitating neurological disorder of sleep-wake state instability. This instability underlies all narcolepsy symptoms, including excessive daytime sleepiness (EDS), symptoms of rapid eye movement (REM) sleep dysregulation (ie, cataplexy, hypnagogic/hypnopompic hallucinations, sleep paralysis), and disrupted nighttime sleep. Several neurotransmitter systems promote wakefulness, and various neural pathways are involved in regulating REM sleep-related muscle atonia, providing multiple targets for pharmacologic intervention to reduce EDS and cataplexy. Medications approved by the US Food and Drug Administration (FDA) for the treatment of EDS in narcolepsy include traditional stimulants (eg, amphetamines, methylphenidate), wake-promoting agents (eg, modafinil, armodafinil), and solriamfetol, which mainly act on dopaminergic and noradrenergic pathways. Sodium oxybate (thought to act via GABAB receptors) is FDA-approved for the treatment of EDS and cataplexy. Pitolisant, a histamine 3 (H3)-receptor antagonist/inverse agonist, is approved by the European Medicines Agency (EMA) for the treatment of narcolepsy with or without cataplexy in adults and by the FDA for the treatment of EDS in adults with narcolepsy. Pitolisant increases the synthesis and release of histamine in the brain and modulates the release of other neurotransmitters (eg, norepinephrine, dopamine). Antidepressants that inhibit reuptake of serotonin and/or norepinephrine are widely used off label to manage cataplexy. In many patients with narcolepsy, combination treatment with medications that act via different neural pathways is necessary for optimal symptom management. Mechanism of action, pharmacokinetics, and abuse potential are important considerations in treatment selection and subsequent medication adjustments to maximize efficacy and mitigate adverse effects in the treatment of patients with narcolepsy.
Collapse
Affiliation(s)
- Michael J Thorpy
- Sleep-Wake Disorders Center, Montefiore Medical Center, Albert Einstein College of Medicine, 3411 Wayne Ave, Bronx, NY, 10467, USA.
| | - Richard K Bogan
- SleepMed Inc., Bogan Sleep Consultants, LLC, 1333 Taylor Street, Columbia, SC, USA.
| |
Collapse
|
43
|
The role of co-neurotransmitters in sleep and wake regulation. Mol Psychiatry 2019; 24:1284-1295. [PMID: 30377299 PMCID: PMC6491268 DOI: 10.1038/s41380-018-0291-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 09/17/2018] [Accepted: 10/08/2018] [Indexed: 12/11/2022]
Abstract
Sleep and wakefulness control in the mammalian brain requires the coordination of various discrete interconnected neurons. According to the most conventional sleep model, wake-promoting neurons (WPNs) and sleep-promoting neurons (SPNs) compete for network dominance, creating a systematic "switch" that results in either the sleep or awake state. WPNs and SPNs are ubiquitous in the brainstem and diencephalon, areas that together contain <1% of the neurons in the human brain. Interestingly, many of these WPNs and SPNs co-express and co-release various types of the neurotransmitters that often have opposing modulatory effects on the network. Co-transmission is often beneficial to structures with limited numbers of neurons because it provides increasing computational capability and flexibility. Moreover, co-transmission allows subcortical structures to bi-directionally control postsynaptic neurons, thus helping to orchestrate several complex physiological functions such as sleep. Here, we present an in-depth review of co-transmission in hypothalamic WPNs and SPNs and discuss its functional significance in the sleep-wake network.
Collapse
|
44
|
Varga AW, Mokhlesi B. REM obstructive sleep apnea: risk for adverse health outcomes and novel treatments. Sleep Breath 2019; 23:413-423. [PMID: 30232681 PMCID: PMC6424642 DOI: 10.1007/s11325-018-1727-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 09/04/2018] [Accepted: 09/14/2018] [Indexed: 02/07/2023]
Abstract
Rapid eye movement (REM) sleep was discovered nearly 60 years ago. This stage of sleep accounts for approximately a quarter of total sleep time in healthy adults, and it is mostly concentrated in the second half of the sleep period. The majority of research on REM sleep has focused on neurocognition. More recently, however, there has been a growing interest in understanding whether obstructive sleep apnea (OSA) during the two main stages of sleep (REM and non-REM sleep) leads to different cardiometabolic and neurocognitive risk. In this review, we discuss the growing evidence indicating that OSA during REM sleep is a prevalent disorder that is independently associated with adverse cardiovascular, metabolic, and neurocognitive outcomes. From a therapeutic standpoint, we discuss limitations of continuous positive airway pressure (CPAP) therapy given that 3 or 4 h of CPAP use from the beginning of the sleep period would leave 75% or 60% of obstructive events during REM sleep untreated. We also review potential pharmacologic approaches to treating OSA during REM sleep. Undoubtedly, further research is needed to establish best treatment strategies in order to effectively treat REM OSA. Moreover, it is critical to understand whether treatment of REM OSA will translate into better patient outcomes.
Collapse
Affiliation(s)
- Andrew W Varga
- Mount Sinai Integrative Sleep Center, Icahn School of Medicine at Mount Sinai, Annenberg 21-44, One Gustave L. Levy Place, New York, NY, 10029, USA.
| | - Babak Mokhlesi
- Section of Pulmonary and Critical Care Medicine, Sleep Disorders Center, University of Chicago, Chicago, IL, USA
| |
Collapse
|
45
|
Komagata N, Latifi B, Rusterholz T, Bassetti CLA, Adamantidis A, Schmidt MH. Dynamic REM Sleep Modulation by Ambient Temperature and the Critical Role of the Melanin-Concentrating Hormone System. Curr Biol 2019; 29:1976-1987.e4. [PMID: 31155350 DOI: 10.1016/j.cub.2019.05.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/02/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
Ambient temperature (Ta) warming toward the high end of the thermoneutral zone (TNZ) preferentially increases rapid eye movement (REM) sleep over non-REM (NREM) sleep across species. The control and function of this temperature-induced REM sleep expression have remained unknown. Melanin-concentrating hormone (MCH) neurons play an important role in REM sleep control. We hypothesize that the MCH system may modulate REM sleep as a function of Ta. Here, we show that wild-type (WT) mice dynamically increased REM sleep durations specifically during warm Ta pulsing within the TNZ, compared to both the TNZ cool and baseline constant Ta conditions, without significantly affecting either wake or NREM sleep durations. However, genetically engineered MCH receptor-1 knockout (MCHR1-KO) mice showed no significant changes in REM sleep as a function of Ta, even with increased sleep pressure following a 4-h sleep deprivation. Using MCH-cre mice transduced with channelrhodopsin, we then optogenetically activated MCH neurons time locked with Ta warming, showing an increase in REM sleep expression beyond what Ta warming in yellow fluorescent protein (YFP) control mice achieved. Finally, in mice transduced with archaerhodopsin-T, semi-chronic optogenetic MCH neuronal silencing during Ta warming completely blocked the increase in REM sleep seen in YFP controls. These data demonstrate a previously unknown role for the MCH system in the dynamic output expression of REM sleep during Ta manipulation. These findings are consistent with the energy allocation hypothesis of sleep function, suggesting that endotherms have evolved neural circuits to opportunistically express REM sleep when the need for thermoregulatory defense is minimized.
Collapse
Affiliation(s)
- Noëmie Komagata
- Bern University Hospital (Inselspital), University of Bern, 3010 Bern, Switzerland
| | - Blerina Latifi
- Bern University Hospital (Inselspital), University of Bern, 3010 Bern, Switzerland
| | - Thomas Rusterholz
- Center for Experimental Neurology, Department of Neurology, Bern University Hospital (Inselspital), University of Bern, 3010 Bern, Switzerland
| | - Claudio L A Bassetti
- Department of Neurology, Bern University Hospital (Inselspital), University of Bern, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Antoine Adamantidis
- Center for Experimental Neurology, Department of Neurology, Bern University Hospital (Inselspital), University of Bern, 3010 Bern, Switzerland; Department of Biomedical Research (DBMR), Bern University Hospital (Inselspital), University of Bern, 3010 Bern, Switzerland
| | - Markus H Schmidt
- Center for Experimental Neurology, Department of Neurology, Bern University Hospital (Inselspital), University of Bern, 3010 Bern, Switzerland; Department of Neurology, Bern University Hospital (Inselspital), University of Bern, Freiburgstrasse 18, 3010 Bern, Switzerland; Ohio Sleep Medicine Institute, 4975 Bradenton Avenue, Dublin, OH 43017, USA.
| |
Collapse
|
46
|
Chen ZK, Yuan XS, Dong H, Wu YF, Chen GH, He M, Qu WM, Huang ZL. Whole-Brain Neural Connectivity to Lateral Pontine Tegmentum GABAergic Neurons in Mice. Front Neurosci 2019; 13:375. [PMID: 31068780 PMCID: PMC6491572 DOI: 10.3389/fnins.2019.00375] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/01/2019] [Indexed: 01/22/2023] Open
Abstract
The GABAergic neurons in the lateral pontine tegmentum (LPT) play key roles in the regulation of sleep and locomotion. The dysfunction of the LPT is related to neurological disorders such as rapid eye movement sleep behavior disorder and ocular flutter. However, the whole-brain neural connectivity to LPT GABAergic neurons remains poorly understood. Using virus-based, cell-type-specific, retrograde and anterograde tracing systems, we mapped the monosynaptic inputs and axonal projections of LPT GABAergic neurons in mice. We found that LPT GABAergic neurons received inputs mainly from the superior colliculus, substantia nigra pars reticulata, dorsal raphe nucleus (DR), lateral hypothalamic area (LHA), parasubthalamic nucleus, and periaqueductal gray (PAG), as well as the limbic system (e.g., central nucleus of the amygdala). Further immunofluorescence assays revealed that the inputs to LPT GABAergic neurons were colocalized with several markers associated with important neural functions, especially the sleep-wake cycle. Moreover, numerous LPT GABAergic neuronal varicosities were observed in the medial and midline part of the thalamus, the LHA, PAG, DR, and parabrachial nuclei. Interestingly, LPT GABAergic neurons formed reciprocal connections with areas related to sleep-wake and motor control, including the LHA, PAG, DR, parabrachial nuclei, and superior colliculus, only the LPT-DR connections were in an equally bidirectional manner. These results provide a structural framework to understand the underlying neural mechanisms of rapid eye movement sleep behavior disorder and disorders of saccades.
Collapse
Affiliation(s)
- Ze-Ka Chen
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiang-Shan Yuan
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hui Dong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yong-Fang Wu
- Department of Neurology (Sleep Disorders), Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Miao He
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wei-Min Qu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhi-Li Huang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
47
|
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
|
48
|
Kroeger D, Bandaru SS, Madara JC, Vetrivelan R. Ventrolateral periaqueductal gray mediates rapid eye movement sleep regulation by melanin-concentrating hormone neurons. Neuroscience 2019; 406:314-324. [PMID: 30890480 DOI: 10.1016/j.neuroscience.2019.03.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 02/20/2019] [Accepted: 03/08/2019] [Indexed: 11/28/2022]
Abstract
Neurons containing melanin-concentrating hormone (MCH) in the lateral hypothalamic area (LH) have been shown to promote rapid eye movement sleep (REMs) in mice. However, the downstream neural pathways through which MCH neurons influence REMs remained unclear. Because MCH neurons are considered to be primarily inhibitory, we hypothesized that these neurons inhibit the midbrain 'REMs-suppressing' region consisting of the ventrolateral periaqueductal gray and the lateral pontine tegmentum (vlPAG/LPT) to promote REMs. To test this hypothesis, we optogenetically inhibited MCH terminals in the vlPAG/LPT under baseline conditions as well as with simultaneous chemogenetic activation of MCH soma. We found that inhibition of MCH terminals in the vlPAG/LPT significantly reduced transitions into REMs during spontaneous sleep-wake cycles and prevented the increase in REMs transitions observed after chemogenetic activation of MCH neurons. These results strongly suggest that the vlPAG/LPT may be an essential relay through which MCH neurons modulate REMs.
Collapse
Affiliation(s)
- Daniel Kroeger
- Department of Neurology, Program in Neuroscience and Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA-02215, United States.
| | - Sathyajit S Bandaru
- Department of Neurology, Program in Neuroscience and Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA-02215, United States.
| | - Joseph C Madara
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA-02215, United States.
| | - Ramalingam Vetrivelan
- Department of Neurology, Program in Neuroscience and Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA-02215, United States.
| |
Collapse
|
49
|
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
|
50
|
Winsky-Sommerer R, de Oliveira P, Loomis S, Wafford K, Dijk DJ, Gilmour G. Disturbances of sleep quality, timing and structure and their relationship with other neuropsychiatric symptoms in Alzheimer’s disease and schizophrenia: Insights from studies in patient populations and animal models. Neurosci Biobehav Rev 2019; 97:112-137. [DOI: 10.1016/j.neubiorev.2018.09.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 08/31/2018] [Accepted: 09/30/2018] [Indexed: 02/06/2023]
|