51
|
Gangitano E, Martinez-Sanchez N, Bellini MI, Urciuoli I, Monterisi S, Mariani S, Ray D, Gnessi L. Weight Loss and Sleep, Current Evidence in Animal Models and Humans. Nutrients 2023; 15:3431. [PMID: 37571368 PMCID: PMC10420950 DOI: 10.3390/nu15153431] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Sleep is a vital process essential for survival. The trend of reduction in the time dedicated to sleep has increased in industrialized countries, together with the dramatic increase in the prevalence of obesity and diabetes. Short sleep may increase the risk of obesity, diabetes and cardiovascular disease, and on the other hand, obesity is associated with sleep disorders, such as obstructive apnea disease, insomnia and excessive daytime sleepiness. Sleep and metabolic disorders are linked; therefore, identifying the physiological and molecular pathways involved in sleep regulation and metabolic homeostasis can play a major role in ameliorating the metabolic health of the individual. Approaches aimed at reducing body weight could provide benefits for both cardiometabolic risk and sleep quality, which indirectly, in turn, may determine an amelioration of the cardiometabolic phenotype of individuals. We revised the literature on weight loss and sleep, focusing on the mechanisms and the molecules that may subtend this relationship in humans as in animal models.
Collapse
Affiliation(s)
- Elena Gangitano
- OCDEM Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Noelia Martinez-Sanchez
- OCDEM Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
| | | | - Irene Urciuoli
- Department of Surgery, Sapienza University of Rome, 00161 Rome, Italy
| | - Stefania Monterisi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Stefania Mariani
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - David Ray
- OCDEM Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, UK
| | - Lucio Gnessi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
52
|
Lo YJ, Mishra VK, Lo HY, Dubey NK, Lo WC. Clinical Spectrum and Trajectory of Innovative Therapeutic Interventions for Insomnia: A Perspective. Aging Dis 2023; 14:1038-1069. [PMID: 37163444 PMCID: PMC10389812 DOI: 10.14336/ad.2022.1203] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/03/2022] [Indexed: 05/12/2023] Open
Abstract
Increasing incidences of insomnia in adults, as well as the aging population, have been reported for their negative impact on the quality of life. Insomnia episodes may be associated with neurocognitive, musculoskeletal, cardiovascular, gastrointestinal, renal, hepatic, and metabolic disorders. Epidemiological evidence also revealed the association of insomnia with oncologic and asthmatic complications, which has been indicated as bidirectional. Two therapeutic approaches including cognitive behavioral therapy (CBT) and drugs-based therapies are being practiced for a long time. However, the adverse events associated with drugs limit their wide and long-term application. Further, Traditional Chinese medicine, acupressure, and pulsed magnetic field therapy may also provide therapeutic relief. Notably, the recently introduced cryotherapy has been demonstrated as a potential candidate for insomnia which could reduce pain, by suppressing oxidative stress and inflammation. It seems that the synergistic therapeutic approach of cryotherapy and the above-mentioned approaches might offer promising prospects to further improve efficacy and safety. Considering these facts, this perspective presents a comprehensive summary of recent advances in pathological aetiologies of insomnia including COVID-19, and its therapeutic management with a greater emphasis on cryotherapy.
Collapse
Affiliation(s)
| | | | | | - Navneet Kumar Dubey
- Victory Biotechnology Co., Ltd., Taipei 114757, Taiwan.
- ShiNeo Technology Co., Ltd., New Taipei City 24262, Taiwan.
| | - Wen-Cheng Lo
- Department of Surgery, Division of Neurosurgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 11031, Taiwan.
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
53
|
Dawson M, Terstege DJ, Jamani N, Tsutsui M, Pavlov D, Bugescu R, Epp JR, Leinninger GM, Sargin D. Hypocretin/orexin neurons encode social discrimination and exhibit a sex-dependent necessity for social interaction. Cell Rep 2023; 42:112815. [PMID: 37459234 DOI: 10.1016/j.celrep.2023.112815] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 05/20/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023] Open
Abstract
The hypothalamus plays a crucial role in the modulation of social behavior by encoding internal states. The hypothalamic hypocretin/orexin neurons, initially identified as regulators of sleep and appetite, are important for emotional and motivated behaviors. However, their role in social behavior remains unclear. Using fiber photometry and behavioral analysis, we show here that hypocretin neurons differentially encode social discrimination based on the nature of social encounters. The optogenetic inhibition of hypocretin neuron activity or blocking of hcrt-1 receptors reduces the amount of time mice are engaged in social interaction in males but not in females. Reduced hcrt-1 receptor signaling during social interaction is associated with altered activity in the insular cortex and ventral tegmental area in males. Our data implicating hypocretin neurons as sexually dimorphic regulators within social networks have significant implications for the treatment of neuropsychiatric diseases with social dysfunction, particularly considering varying prevalence among sexes.
Collapse
Affiliation(s)
- Matthew Dawson
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Dylan J Terstege
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Naila Jamani
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Mio Tsutsui
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Dmitrii Pavlov
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Raluca Bugescu
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Jonathan R Epp
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Gina M Leinninger
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Derya Sargin
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
54
|
Bian WJ, González OC, de Lecea L. Adolescent sleep defects and dopaminergic hyperactivity in mice with a schizophrenia-linked Shank3 mutation. Sleep 2023; 46:zsad131. [PMID: 37144901 PMCID: PMC10334736 DOI: 10.1093/sleep/zsad131] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/03/2023] [Indexed: 05/06/2023] Open
Abstract
Shank3 is a shared risk gene for autism spectrum disorders and schizophrenia. Sleep defects have been characterized for autism models with Shank3 mutations; however, evidence has been lacking for the potential sleep defects caused by Shank3 mutation associated with schizophrenia and how early in development these defects may occur. Here we characterized the sleep architecture of adolescent mice carrying a schizophrenia-linked, R1117X mutation in Shank3. We further employed GRABDA dopamine sensor and fiber photometry to record dopamine release in the nucleus accumbens during sleep/wake states. Our results show that homozygous mutant R1117X mice have significantly reduced sleep in the dark phase during adolescence, altered electroencephalogram power, especially during the rapid-eye-movement sleep, and dopamine hyperactivity during sleep but not during wakefulness. Further analyses suggest that these adolescent defects in sleep architecture and dopaminergic neuromodulation tightly correlate with the social novelty preference later in adulthood and predict adult social performance during same-sex social interactions. Our results provide novel insights into the sleep phenotypes in mouse models of schizophrenia and the potential use of developmental sleep as a predictive metric for adult social symptoms. Together with recent studies in other Shank3 models, our work underscores the idea that Shank3-involved circuit disruptions may be one of the shared pathologies in certain types of schizophrenia and autism. Future research is needed to establish the causal relationship among adolescent sleep defects, dopaminergic dysregulation, and adult behavioral changes in Shank3 mutation animals and other models.
Collapse
Affiliation(s)
- Wen-Jie Bian
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Oscar C González
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
55
|
Nir Y, de Lecea L. Sleep and vigilance states: Embracing spatiotemporal dynamics. Neuron 2023; 111:1998-2011. [PMID: 37148873 DOI: 10.1016/j.neuron.2023.04.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/08/2023] [Accepted: 04/12/2023] [Indexed: 05/08/2023]
Abstract
The classic view of sleep and vigilance states is a global stationary perspective driven by the interaction between neuromodulators and thalamocortical systems. However, recent data are challenging this view by demonstrating that vigilance states are highly dynamic and regionally complex. Spatially, sleep- and wake-like states often co-occur across distinct brain regions, as in unihemispheric sleep, local sleep in wakefulness, and during development. Temporally, dynamic switching prevails around state transitions, during extended wakefulness, and in fragmented sleep. This knowledge, together with methods monitoring brain activity across multiple regions simultaneously at millisecond resolution with cell-type specificity, is rapidly shifting how we consider vigilance states. A new perspective incorporating multiple spatial and temporal scales may have important implications for considering the governing neuromodulatory mechanisms, the functional roles of vigilance states, and their behavioral manifestations. A modular and dynamic view highlights novel avenues for finer spatiotemporal interventions to improve sleep function.
Collapse
Affiliation(s)
- Yuval Nir
- Department of Physiology and Pharmacology, Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; The Sieratzki-Sagol Center for Sleep Medicine, Tel-Aviv Sourasky Medical Center, Tel-Aviv 64239, Israel.
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
56
|
Glickman B, LaLumiere RT. Theoretical Considerations for Optimizing the Use of Optogenetics with Complex Behavior. Curr Protoc 2023; 3:e836. [PMID: 37439512 PMCID: PMC10406170 DOI: 10.1002/cpz1.836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Optogenetic approaches have allowed researchers to address complex questions about behavior that were previously unanswerable. However, as optogenetic procedures involve a large parameter space across multiple dimensions, it is crucial to consider such parameters in conjunction with the behaviors under study. Here, we discuss strategies to optimize optogenetic approaches with complex behavior by identifying critical experimental design considerations, including frequency specificity, temporal precision, activity-controlled optogenetics, stimulation pattern, and cell-type specificity. We highlight potential limitations or theoretical considerations to be made when manipulating each of these factors of optogenetic experiments. This overview emphasizes the importance of optimizing optogenetic study design to enhance the conclusions that can be drawn about the neuroscience of behavior. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Bess Glickman
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA 52242
| | - Ryan T. LaLumiere
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA 52242
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
57
|
Rivas M, Ferreira A, Torterolo P, Benedetto L. Hypocretins, sleep, and maternal behavior. Front Behav Neurosci 2023; 17:1184885. [PMID: 37456808 PMCID: PMC10347526 DOI: 10.3389/fnbeh.2023.1184885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
The postpartum period is a demanding time during which mothers experience numerous physiological adaptations that enable them to care for their offspring while maintaining their wellbeing. Hypocretins, also known as orexins, are neuropeptides synthesized by hypothalamic neurons that play a fundamental role in several functions, including the promotion of wakefulness and motivated behaviors, such as maternal care. In this regard, several findings suggest that the activity of the hypocretinergic system increases in the early postpartum period and begins to decline as weaning approaches. In particular, hypocretins within the medial preoptic area, a crucial region during this period, modulate both maternal behavior and sleep. Although further studies are necessary to obtain a comprehensive understanding of the role of hypocretins in lactating females, current research suggests that this system participates in promoting active components of maternal behavior and regulating wakefulness and sleep adjustments during the postpartum period, potentially leading to increased wakefulness during this stage. These adaptive adjustments enable the mother to cope with the continuously changing demands of the pups.
Collapse
Affiliation(s)
- Mayda Rivas
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Annabel Ferreira
- Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Pablo Torterolo
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Luciana Benedetto
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
58
|
Zhou Z, Wang X, Li X, Liao L. A bibliometric profile of optogenetics: quantitative and qualitative analyses. Front Neurosci 2023; 17:1221316. [PMID: 37424998 PMCID: PMC10323434 DOI: 10.3389/fnins.2023.1221316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Optogenetics is a rapidly developing field combining optics and genetics, with promising applications in neuroscience and beyond. However, there is currently a lack of bibliometric analyses examining publications in this area. Method Publications on optogenetics were gathered from the Web of Science Core Collection Database. A quantitative analysis was conducted to gain insights into the annual scientific output, and distribution of authors, journals, subject categories, countries, and institutions. Additionally, qualitative analysis, such as co-occurrence network analysis, thematic analysis, and theme evolution, were performed to identify the main areas and trends of optogenetics articles. Results A total of 6,824 publications were included for analysis. The number of articles has rapidly grown since 2010, with an annual growth rate of 52.82%. Deisseroth K, Boyden ES, and Hegemann P were the most prolific contributors to the field. The United States contributed the most articles (3,051 articles), followed by China (623 articles). A majority of optogenetics-related articles are published in high-quality journals, including NATURE, SCIENCE, and CELL. These articles mainly belong to four subjects: neurosciences, biochemistry and molecular biology, neuroimaging, and materials science. Co-occurrence keyword network analysis identified three clusters: optogenetic components and techniques, optogenetics and neural circuitry, optogenetics and disease. Conclusion The results suggest that optogenetics research is flourishing, focusing on optogenetic techniques and their applications in neural circuitry exploration and disease intervention. Optogenetics is expected to remain a hot topic in various fields in the future.
Collapse
Affiliation(s)
- Zhonghan Zhou
- Shandong University, Jinan, Shandong, China
- Department of Urology, China Rehabilitation Research Center, Beijing, China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
- China Rehabilitation Science Institute, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Xuesheng Wang
- Department of Urology, China Rehabilitation Research Center, Beijing, China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
- China Rehabilitation Science Institute, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| | - Xunhua Li
- Department of Urology, China Rehabilitation Research Center, Beijing, China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
- China Rehabilitation Science Institute, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| | - Limin Liao
- Shandong University, Jinan, Shandong, China
- Department of Urology, China Rehabilitation Research Center, Beijing, China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
- China Rehabilitation Science Institute, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| |
Collapse
|
59
|
Pai SK. Mechanisms underlying fragmented sleep in aging. AGING BRAIN 2023; 3:100077. [PMID: 37304173 PMCID: PMC10248856 DOI: 10.1016/j.nbas.2023.100077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 06/13/2023] Open
|
60
|
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: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 04/10/2023] [Indexed: 05/17/2023]
Abstract
Early Alzheimer's disease (AD) is associated with hippocampal hyperactivity and decreased sleep quality. Here we show that homeostatic mechanisms transiently counteract the increased excitatory drive to CA1 neurons in AppNL-G-F mice, but that this mechanism fails in older mice. Spatial transcriptomics analysis identifies Pmch as part of the adaptive response in AppNL-G-F mice. Pmch encodes melanin-concentrating hormone (MCH), which is produced in sleep-active lateral hypothalamic neurons that project to CA1 and modulate memory. We show that MCH downregulates synaptic transmission, modulates firing rate homeostasis in hippocampal neurons and reverses the increased excitatory drive to CA1 neurons in AppNL-G-F mice. AppNL-G-F mice spend less time in rapid eye movement (REM) sleep. AppNL-G-F mice and individuals with AD show progressive changes in morphology of CA1-projecting MCH axons. Our findings identify the MCH system as vulnerable in early AD and suggest that impaired MCH-system function contributes to aberrant excitatory drive and sleep defects, which can compromise hippocampus-dependent functions.
Collapse
Affiliation(s)
- Sara Calafate
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium.
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Gökhan Özturan
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Nicola Thrupp
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Jeroen Vanderlinden
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Luísa Santa-Marinha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rafaela Morais-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Antonella Ruggiero
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ivan Bozic
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| | - Thomas Rusterholz
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Blanca Lorente-Echeverría
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Marcelo Dias
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Wei-Ting Chen
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Mark Fiers
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Ashley Lu
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Ine Vlaeminck
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Eline Creemers
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Katleen Craessaerts
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Joris Vandenbempt
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Luuk van Boekholdt
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
- KU Leuven, Department of Otorhinolaryngology, Leuven, Belgium
| | - Suresh Poovathingal
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Kristofer Davie
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Dietmar Rudolf Thal
- Department of Imaging and Pathology, Laboratory of Neuropathology, and Leuven Brain Institute, KU-Leuven, O&N IV, Leuven, Belgium
- Department of Pathology, UZ Leuven, Leuven, Belgium
| | - Keimpe Wierda
- VIB Center for Brain & Disease Research, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Antoine Adamantidis
- Zentrum für Experimentelle Neurologie, Department of Neurology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Bart De Strooper
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium.
- UK Dementia Research Institute (UK DRI@UCL) at University College London, London, UK.
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium.
| |
Collapse
|
61
|
Ballester Roig MN, Leduc T, Dufort-Gervais J, Maghmoul Y, Tastet O, Mongrain V. Probing pathways by which rhynchophylline modifies sleep using spatial transcriptomics. Biol Direct 2023; 18:21. [PMID: 37143153 PMCID: PMC10161643 DOI: 10.1186/s13062-023-00377-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/12/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Rhynchophylline (RHY) is an alkaloid component of Uncaria, which are plants extensively used in traditional Asian medicines. Uncaria treatments increase sleep time and quality in humans, and RHY induces sleep in rats. However, like many traditional natural treatments, the mechanisms of action of RHY and Uncaria remain evasive. Moreover, it is unknown whether RHY modifies key brain oscillations during sleep. We thus aimed at defining the effects of RHY on sleep architecture and oscillations throughout a 24-h cycle, as well as identifying the underlying molecular mechanisms. Mice received systemic RHY injections at two times of the day (beginning and end of the light period), and vigilance states were studied by electrocorticographic recordings. RESULTS RHY enhanced slow wave sleep (SWS) after both injections, suppressed paradoxical sleep (PS) in the light but enhanced PS in the dark period. Furthermore, RHY modified brain oscillations during both wakefulness and SWS (including delta activity dynamics) in a time-dependent manner. Interestingly, most effects were larger in females. A brain spatial transcriptomic analysis showed that RHY modifies the expression of genes linked to cell movement, apoptosis/necrosis, and transcription/translation in a brain region-independent manner, and changes those linked to sleep regulation (e.g., Hcrt, Pmch) in a brain region-specific manner (e.g., in the hypothalamus). CONCLUSIONS The findings provide support to the sleep-inducing effect of RHY, expose the relevance to shape wake/sleep oscillations, and highlight its effects on the transcriptome with a high spatial resolution. The exposed molecular mechanisms underlying the effect of a natural compound should benefit sleep- and brain-related medicine.
Collapse
Affiliation(s)
- Maria Neus Ballester Roig
- Department of Neuroscience, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Center for Advanced Research in Sleep Medicine, Recherche CIUSSS-NIM, Montréal, QC, H4J 1C5, Canada
| | - Tanya Leduc
- Department of Neuroscience, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Center for Advanced Research in Sleep Medicine, Recherche CIUSSS-NIM, Montréal, QC, H4J 1C5, Canada
| | - Julien Dufort-Gervais
- Center for Advanced Research in Sleep Medicine, Recherche CIUSSS-NIM, Montréal, QC, H4J 1C5, Canada
| | - Yousra Maghmoul
- Center for Advanced Research in Sleep Medicine, Recherche CIUSSS-NIM, Montréal, QC, H4J 1C5, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Olivier Tastet
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal, 900 rue St-Denis, Tour Viger, Montréal, QC, H2X 0A9, Canada
| | - Valérie Mongrain
- Department of Neuroscience, Université de Montréal, Montréal, QC, H3T 1J4, Canada.
- Center for Advanced Research in Sleep Medicine, Recherche CIUSSS-NIM, Montréal, QC, H4J 1C5, Canada.
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal, 900 rue St-Denis, Tour Viger, Montréal, QC, H2X 0A9, Canada.
| |
Collapse
|
62
|
Maxwell ND, Smiley CE, Sadek AT, Loyo-Rosado FZ, Giles DC, Macht VA, Woodruff JL, Taylor DL, Wilson SP, Fadel JR, Reagan LP, Grillo CA. Leptin activation of dorsal raphe neurons inhibits feeding behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.24.538086. [PMID: 37162932 PMCID: PMC10168215 DOI: 10.1101/2023.04.24.538086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Leptin is a homeostatic regulatory element that signals the presence of energy stores -in the form of adipocytes-which ultimately reduces food intake and increases energy expenditure. Similarly, serotonin (5-HT), a signaling molecule found in both the central and peripheral nervous systems, also regulates food intake. Here we use a combination of pharmacological manipulations, optogenetics, retrograde tracing, and in situ hybridization, combined with behavioral endpoints to physiologically and anatomically identify a novel leptin-mediated pathway between 5-HT neurons in the dorsal raphe nucleus (DRN) and hypothalamic arcuate nucleus (ARC) that controls food intake. In this study, we show that microinjecting leptin directly into the DRN reduces food intake in male Sprague-Dawley rats. This effect is mediated by leptin-receptor expressing neurons in the DRN as selective optogenetic activation of these neurons at either their ARC terminals or DRN cell bodies also reduces food intake. Anatomically, we identified a unique population of serotonergic raphe neurons expressing leptin receptors that send projections to the ARC. Finally, by utilizing in vivo microdialysis and high-performance liquid chromatography, we show that leptin administration to the DRN increases 5-HT efflux into the ARC. Overall, this study identifies a novel circuit for leptin-mediated control of food intake through a DRN-ARC pathway, utilizing 5-HT as a mechanism to control feeding behavior. Characterization of this new pathway creates opportunities for understanding how the brain controls eating behavior, as well as opens alternative routes for the treatment of eating disorders. Significance Leptin and serotonin both play a vital role in the regulation of food intake, yet there is still uncertainty in how these two molecules interact to control appetite. The purpose of this study is to further understand the anatomical and functional connections between leptin receptor expressing neurons in the dorsal raphe nucleus, the main source of serotonin, and the arcuate nucleus of the hypothalamus, and how serotonin plays a role in this pathway to reduce food intake. Insight gained from this study will contribute to a more thorough understanding of the networks that regulate food intake, and open alternative avenues for the development of treatments for obesity and eating disorders.
Collapse
|
63
|
Hung C, Yamanaka A. The role of orexin neuron activity in sleep/wakefulness regulation. Peptides 2023; 165:171007. [PMID: 37030519 DOI: 10.1016/j.peptides.2023.171007] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 04/10/2023]
Abstract
Orexin (also known as hypocretin) is a neuropeptide exclusively synthesized in the neurons of the lateral hypothalamus (LH). Initially orexin was thought to be involved in the regulation of feeding behavior. However, it is now known to also be a critical regulator of sleep/wakefulness, especially the maintenance of wakefulness. Although the somas of orexin neurons are exclusively located in the LH, these neurons send axons throughout the brain and spinal cord. Orexin neurons integrate inputs from various brain regions and project to neurons that are involved in the regulation of sleep/wakefulness. Orexin knockout mice have a fragmentation of sleep/wakefulness and cataplexy-like behavior arrest, which is similar to the sleep disorder narcolepsy. Recent progress with manipulation of neural activity of targeted neurons, using experimental tools such as optogenetics and chemogenetics, has emphasized the role of orexin neuron activity on the regulation of sleep/wakefulness. Recording of orexin neuron activity in vivo using electrophysiological and gene-encoded calcium indicator proteins revealed that these cells have specific activity patterns across sleep/wakefulness state changes. Here, we also discuss not only the role of the orexin peptide, but also the role of other co-transmitters that are synthesized and released from orexin neurons and involved in sleep/wakefulness regulation.
Collapse
Affiliation(s)
- Chijung Hung
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Akihiro Yamanaka
- Chinese Institute for Brain Research, Beijing (CIBR), Beijing, 102206, China; National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8585 Japan; Division of Brain Sciences Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| |
Collapse
|
64
|
Ten-Blanco M, Flores Á, Cristino L, Pereda-Pérez I, Berrendero F. Targeting the orexin/hypocretin system for the treatment of neuropsychiatric and neurodegenerative diseases: from animal to clinical studies. Front Neuroendocrinol 2023; 69:101066. [PMID: 37015302 DOI: 10.1016/j.yfrne.2023.101066] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/15/2023] [Accepted: 03/30/2023] [Indexed: 04/06/2023]
Abstract
Orexins (also known as hypocretins) are neuropeptides located exclusively in hypothalamic neurons that have extensive projections throughout the central nervous system and bind two different G protein-coupled receptors (OX1R and OX2R). Since its discovery in 1998, the orexin system has gained the interest of the scientific community as a potential therapeutic target for the treatment of different pathological conditions. Considering previous basic science research, a dual orexin receptor antagonist, suvorexant, was the first orexin agent to be approved by the US Food and Drug Administration to treat insomnia. In this review, we discuss and update the main preclinical and human studies involving the orexin system with several psychiatric and neurodegenerative diseases. This system constitutes a nice example of how basic scientific research driven by curiosity can be the best route to the generation of new and powerful pharmacological treatments.
Collapse
Affiliation(s)
- Marc Ten-Blanco
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - África Flores
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Neurosciences Institute, University of Barcelona and Bellvitge University Hospital-IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Luigia Cristino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy
| | - Inmaculada Pereda-Pérez
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Fernando Berrendero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Pozuelo de Alarcón, Madrid, Spain.
| |
Collapse
|
65
|
Kataoka M, Terakita A. Recent advances in optogenetics: Report for the session 12 at the 19th International Conference on Retinal Proteins. Biophys Physicobiol 2023; 20:e201002. [PMID: 38362320 PMCID: PMC10865861 DOI: 10.2142/biophysico.bppb-v20.s002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Affiliation(s)
- Mikio Kataoka
- Nara Institute of Science and Technology, Ikoma, Nara 630-0182, Japan
| | - Akihisa Terakita
- Osaka Metropolitan University, Sumiyoshi-ku, Osaka 558-8585, Japan
| |
Collapse
|
66
|
Ma C, Zhou N, Ma K, Niu J, Mi T, He Z, Wen Y, Liu C, He Z, Niu J. Neural pathways from hypothalamic orexin neurons to the ventrolateral preoptic area mediate sleep impairments induced by conditioned fear. Front Neurosci 2023; 17:1122803. [PMID: 36998723 PMCID: PMC10043189 DOI: 10.3389/fnins.2023.1122803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
IntroductionFear and sleep impairments common co-exist, but the underlying mechanisms remain unclear. Hypothalamic orexinergic neurons are involved in the regulation of sleep-wake and fear expression. The ventrolateral preoptic area (VLPO) is an essential brain region to promote sleep, and orexinergic axonal fibers projecting to the VLPO are involved in the maintenance of sleep-wake. Neural pathways from hypothalamic orexin neurons to the VLPO might mediate sleep impairments induced by conditioned fear.MethodsTo verify above hypothesis, electroencephalogram (EEG) and electromyogram (EMG) were recorded for analysis of sleep-wake states before and 24 h after conditioned fear training. The retrograde tracing technique and immunofluorescence staining was used to identify the projections from the hypothalamic orexin neurons to the VLPO and to observe their activation in mice with conditioned fear. Moreover, optogenetic activation or inhibition of hypothalamic orexin-VLPO pathways was performed to observe whether the sleep-wake can be regulated in mice with conditioned fear. Finally, orexin-A and orexin receptor antagonist was administered into the VLPO to certify the function of hypothalamic orexin-VLPO pathways on mediating sleep impairments induced by conditioned fear.ResultsIt was found that there was a significant decrease in the non-rapid eye movement (NREM) and rapid eye movement (REM) sleep time and a significant increase in the wakefulness time in mice with conditioned fear. The results of retrograde tracing technique and immunofluorescence staining showed that hypothalamic orexin neurons projected to the VLPO and observed the CTB labeled orexin neurons were significantly activated (c-Fos+) in the hypothalamus in mice with conditioned fear. Optogenetic activation of hypothalamic orexin to the VLPO neural pathways significantly decreased NREM and REM sleep time and increased wakefulness time in mice with conditioned fear. A significant decrease in NREM and REM sleep time and an increase in wakefulness time were observed after the injection of orexin-A into the VLPO, and the effects of orexin-A in the VLPO were blocked by a pre-administrated dual orexin antagonist (DORA).ConclusionThese findings suggest that the neural pathways from hypothalamic orexinergic neurons to the VLPO mediate sleep impairments induced by conditioned fear.
Collapse
Affiliation(s)
- Caifen Ma
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Ning Zhou
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Kang Ma
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Jiandong Niu
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Ting Mi
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Zhenquan He
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Yujun Wen
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Chunhong Liu
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
- Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zhongyi He
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- *Correspondence: Zhongyi He,
| | - Jianguo Niu
- Department of Human Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
- Jianguo Niu,
| |
Collapse
|
67
|
Shin A, Park S, Shin W, Woo J, Jeong M, Kim J, Kim D. A brainstem-to-mediodorsal thalamic pathway mediates sound-induced arousal from slow-wave sleep. Curr Biol 2023; 33:875-885.e5. [PMID: 36754050 DOI: 10.1016/j.cub.2023.01.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 10/25/2022] [Accepted: 01/17/2023] [Indexed: 02/09/2023]
Abstract
Auditory-induced arousal is a defense mechanism of animals against potential dangers. Although the thalamus is the neural substrate that relays sensory information to the cortex, its function is reduced during slow-wave sleep (SWS), also known as deep sleep. Despite this, animals are capable of waking up in response to external sensory stimuli, suggesting the existence of neural circuits that are involved in this response. Here, we report that kainate-class-type ionotropic glutamate receptor subunit 4 (GRIK4)-positive mediodorsal (MD) thalamic neurons act as a neural substrate for arousals from SWS. These neurons become active during arousal from SWS and their photoactivation can induce arousal from SWS. Moreover, we show that these neurons are influenced by glutamatergic neurons in the brainstem, the activity of which increases during auditory-induced arousals. These results suggest that this brainstem-MD pathway can mediate wakefulness from SWS.
Collapse
Affiliation(s)
- Anna Shin
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Seahyung Park
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Wooyeon Shin
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Jeonghoon Woo
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Minju Jeong
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jeongjin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea.
| | - Daesoo Kim
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
68
|
Cre-dependent ACR2-expressing reporter mouse strain for efficient long-lasting inhibition of neuronal activity. Sci Rep 2023; 13:3966. [PMID: 36894577 PMCID: PMC9998869 DOI: 10.1038/s41598-023-30907-2] [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: 12/14/2022] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Optogenetics is a powerful tool for manipulating neuronal activity by light illumination with high temporal and spatial resolution. Anion-channelrhodopsins (ACRs) are light-gated anion channels that allow researchers to efficiently inhibit neuronal activity. A blue light-sensitive ACR2 has recently been used in several in vivo studies; however, the reporter mouse strain expressing ACR2 has not yet been reported. Here, we generated a new reporter mouse strain, LSL-ACR2, in which ACR2 is expressed under the control of Cre recombinase. We crossed this strain with a noradrenergic neuron-specific driver mouse (NAT-Cre) to generate NAT-ACR2 mice. We confirmed Cre-dependent expression and function of ACR2 in the targeted neurons by immunohistochemistry and electrophysiological recordings in vitro, and confirmed physiological function using an in vivo behavioral experiment. Our results show that the LSL-ACR2 mouse strain can be applied for optogenetic inhibition of targeted neurons, particularly for long-lasting continuous inhibition, upon crossing with Cre-driver mouse strains. The LSL-ACR2 strain can be used to prepare transgenic mice with homogenous expression of ACR2 in targeted neurons with a high penetration ratio, good reproducibility, and no tissue invasion.
Collapse
|
69
|
Krohn F, Novello M, van der Giessen RS, De Zeeuw CI, Pel JJM, Bosman LWJ. The integrated brain network that controls respiration. eLife 2023; 12:83654. [PMID: 36884287 PMCID: PMC9995121 DOI: 10.7554/elife.83654] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/29/2023] [Indexed: 03/09/2023] Open
Abstract
Respiration is a brain function on which our lives essentially depend. Control of respiration ensures that the frequency and depth of breathing adapt continuously to metabolic needs. In addition, the respiratory control network of the brain has to organize muscular synergies that integrate ventilation with posture and body movement. Finally, respiration is coupled to cardiovascular function and emotion. Here, we argue that the brain can handle this all by integrating a brainstem central pattern generator circuit in a larger network that also comprises the cerebellum. Although currently not generally recognized as a respiratory control center, the cerebellum is well known for its coordinating and modulating role in motor behavior, as well as for its role in the autonomic nervous system. In this review, we discuss the role of brain regions involved in the control of respiration, and their anatomical and functional interactions. We discuss how sensory feedback can result in adaptation of respiration, and how these mechanisms can be compromised by various neurological and psychological disorders. Finally, we demonstrate how the respiratory pattern generators are part of a larger and integrated network of respiratory brain regions.
Collapse
Affiliation(s)
- Friedrich Krohn
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Manuele Novello
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands.,Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Johan J M Pel
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | |
Collapse
|
70
|
Zheng D, Pisano F, Collard L, Balena A, Pisanello M, Spagnolo B, Mach-Batlle R, Tantussi F, Carbone L, De Angelis F, Valiente M, de la Prida LM, Ciracì C, De Vittorio M, Pisanello F. Toward Plasmonic Neural Probes: SERS Detection of Neurotransmitters through Gold-Nanoislands-Decorated Tapered Optical Fibers with Sub-10 nm Gaps. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2200902. [PMID: 36479741 DOI: 10.1002/adma.202200902] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 10/21/2022] [Indexed: 06/17/2023]
Abstract
Integration of plasmonic nanostructures with fiber-optics-based neural probes enables label-free detection of molecular fingerprints via surface-enhanced Raman spectroscopy (SERS), and it represents a fascinating technological horizon to investigate brain function. However, developing neuroplasmonic probes that can interface with deep brain regions with minimal invasiveness while providing the sensitivity to detect biomolecular signatures in a physiological environment is challenging, in particular because the same waveguide must be employed for both delivering excitation light and collecting the resulting scattered photons. Here, a SERS-active neural probe based on a tapered optical fiber (TF) decorated with gold nanoislands (NIs) that can detect neurotransmitters down to the micromolar range is presented. To do this, a novel, nonplanar repeated dewetting technique to fabricate gold NIs with sub-10 nm gaps, uniformly distributed on the wide (square millimeter scale in surface area), highly curved surface of TF is developed. It is experimentally and numerically shown that the amplified broadband near-field enhancement of the high-density NIs layer allows for achieving a limit of detection in aqueous solution of 10-7 m for rhodamine 6G and 10-5 m for serotonin and dopamine through SERS at near-infrared wavelengths. The NIs-TF technology is envisioned as a first step toward the unexplored frontier of in vivo label-free plasmonic neural interfaces.
Collapse
Affiliation(s)
- Di Zheng
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Filippo Pisano
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Liam Collard
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Antonio Balena
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Marco Pisanello
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Barbara Spagnolo
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Rosa Mach-Batlle
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Francesco Tantussi
- Istituto Italiano di Tecnologia, Center for Convergent Technologies, Genova, 16163, Italy
| | - Luigi Carbone
- CNR NANOTEC - Institute of Nanotechnology, University of Salento, Lecce, 73100, Italy
| | - Francesco De Angelis
- Istituto Italiano di Tecnologia, Center for Convergent Technologies, Genova, 16163, Italy
| | - Manuel Valiente
- Brain Metastasis Group, Spanish National Cancer Research Center (CNIO), Madrid, 28029, Spain
| | | | - Cristian Ciracì
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| | - Massimo De Vittorio
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
- Dipartimento di Ingegneria Dell'Innovazione, Università del Salento, Lecce, 73100, Italy
| | - Ferruccio Pisanello
- Istituto Italiano di Tecnologia, Center for Biomolecular Nanotechnologies, Arnesano, LE, 73010, Italy
| |
Collapse
|
71
|
Sex-dependent role of orexin deficiency in feeding behavior and affective state of mice following intermittent access to a Western diet - Implications for binge-like eating behavior. Physiol Behav 2023; 260:114069. [PMID: 36572152 DOI: 10.1016/j.physbeh.2022.114069] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/02/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
Binge eating disorder is a debilitating disease characterized by recurrent episodes of excessive food consumption and associated with psychiatric comorbidities. Despite a growing body of research investigating the neurobiological underpinnings of eating disorders, specific treatments are lacking. Given its fundamental role in feeding behaviors, we investigated the role of the orexin (hypocretin) neuropeptide system in binge-like eating and associated phenotypes. Specifically, we submitted female and male orexin-deficient mice to a paradigm of intermittent access (once weekly for 24 h) to a Western diet (WD) to induce binge-like eating. Additionally, we measured their anxiety-like behavior and plasma corticosterone levels. All mice showed binge-like eating in response to the intermittent WD access, but females did so to a greater extent than males. While orexin deficiency did not affect binge-like eating in this paradigm, we found that female orexin-deficient mice generally weighed more, and they expressed increased hypophagia and stress levels compared to wild-type mice following binge-like eating episodes. These detrimental effects of orexin deficiency were marginal or absent in males. Moreover, male wild-type mice expressed post-binge anxiety, but orexin-deficient mice did not. In conclusion, these results extend our knowledge of orexin's role in dysregulated eating and associated negative affective states, and contribute to the growing body of evidence indicating a sexual dimorphism of the orexin system. Considering that many human disorders, and especially eating disorders, have a strong sex bias, our findings further emphasize the importance of testing both female and male subjects.
Collapse
|
72
|
Amezawa M, Yamamoto N, Nagumo Y, Kutsumura N, Ishikawa Y, Yanagisawa M, Nagase H, Saitoh T. Design and synthesis of novel orexin 2 receptor agonists with a 1,3,5‑trioxazatriquinane skeleton. Bioorg Med Chem Lett 2023; 82:129151. [PMID: 36690040 DOI: 10.1016/j.bmcl.2023.129151] [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: 12/04/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023]
Abstract
A novel series of 1,3,5‑trioxazatriquinane with multiple effective residues (TriMER) derivatives with amino-methylene side chains was designed and synthesized based on the docking-simulation results between orexin receptors (OXRs) and TriMER-type OXR antagonists. In vitro screening against orexin receptors identified six TriMER derivatives with a cis side-chain configuration, and, among these, 20d and 28d showed full agonist activity against OX2R at a concentration of 10 µM. To determine the absolute stereochemistry of these hit compounds, we also conducted the first asymmetric synthesis of a 1,3,5‑trioxazatriquinane skeleton using a Katsuki-Sharpless asymmetric epoxidation as the key reaction and obtained a set of the individual stereoisomers. After evaluating their activity, (+)-20d (EC50 = 3.87 μM for OX2R) and (+)-28d (EC50 = 1.62 μM for OX2R) were determined as eutomers for OX2R agonist activity. Our results provide a new class of skeleton consisting of an (R)-1,3,5‑trioxazatriquinane core with flexible methylene linkers and hydrophobic substituents at the terminals of the side chains via carbamates/sulfonamides as OX2R agonists.
Collapse
Affiliation(s)
- Mao Amezawa
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Naoshi Yamamoto
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yasuyuki Nagumo
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Noriki Kutsumura
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan; International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yukiko Ishikawa
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; R&D Center for Frontiers of Mirai in Policy and Technology (F-MIRAI), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, US
| | - Hiroshi Nagase
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan; International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Tsuyoshi Saitoh
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
73
|
Leon LES, Sillitoe RV. Disrupted sleep in dystonia depends on cerebellar function but not motor symptoms in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.09.527916. [PMID: 36798256 PMCID: PMC9934608 DOI: 10.1101/2023.02.09.527916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Although dystonia is the third most common movement disorder, patients often also experience debilitating nonmotor defects including impaired sleep. The cerebellum is a central component of a "dystonia network" that plays various roles in sleep regulation. Importantly, the primary driver of sleep impairments in dystonia remains poorly understood. The cerebellum, along with other nodes in the motor circuit, could disrupt sleep. However, it is unclear how the cerebellum might alter sleep and mobility. To disentangle the impact of cerebellar dysfunction on motion and sleep, we generated two mouse genetic models of dystonia that have overlapping cerebellar circuit miswiring but show differing motor phenotype severity: Ptf1a Cre ;Vglut2 fx/fx and Pdx1 Cre ;Vglut2 fx/fx mice. In both models, excitatory climbing fiber to Purkinje cell neurotransmission is blocked, but only the Ptf1a Cre ;Vglut2 fx/fx mice have severe twisting. Using in vivo ECoG and EMG recordings we found that both mutants spend greater time awake and in NREM sleep at the expense of REM sleep. The increase in awake time is driven by longer awake bouts rather than an increase in bout number. We also found a longer latency to reach REM in both mutants, which is similar to what is reported in human dystonia. We uncovered independent but parallel roles for cerebellar circuit dysfunction and motor defects in promoting sleep quality versus posture impairments in dystonia.
Collapse
|
74
|
Sharma A, Tripathi V, Kumar V. Hypothalamic molecular correlates of photoperiod-induced spring migration in intact and castrated male redheaded buntings. Mol Cell Endocrinol 2023; 561:111829. [PMID: 36526025 DOI: 10.1016/j.mce.2022.111829] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
This study investigated the molecular changes associated with neural plasticity in photoperiodic induction of spring migration in intact and castrated redheaded bunting, Emberiza bruniceps. We measured the hypothalamic mRNA expression of genes in birds that were photostimulated into winter non-migratory and spring (vernal) migratory phenotypes under short and long photoperiods, respectively. These included genes associated with the appetitive phase of reproduction (spring migration drive, th and ddc genes encoding for tyrosine hydroxylase and dopamine decarboxylase enzymes, respectively), sleep/awake state (pmch gene encoding for pro-melanin concentrating hormone; hcrt and hcrtr2 encoding for the hypocretin/orexin and its receptor, respectively) and neurogenesis (dcx and neuN coding for doublecortin and neuronal nuclear proteins, respectively). Higher th mRNA levels suggested an upregulated dopamine synthesis in the hypothalamus of spring migrants. Similarly, elevated hcrt and hcrtr2 mRNA levels suggested an increased wakefulness, and those of dcx and neuN genes suggested an enhanced neurogenesis during the spring migration state. Further, compared to intact birds, the lower th and pmch, and higher hcrtr2 and neuN mRNA levels in castrates suggested a role of testicular steroids in modulation of the appetitive phase of reproduction, sleep and awake states, and neurogenesis during spring migration period. These results provide insights into molecular changes linked with important hypothalamic molecular pathways and steroidal influence in the photoperiodic induction of spring migration in obligate migratory songbirds.
Collapse
Affiliation(s)
- Aakansha Sharma
- IndoUS Center in Chronobiology, Department of Zoology, University of Delhi, Delhi, 110007, India.
| | - Vatsala Tripathi
- Department of Zoology, Dyal Singh College, University of Delhi, Delhi, 110003, India.
| | - Vinod Kumar
- IndoUS Center in Chronobiology, Department of Zoology, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
75
|
Sulaman BA, Wang S, Tyan J, Eban-Rothschild A. Neuro-orchestration of sleep and wakefulness. Nat Neurosci 2023; 26:196-212. [PMID: 36581730 DOI: 10.1038/s41593-022-01236-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/16/2022] [Indexed: 12/31/2022]
Abstract
Although considered an inactive state for centuries, sleep entails many active processes occurring at the cellular, circuit and organismal levels. Over the last decade, several key technological advances, including calcium imaging and optogenetic and chemogenetic manipulations, have facilitated a detailed understanding of the functions of different neuronal populations and circuits in sleep-wake regulation. Here, we present recent progress and summarize our current understanding of the circuitry underlying the initiation, maintenance and coordination of wakefulness, rapid eye movement sleep (REMS) and non-REMS (NREMS). We propose a de-arousal model for sleep initiation, in which the neuromodulatory milieu necessary for sleep initiation is achieved by engaging in repetitive pre-sleep behaviors that gradually reduce vigilance to the external environment and wake-promoting neuromodulatory tone. We also discuss how brain processes related to thermoregulation, hunger and fear intersect with sleep-wake circuits to control arousal. Lastly, we discuss controversies and lingering questions in the sleep field.
Collapse
Affiliation(s)
- Bibi A Sulaman
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Su Wang
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Jean Tyan
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
76
|
Grady FS, Graff SA, Resch JM, Geerling JC. Parabrachial-insular stimulation does not wake mice. J Neurophysiol 2023; 129:347-355. [PMID: 36542422 PMCID: PMC9886350 DOI: 10.1152/jn.00318.2022] [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: 07/26/2022] [Revised: 11/28/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
The parabrachial nucleus (PB) in the upper brainstem receives interoceptive information and sends a massive output projection directly to the cerebral cortex. Its glutamatergic axons primarily target the midinsular cortex, and we have proposed that this PB-insular projection promotes arousal. Here, we test whether stimulating this projection causes wakefulness. We combined optogenetics and video-electroencephalography (vEEG) in mice to test this hypothesis by stimulating PB axons in the insular cortex. Stimulating this projection did not alter the cortical EEG or awaken mice. Also, despite a tendency toward aversion, PB-insular stimulation did not significantly alter real-time place preference (RTPP). These results are not consistent with the hypothesis that the direct PB-insular projection is part of the ascending arousal system.NEW & NOTEWORTHY A brainstem region critical for wakefulness overlaps the medial parabrachial nucleus (PB) and has functional and direct axonal connectivity with the insular cortex. In this study, we hypothesized that this direct projection from the PB to the insular cortex promotes arousal. However, photostimulating PB axons in the insular cortex did not alter the cortical EEG or awaken mice. This information constrains the possible circuit connections through which brainstem neurons may sustain arousal.
Collapse
Affiliation(s)
- Fillan S Grady
- Department of Neurology, University of Iowa, Iowa City, Iowa
| | | | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa
| | - Joel C Geerling
- Department of Neurology, University of Iowa, Iowa City, Iowa
| |
Collapse
|
77
|
Mogavero MP, Silvani A, Lanza G, DelRosso LM, Ferini-Strambi L, Ferri R. Targeting Orexin Receptors for the Treatment of Insomnia: From Physiological Mechanisms to Current Clinical Evidence and Recommendations. Nat Sci Sleep 2023; 15:17-38. [PMID: 36713640 PMCID: PMC9879039 DOI: 10.2147/nss.s201994] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/08/2023] [Indexed: 01/23/2023] Open
Abstract
After a detailed description of orexins and their roles in sleep and other medical disorders, we discuss here the current clinical evidence on the effects of dual (DORAs) or selective (SORAs) orexin receptor antagonists on insomnia with the aim to provide recommendations for their further assessment in a context of personalized and precision medicine. In the last decade, many trials have been conducted with orexin receptor antagonists, which represent an innovative and valid therapeutic option based on the multiple mechanisms of action of orexins on different biological circuits, both centrally and peripherally, and their role in a wide range of medical conditions which are often associated with insomnia. A very interesting aspect of this new category of drugs is that they have limited abuse liability and their discontinuation does not seem associated with significant rebound effects. Further studies on the efficacy of DORAs are required, especially on children and adolescents and in particular conditions, such as menopause. Which DORA is most suitable for each patient, based on comorbidities and/or concomitant treatments, should be the focus of further careful research. On the contrary, studies on SORAs, some of which seem to be appropriate also in insomnia in patients with psychiatric diseases, are still at an early stage and, therefore, do not allow to draw definite conclusions.
Collapse
Affiliation(s)
- Maria P Mogavero
- Vita-Salute San Raffaele University, Milan, Italy
- Sleep Disorders Center, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Silvani
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe Lanza
- Sleep Research Centre, Oasi Research Institute - IRCCS, Troina, Italy
- Department of Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Lourdes M DelRosso
- Pulmonary and Sleep Medicine, University of California San Francisco-Fresno, Fresno, CA, USA
| | - Luigi Ferini-Strambi
- Vita-Salute San Raffaele University, Milan, Italy
- Sleep Disorders Center, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Raffaele Ferri
- Sleep Research Centre, Oasi Research Institute - IRCCS, Troina, Italy
| |
Collapse
|
78
|
MacRae CA, Peterson RT. Zebrafish as a Mainstream Model for In Vivo Systems Pharmacology and Toxicology. Annu Rev Pharmacol Toxicol 2023; 63:43-64. [PMID: 36151053 DOI: 10.1146/annurev-pharmtox-051421-105617] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Pharmacology and toxicology are part of a much broader effort to understand the relationship between chemistry and biology. While biomedicine has necessarily focused on specific cases, typically of direct human relevance, there are real advantages in pursuing more systematic approaches to characterizing how health and disease are influenced by small molecules and other interventions. In this context, the zebrafish is now established as the representative screenable vertebrate and, through ongoing advances in the available scale of genome editing and automated phenotyping, is beginning to address systems-level solutions to some biomedical problems. The addition of broader efforts to integrate information content across preclinical model organisms and the incorporation of rigorous analytics, including closed-loop deep learning, will facilitate efforts to create systems pharmacology and toxicology with the ability to continuously optimize chemical biological interactions around societal needs. In this review, we outline progress toward this goal.
Collapse
Affiliation(s)
- Calum A MacRae
- Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA;
| | | |
Collapse
|
79
|
Tyree SM, Jennings KJ, Gonzalez OC, Li SB, Nicholson JR, von Heimendahl M, de Lecea L. Optogenetic and pharmacological interventions link hypocretin neurons to impulsivity in mice. Commun Biol 2023; 6:74. [PMID: 36658362 PMCID: PMC9852239 DOI: 10.1038/s42003-023-04409-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 01/03/2023] [Indexed: 01/20/2023] Open
Abstract
Neurons in the lateral hypothalamus expressing the neuropeptide Hypocretin, also known as orexin, are known critical modulators of arousal stability. However, their role in the different components of the arousal construct such as attention and decision making is poorly understood. Here we study Hypocretin neuronal circuit dynamics during stop action impulsivity in a Go/NoGo task in mice. We show that Hypocretin neuronal activity correlates with anticipation of reward. We then assessed the causal role of Hypocretin neuronal activity using optogenetics in a Go/NoGo task. We show that stimulation of Hypocretin neurons during the cue period dramatically increases the number of premature responses. These effects are mimicked by amphetamine, reduced by atomoxetine, a norepinephrine uptake inhibitor, and blocked by a Hypocretin receptor 1 selective antagonist. We conclude that Hypocretin neurons have a key role in the integration of salient stimuli during wakefulness to produce appropriate and timely responses to rewarding and aversive cues.
Collapse
Affiliation(s)
- Susan M. Tyree
- grid.168010.e0000000419368956Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford, CA USA ,Present Address: Atlantia Clinical Trials, Cork, Ireland
| | - Kimberly J. Jennings
- grid.168010.e0000000419368956Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford, CA USA ,grid.55460.320000000121548364Present Address: University of Texas, Austin, TX USA
| | - Oscar C. Gonzalez
- grid.168010.e0000000419368956Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford, CA USA
| | - Shi-bin Li
- grid.168010.e0000000419368956Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford, CA USA
| | - Janet R. Nicholson
- grid.420061.10000 0001 2171 7500Boehringer Ingelheim Pharma GmbH & Co.KG, Biberach, Germany
| | - Moritz von Heimendahl
- grid.420061.10000 0001 2171 7500Boehringer Ingelheim Pharma GmbH & Co.KG, Biberach, Germany
| | - Luis de Lecea
- grid.168010.e0000000419368956Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford, CA USA
| |
Collapse
|
80
|
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: 0] [Impact Index Per Article: 0] [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
|
81
|
Zhao YN, Jiang JB, Tao SY, Zhang Y, Chen ZK, Qu WM, Huang ZL, Yang SR. GABAergic neurons in the rostromedial tegmental nucleus are essential for rapid eye movement sleep suppression. Nat Commun 2022; 13:7552. [PMID: 36477665 PMCID: PMC9729601 DOI: 10.1038/s41467-022-35299-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Rapid eye movement (REM) sleep disturbances are prevalent in various psychiatric disorders. However, the neural circuits that regulate REM sleep remain poorly understood. Here, we found that in male mice, optogenetic activation of rostromedial tegmental nucleus (RMTg) GABAergic neurons immediately converted REM sleep to arousal and then initiated non-REM (NREM) sleep. Conversely, laser-mediated inactivation completely converted NREM to REM sleep and prolonged REM sleep duration. The activity of RMTg GABAergic neurons increased to a high discharge level at the termination of REM sleep. RMTg GABAergic neurons directly converted REM sleep to wakefulness and NREM sleep via inhibitory projections to the laterodorsal tegmentum (LDT) and lateral hypothalamus (LH), respectively. Furthermore, LDT glutamatergic neurons were responsible for the REM sleep-wake transitions following photostimulation of the RMTgGABA-LDT circuit. Thus, RMTg GABAergic neurons are essential for suppressing the induction and maintenance of REM sleep.
Collapse
Affiliation(s)
- Ya-Nan Zhao
- grid.8547.e0000 0001 0125 2443Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science; Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| | - Jian-Bo Jiang
- grid.8547.e0000 0001 0125 2443Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science; Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| | - Shi-Yuan Tao
- grid.8547.e0000 0001 0125 2443Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science; Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| | - Yang Zhang
- grid.8547.e0000 0001 0125 2443Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science; Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| | - Ze-Ka Chen
- grid.8547.e0000 0001 0125 2443Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science; Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| | - Wei-Min Qu
- grid.8547.e0000 0001 0125 2443Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science; Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| | - Zhi-Li Huang
- grid.8547.e0000 0001 0125 2443Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science; Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| | - Su-Rong Yang
- grid.8547.e0000 0001 0125 2443Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science; Institutes of Brain Science, Fudan University, Shanghai, 200032 China
| |
Collapse
|
82
|
Bigalke JA, Shan Z, Carter JR. Orexin, Sleep, Sympathetic Neural Activity, and Cardiovascular Function. Hypertension 2022; 79:2643-2655. [PMID: 36148653 PMCID: PMC9649879 DOI: 10.1161/hypertensionaha.122.19796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Inadequate sleep duration and quality are associated with reduced cardiovascular health and increased mortality. Experimental evidence points to the sympathetic nervous system as a key mediator in the observed relationship between poor sleep and cardiovascular dysfunction. However, brain mechanisms underpinning the impaired sympathetic function associated with poor sleep remain unclear. Recent evidence suggests the central orexin system, particularly orexins A and B and their receptors, have a key regulatory role for sleep in animal and human models. While orexin system activity has been observed to significantly impact sympathetic regulation in animals, the extension of these findings to humans has been difficult due to an inability to directly assess orexin system activity in humans. However, direct measures of sympathetic activity in populations with narcolepsy and chronic insomnia, 2 sleep disorders associated with deficient and excessive orexin neural activity, have allowed indirect assessment of the relationships between orexin, sleep, and sympathetic regulation. Further, the recent pharmaceutical development of dual orexin receptor antagonists for use in clinical insomnia populations offers an unprecedented opportunity to examine the mechanistic role of orexin in sleep and cardiovascular health in humans. The current review assesses the role of orexin in both sleep and sympathetic regulation from a translational perspective, spanning animal and human studies. The review concludes with future research directions necessary to fully elucidate the mechanistic role for orexin in sleep and sympathetic regulation in humans.
Collapse
Affiliation(s)
- Jeremy A. Bigalke
- Department of Health and Human Development, Montana State University, Bozeman, Montana
- Department of Psychology, Montana State University, Bozeman, Montana
| | - Zhiying Shan
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, Michigan
| | - Jason R. Carter
- Department of Health and Human Development, Montana State University, Bozeman, Montana
- Department of Psychology, Montana State University, Bozeman, Montana
| |
Collapse
|
83
|
Mutlu-Burnaz O, Yulug B, Oncul M, Celik E, Atasoy NS, Cankaya S, Hanoglu L, Velioglu HA. Chemogenetic inhibition of MCH neurons does not alter memory performance in mice. Biomed Pharmacother 2022; 155:113771. [DOI: 10.1016/j.biopha.2022.113771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
|
84
|
Jung J, Kim T. General anesthesia and sleep: like and unlike. Anesth Pain Med (Seoul) 2022; 17:343-351. [DOI: 10.17085/apm.22227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
General anesthesia and sleep have long been discussed in the neurobiological context owingto their commonalities, such as unconsciousness, immobility, non-responsiveness to externalstimuli, and lack of memory upon returning to consciousness. Sleep is regulated bycomplex interactions between wake-promoting and sleep-promoting neural circuits. Anestheticsexert their effects partly by inhibiting wake-promoting neurons or activating sleep-promotingneurons. Unconscious but arousable sedation is more related to sleep-wake circuitries,whereas unconscious and unarousable anesthesia is independent of them. Generalanesthesia is notable for its ability to decrease sleep propensity. Conversely, increasedsleep propensity due to insufficient sleep potentiates anesthetic effects. Taken together, it isplausible that sleep and anesthesia are closely related phenomena but not the same ones.Further investigations on the relationship between sleep and anesthesia are warranted.
Collapse
|
85
|
Stüdemann T, Rössinger J, Manthey C, Geertz B, Srikantharajah R, von Bibra C, Shibamiya A, Köhne M, Wiehler A, Wiegert JS, Eschenhagen T, Weinberger F. Contractile Force of Transplanted Cardiomyocytes Actively Supports Heart Function After Injury. Circulation 2022; 146:1159-1169. [PMID: 36073365 PMCID: PMC9555755 DOI: 10.1161/circulationaha.122.060124] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Transplantation of pluripotent stem cell-derived cardiomyocytes represents a promising therapeutic strategy for cardiac regeneration, and the first clinical studies in patients with heart failure have commenced. Yet, little is known about the mechanism of action underlying graft-induced benefits. Here, we explored whether transplanted cardiomyocytes actively contribute to heart function. METHODS We injected cardiomyocytes with an optogenetic off-on switch in a guinea pig cardiac injury model. RESULTS Light-induced inhibition of engrafted cardiomyocyte contractility resulted in a rapid decrease of left ventricular function in ≈50% (7/13) animals that was fully reversible with the offset of photostimulation. CONCLUSIONS Our optogenetic approach demonstrates that transplanted cardiomyocytes can actively participate in heart function, supporting the hypothesis that the delivery of new force-generating myocardium can serve as a regenerative therapeutic strategy.
Collapse
Affiliation(s)
- Tim Stüdemann
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lubeck, Germany (T.S., J.R., C.M., R.S., C.v.B., A.S., M.K., T.E., F.W.)
| | - Judith Rössinger
- Department of Experimental Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Germany (T.S., J.R., C.M., B.G., R.S., C.v.B., A.S., M.K., T.E., F.W.).,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lubeck, Germany (T.S., J.R., C.M., R.S., C.v.B., A.S., M.K., T.E., F.W.)
| | - Christoph Manthey
- Department of Experimental Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Germany (T.S., J.R., C.M., B.G., R.S., C.v.B., A.S., M.K., T.E., F.W.).,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lubeck, Germany (T.S., J.R., C.M., R.S., C.v.B., A.S., M.K., T.E., F.W.)
| | - Birgit Geertz
- Department of Experimental Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Germany (T.S., J.R., C.M., B.G., R.S., C.v.B., A.S., M.K., T.E., F.W.)
| | - Rajiven Srikantharajah
- Department of Experimental Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Germany (T.S., J.R., C.M., B.G., R.S., C.v.B., A.S., M.K., T.E., F.W.).,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lubeck, Germany (T.S., J.R., C.M., R.S., C.v.B., A.S., M.K., T.E., F.W.)
| | - Constantin von Bibra
- Department of Experimental Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Germany (T.S., J.R., C.M., B.G., R.S., C.v.B., A.S., M.K., T.E., F.W.).,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lubeck, Germany (T.S., J.R., C.M., R.S., C.v.B., A.S., M.K., T.E., F.W.)
| | - Aya Shibamiya
- Department of Experimental Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Germany (T.S., J.R., C.M., B.G., R.S., C.v.B., A.S., M.K., T.E., F.W.).,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lubeck, Germany (T.S., J.R., C.M., R.S., C.v.B., A.S., M.K., T.E., F.W.)
| | - Maria Köhne
- Department of Experimental Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Germany (T.S., J.R., C.M., B.G., R.S., C.v.B., A.S., M.K., T.E., F.W.).,Surgery for Congenital Heart Disease, University Heart & Vascular Center Hamburg, Germany (M.K.).,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lubeck, Germany (T.S., J.R., C.M., R.S., C.v.B., A.S., M.K., T.E., F.W.)
| | - Antonius Wiehler
- Department of Psychiatry, Service Hospitalo-Universitaire, Groupe Hospitalier Universitaire Paris Psychiatrie & Neurosciences, Universite de Paris, France (A.W.)
| | - J. Simon Wiegert
- Research Group Synaptic Wiring and Information Processing, Centre for Molecular Neurobiology Hamburg, Germany (J.S.W.)
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Germany (T.S., J.R., C.M., B.G., R.S., C.v.B., A.S., M.K., T.E., F.W.).,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lubeck, Germany (T.S., J.R., C.M., R.S., C.v.B., A.S., M.K., T.E., F.W.)
| | - Florian Weinberger
- Department of Experimental Pharmacology and Toxicology, University Medical Centre Hamburg-Eppendorf, Germany (T.S., J.R., C.M., B.G., R.S., C.v.B., A.S., M.K., T.E., F.W.).,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lubeck, Germany (T.S., J.R., C.M., R.S., C.v.B., A.S., M.K., T.E., F.W.)
| |
Collapse
|
86
|
Sylwestrak EL, Jo Y, Vesuna S, Wang X, Holcomb B, Tien RH, Kim DK, Fenno L, Ramakrishnan C, Allen WE, Chen R, Shenoy KV, Sussillo D, Deisseroth K. Cell-type-specific population dynamics of diverse reward computations. Cell 2022; 185:3568-3587.e27. [PMID: 36113428 PMCID: PMC10387374 DOI: 10.1016/j.cell.2022.08.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/16/2022] [Accepted: 08/17/2022] [Indexed: 01/26/2023]
Abstract
Computational analysis of cellular activity has developed largely independently of modern transcriptomic cell typology, but integrating these approaches may be essential for full insight into cellular-level mechanisms underlying brain function and dysfunction. Applying this approach to the habenula (a structure with diverse, intermingled molecular, anatomical, and computational features), we identified encoding of reward-predictive cues and reward outcomes in distinct genetically defined neural populations, including TH+ cells and Tac1+ cells. Data from genetically targeted recordings were used to train an optimized nonlinear dynamical systems model and revealed activity dynamics consistent with a line attractor. High-density, cell-type-specific electrophysiological recordings and optogenetic perturbation provided supporting evidence for this model. Reverse-engineering predicted how Tac1+ cells might integrate reward history, which was complemented by in vivo experimentation. This integrated approach describes a process by which data-driven computational models of population activity can generate and frame actionable hypotheses for cell-type-specific investigation in biological systems.
Collapse
Affiliation(s)
- Emily L Sylwestrak
- Department of Biology, University of Oregon, Eugene, OR 97403, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA.
| | - YoungJu Jo
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Sam Vesuna
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Xiao Wang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Blake Holcomb
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - Rebecca H Tien
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Doo Kyung Kim
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Lief Fenno
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - William E Allen
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Neurosciences Interdepartmental Program, Stanford University, Stanford, CA 94303, USA
| | - Ritchie Chen
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Krishna V Shenoy
- Department of Neurobiology, Stanford University, Stanford, CA 94303, USA; Department of Electrical Engineering, Stanford University, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - David Sussillo
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
87
|
Klaus S, Carolan A, O'Rourke D, Kennedy B. What respiratory physicians should know about narcolepsy and other hypersomnias. Breathe (Sheff) 2022; 18:220157. [PMID: 36865656 PMCID: PMC9973529 DOI: 10.1183/20734735.0157-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Narcolepsy and related central disorders of hypersomnolence may present to the sleep clinic with excessive daytime sleepiness. A strong clinical suspicion and awareness of the diagnostic clues, such as cataplexy, are essential to avoid unnecessary diagnostic delay. This review provides an overview of the epidemiology, pathophysiology, clinical features, diagnostic criteria and management of narcolepsy and related disorders, including idiopathic hypersomnia, Kleine-Levin syndrome (recurrent episodic hypersomnia) and secondary central disorders of hypersomnolence.
Collapse
Affiliation(s)
- Stephen Klaus
- Department of Sleep Medicine, St James's Hospital, Dublin, Ireland
| | - Aoife Carolan
- Department of Sleep Medicine, St James's Hospital, Dublin, Ireland
| | - Deirdre O'Rourke
- Department of Sleep Medicine, St James's Hospital, Dublin, Ireland
| | - Barry Kennedy
- Department of Sleep Medicine, St James's Hospital, Dublin, Ireland,Corresponding author: Barry Kennedy ()
| |
Collapse
|
88
|
Eiden LE, Hernández VS, Jiang SZ, Zhang L. Neuropeptides and small-molecule amine transmitters: cooperative signaling in the nervous system. Cell Mol Life Sci 2022; 79:492. [PMID: 35997826 PMCID: PMC11072502 DOI: 10.1007/s00018-022-04451-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/15/2022] [Accepted: 06/23/2022] [Indexed: 12/17/2022]
Abstract
Neuropeptides are expressed in cell-specific patterns throughout mammalian brain. Neuropeptide gene expression has been useful for clustering neurons by phenotype, based on single-cell transcriptomics, and for defining specific functional circuits throughout the brain. How neuropeptides function as first messengers in inter-neuronal communication, in cooperation with classical small-molecule amine transmitters (SMATs) is a current topic of systems neurobiology. Questions include how neuropeptides and SMATs cooperate in neurotransmission at the molecular, cellular and circuit levels; whether neuropeptides and SMATs always co-exist in neurons; where neuropeptides and SMATs are stored in the neuron, released from the neuron and acting, and at which receptors, after release; and how neuropeptides affect 'classical' transmitter function, both directly upon co-release, and indirectly, via long-term regulation of gene transcription and neuronal plasticity. Here, we review an extensive body of data about the distribution of neuropeptides and their receptors, their actions after neuronal release, and their function based on pharmacological and genetic loss- and gain-of-function experiments, that addresses these questions, fundamental to understanding brain function, and development of neuropeptide-based, and potentially combinatorial peptide/SMAT-based, neurotherapeutics.
Collapse
Affiliation(s)
- Lee E Eiden
- Section On Molecular Neuroscience, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 49 Convent Drive, Room 5A38, Bethesda, MD, 20892, USA.
| | - Vito S Hernández
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Sunny Z Jiang
- Section On Molecular Neuroscience, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, 49 Convent Drive, Room 5A38, Bethesda, MD, 20892, USA
| | - Limei Zhang
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico.
| |
Collapse
|
89
|
Clark MG, Gonzalez GA, Luo Y, Aldana-Mendoza JA, Carlsen MS, Eakins G, Dai M, Zhang C. Real-time precision opto-control of chemical processes in live cells. Nat Commun 2022; 13:4343. [PMID: 35896556 PMCID: PMC9329476 DOI: 10.1038/s41467-022-32071-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 07/15/2022] [Indexed: 11/29/2022] Open
Abstract
Precision control of molecular activities and chemical reactions in live cells is a long-sought capability by life scientists. No existing technology can probe molecular targets in cells and simultaneously control the activities of only these targets at high spatial precision. We develop a real-time precision opto-control (RPOC) technology that detects a chemical-specific optical response from molecular targets during laser scanning and uses the optical signal to couple a separate laser to only interact with these molecules without affecting other sample locations. We demonstrate precision control of molecular states of a photochromic molecule in different regions of the cells. We also synthesize a photoswitchable compound and use it with RPOC to achieve site-specific inhibition of microtubule polymerization and control of organelle dynamics in live cells. RPOC can automatically detect and control biomolecular activities and chemical processes in dynamic living samples with submicron spatial accuracy, fast response time, and high chemical specificity. There is a need to control molecular activities at high spatial precision. Here the authors report a real-time precision opto-control technology that detects a chemical-specific optical response from molecular targets, and precisely control photoswitchable microtubule polymerization inhibitors in cells.
Collapse
Affiliation(s)
- Matthew G Clark
- Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN, 47907, USA
| | - Gil A Gonzalez
- Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN, 47907, USA
| | - Yiyang Luo
- Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN, 47907, USA
| | - Jesus A Aldana-Mendoza
- Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN, 47907, USA
| | - Mark S Carlsen
- Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN, 47907, USA
| | - Gregory Eakins
- Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN, 47907, USA
| | - Mingji Dai
- Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN, 47907, USA.,Purdue Center for Cancer Research, 201 S. University St., West Lafayette, IN, 47907, USA
| | - Chi Zhang
- Department of Chemistry, Purdue University, 560 Oval Dr., West Lafayette, IN, 47907, USA. .,Purdue Center for Cancer Research, 201 S. University St., West Lafayette, IN, 47907, USA. .,Purdue Institute of Inflammation, Immunology, and Infectious Disease, 207 S. Martin Jischke Dr., West Lafayette, IN, 47907, USA.
| |
Collapse
|
90
|
Emiliani V, Entcheva E, Hedrich R, Hegemann P, Konrad KR, Lüscher C, Mahn M, Pan ZH, Sims RR, Vierock J, Yizhar O. Optogenetics for light control of biological systems. NATURE REVIEWS. METHODS PRIMERS 2022; 2:55. [PMID: 37933248 PMCID: PMC10627578 DOI: 10.1038/s43586-022-00136-4] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 11/08/2023]
Abstract
Optogenetic techniques have been developed to allow control over the activity of selected cells within a highly heterogeneous tissue, using a combination of genetic engineering and light. Optogenetics employs natural and engineered photoreceptors, mostly of microbial origin, to be genetically introduced into the cells of interest. As a result, cells that are naturally light-insensitive can be made photosensitive and addressable by illumination and precisely controllable in time and space. The selectivity of expression and subcellular targeting in the host is enabled by applying control elements such as promoters, enhancers and specific targeting sequences to the employed photoreceptor-encoding DNA. This powerful approach allows precise characterization and manipulation of cellular functions and has motivated the development of advanced optical methods for patterned photostimulation. Optogenetics has revolutionized neuroscience during the past 15 years and is primed to have a similar impact in other fields, including cardiology, cell biology and plant sciences. In this Primer, we describe the principles of optogenetics, review the most commonly used optogenetic tools, illumination approaches and scientific applications and discuss the possibilities and limitations associated with optogenetic manipulations across a wide variety of optical techniques, cells, circuits and organisms.
Collapse
Affiliation(s)
- Valentina Emiliani
- Wavefront Engineering Microscopy Group, Photonics Department, Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Rainer Hedrich
- Julius-von-Sachs Institute for Biosciences, Molecular Plant Physiology and Biophysics, University of Wuerzburg, Wuerzburg, Germany
| | - Peter Hegemann
- Institute for Biology, Experimental Biophysics, Humboldt-Universitaet zu Berlin, Berlin, Germany
| | - Kai R. Konrad
- Julius-von-Sachs Institute for Biosciences, Molecular Plant Physiology and Biophysics, University of Wuerzburg, Wuerzburg, Germany
| | - Christian Lüscher
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Clinic of Neurology, Department of Clinical Neurosciences, Geneva University Hospital, Geneva, Switzerland
| | - Mathias Mahn
- Department of Neurobiology, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Zhuo-Hua Pan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ruth R. Sims
- Wavefront Engineering Microscopy Group, Photonics Department, Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Johannes Vierock
- Institute for Biology, Experimental Biophysics, Humboldt-Universitaet zu Berlin, Berlin, Germany
- Neuroscience Research Center, Charité – Universitaetsmedizin Berlin, Berlin, Germany
| | - Ofer Yizhar
- Departments of Brain Sciences and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
91
|
Chavan P, Chikahisa S, Shiuchi T, Shimizu N, Dalanon J, Okura K, Séi H, Matsuka Y. Dual orexin receptor antagonist drug suvorexant can help in amelioration of predictable chronic mild stress-induced hyperalgesia. Brain Res Bull 2022; 188:39-46. [PMID: 35868501 DOI: 10.1016/j.brainresbull.2022.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/07/2022] [Accepted: 07/18/2022] [Indexed: 12/31/2022]
Abstract
AIMS This study aimed to evaluate the involvement of the orexin system in predictable chronic mild stress (PCMS) and the effects of suvorexant, a dual orexin receptor antagonist, on nociceptive behavior in PCMS. MATERIALS AND METHODS Male C57BL/6 J mice were separated into various PCMS groups: a control group with sawdust on the floor of the rearing cage (C), a group with mesh wire on the floor (M), and a group with water just below the mesh wire (W). Activation of lateral hypothalamic orexin neurons was assessed using immunofluorescence. In another experiment, half of the mice in each group were administered an intraperitoneal injection of suvorexant (10 mg/kg), and the remaining mice were injected with the same amount of vehicle (normal saline). Thermal hyperalgesia was examined using tail immersion and hot plate tests, while mechanical hyperalgesia was investigated using the tail pinch test after 21 days of PCMS. KEY FINDINGS Animals subjected to PCMS showed an increased percentage of activated orexin neurons in the lateral hypothalamic region after 21 days. Mice raised in the PCMS environment showed increased pain sensitivity in several pain tests; however, the symptoms were significantly reduced by suvorexant administration. SIGNIFICANCE The findings revealed that PCMS activates hypothalamic orexin neuronal activity, and the use of suvorexant can help attenuate PCMS-induced thermal and mechanical hyperalgesia.
Collapse
Affiliation(s)
- Parimal Chavan
- Department of Stomatognathic Function and Occlusal Reconstruction, Tokushima University Graduate School of Biomedical Sciences, Tokushima City, Japan
| | - Sachiko Chikahisa
- Department of Integrative Physiology, Tokushima University Graduate School of Biomedical Sciences, Tokushima City, Japan; Department of Health and Nutrition, Faculty of Human Life Science, Shikoku University, Tokushima City, Japan.
| | - Tetsuya Shiuchi
- Department of Integrative Physiology, Tokushima University Graduate School of Biomedical Sciences, Tokushima City, Japan
| | - Noriyuki Shimizu
- Department of Integrative Physiology, Tokushima University Graduate School of Biomedical Sciences, Tokushima City, Japan
| | - Junhel Dalanon
- Department of Stomatognathic Function and Occlusal Reconstruction, Tokushima University Graduate School of Biomedical Sciences, Tokushima City, Japan
| | - Kazuo Okura
- Department of Stomatognathic Function and Occlusal Reconstruction, Tokushima University Graduate School of Biomedical Sciences, Tokushima City, Japan
| | - Hiroyoshi Séi
- Department of Integrative Physiology, Tokushima University Graduate School of Biomedical Sciences, Tokushima City, Japan
| | - Yoshizo Matsuka
- Department of Stomatognathic Function and Occlusal Reconstruction, Tokushima University Graduate School of Biomedical Sciences, Tokushima City, Japan
| |
Collapse
|
92
|
Gao XB, Horvath TL. From Molecule to Behavior: Hypocretin/orexin Revisited From a Sex-dependent Perspective. Endocr Rev 2022; 43:743-760. [PMID: 34792130 PMCID: PMC9277634 DOI: 10.1210/endrev/bnab042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Indexed: 11/19/2022]
Abstract
The hypocretin/orexin (Hcrt/Orx) system in the perifornical lateral hypothalamus has been recognized as a critical node in a complex network of neuronal systems controlling both physiology and behavior in vertebrates. Our understanding of the Hcrt/Orx system and its array of functions and actions has grown exponentially in merely 2 decades. This review will examine the latest progress in discerning the roles played by the Hcrt/Orx system in regulating homeostatic functions and in executing instinctive and learned behaviors. Furthermore, the gaps that currently exist in our knowledge of sex-related differences in this field of study are discussed.
Collapse
Affiliation(s)
- Xiao-Bing Gao
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
93
|
Pouliopoulos AN, Murillo MF, Noel RL, Batts AJ, Ji R, Kwon N, Yu H, Tong CK, Gelinas JN, Araghy DK, Hussaini SA, Konofagou EE. Non-invasive optogenetics with ultrasound-mediated gene delivery and red-light excitation. Brain Stimul 2022; 15:927-941. [PMID: 35718324 PMCID: PMC9379392 DOI: 10.1016/j.brs.2022.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/30/2022] [Accepted: 06/11/2022] [Indexed: 12/03/2022] Open
Abstract
Optogenetics has revolutionized the capability of controlling genetically modified neurons in vitro and in vivo and has become an indispensable neuroscience tool. Using light as a probe for selective neuronal activation or inhibition and as a means to read out neural activity has dramatically enhanced our understanding of complex neural circuits. However, a common limitation of optogenetic studies to date is their invasiveness and spatiotemporal range. Direct viral injections into the brain tissue along with implantation of optical fibers and recording electrodes can disrupt the neuronal circuitry and cause significant damage. Conventional approaches are spatially limited around the site of the direct injection and insufficient in examining large networks throughout the brain. Lastly, optogenetics is currently not easily scalable to large animals or humans. Here, we demonstrate that optogenetic excitation can be achieved entirely non-invasively through the intact skull in mice. Using a needle-free combination of focused ultrasound-mediated viral delivery and extracorporeal illumination with red light, we achieved selective neuronal activation at depths up to 4 mm in the murine brain, confirmed through cFos expression and electrophysiology measurements within the treated areas. Ultrasound treatment significantly reduced freezing time during recall in fear conditioning experiments, but remote light exposure had a moderate effect on the freezing behavior of mice treated with viral vectors. The proposed method has the potential to open new avenues of studying, but also stimulating, neuronal networks, in an effort to elucidate normal or dysfunctional brain activity and treat neurological diseases. Finally, the same non-invasive methodology could be combined with gene therapy and applied to other organs, such as the eye and the heart.
Collapse
Affiliation(s)
| | - Maria F Murillo
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Rebecca Lynn Noel
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Alec J Batts
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Robin Ji
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Nancy Kwon
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Han Yu
- Department of Electrical Engineering, Columbia University, New York City, NY, USA
| | - Chi-Kun Tong
- Department of Physiology and Cellular Biophysics, Columbia University, New York City, NY, USA
| | | | | | - S Abid Hussaini
- Department of Pathology and Cell Biology, Columbia University, New York City, NY, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA; Department of Radiology, Columbia University, New York City, NY, USA.
| |
Collapse
|
94
|
Beloate LN, Zhang N. Connecting the dots between cell populations, whole-brain activity, and behavior. NEUROPHOTONICS 2022; 9:032208. [PMID: 35350137 PMCID: PMC8957372 DOI: 10.1117/1.nph.9.3.032208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/25/2022] [Indexed: 06/14/2023]
Abstract
Simultaneously manipulating and monitoring both microscopic and macroscopic brain activity in vivo and identifying the linkage to behavior are powerful tools in neuroscience research. These capabilities have been realized with the recent technical advances of optogenetics and its combination with fMRI, here termed "opto-fMRI." Opto-fMRI allows for targeted brain region-, cell-type-, or projection-specific manipulation and targeted Ca 2 + activity measurement to be linked with global brain signaling and behavior. We cover the history, technical advances, applications, and important considerations of opto-fMRI in anesthetized and awake rodents and the future directions of the combined techniques in neuroscience and neuroimaging.
Collapse
Affiliation(s)
- Lauren N. Beloate
- Pennsylvania State University, Department of Biomedical Engineering, Pennsylvania, United States
| | - Nanyin Zhang
- Pennsylvania State University, Department of Biomedical Engineering, Pennsylvania, United States
- Pennsylvania State University, Huck Institutes of the Life Sciences, Pennsylvania, United States
| |
Collapse
|
95
|
Bian WJ, Brewer CL, Kauer JA, de Lecea L. Adolescent sleep shapes social novelty preference in mice. Nat Neurosci 2022; 25:912-923. [PMID: 35618950 PMCID: PMC9283223 DOI: 10.1038/s41593-022-01076-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 04/14/2022] [Indexed: 12/17/2022]
Abstract
Sleep disturbances frequently occur in neurodevelopmental disorders such as autism, but the developmental role of sleep is largely unexplored, and a causal relationship between developmental sleep defects and behavioral consequences in adulthood remains elusive. Here, we show that in mice, sleep disruption (SD) in adolescence, but not in adulthood, causes long-lasting impairment in social novelty preference. Furthermore, adolescent SD alters the activation and release patterns of dopaminergic neurons in the ventral tegmental area (VTA) in response to social novelty. This developmental sleep function is mediated by balanced VTA activity during adolescence; chemogenetic excitation mimics, whereas silencing rescues, the social deficits of adolescent SD. Finally, we show that in Shank3-mutant mice, improving sleep or rectifying VTA activity during adolescence ameliorates adult social deficits. Together, our results identify a critical role of sleep and dopaminergic activity in the development of social interaction behavior.
Collapse
Affiliation(s)
- Wen-Jie Bian
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| | - Chelsie L Brewer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Julie A Kauer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
96
|
Zhang K, Pan J, Yu Y. Regulation of Neural Circuitry under General Anesthesia: New Methods and Findings. Biomolecules 2022; 12:biom12070898. [PMID: 35883456 PMCID: PMC9312763 DOI: 10.3390/biom12070898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 02/01/2023] Open
Abstract
General anesthesia has been widely utilized since the 1840s, but its underlying neural circuits remain to be completely understood. Since both general anesthesia and sleep are reversible losses of consciousness, studies on the neural-circuit mechanisms affected by general anesthesia have mainly focused on the neural nuclei or the pathways known to regulate sleep. Three advanced technologies commonly used in neuroscience, in vivo calcium imaging, chemogenetics, and optogenetics, are used to record and modulate the activity of specific neurons or neural circuits in the brain areas of interest. Recently, they have successfully been used to study the neural nuclei and pathways of general anesthesia. This article reviews these three techniques and their applications in the brain nuclei or pathways affected by general anesthesia, to serve as a reference for further and more accurate exploration of other neural circuits under general anesthesia and to contribute to other research fields in the future.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; (K.Z.); (J.P.)
- Tianjin Institute of Anesthesiology, Tianjin 300052, China
| | - Jiacheng Pan
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; (K.Z.); (J.P.)
- Tianjin Institute of Anesthesiology, Tianjin 300052, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; (K.Z.); (J.P.)
- Tianjin Institute of Anesthesiology, Tianjin 300052, China
- Correspondence:
| |
Collapse
|
97
|
Hee Lee J, Lee S, Kim D, Jae Lee K. Implantable Micro-Light-Emitting Diode (µLED)-based optogenetic interfaces toward human applications. Adv Drug Deliv Rev 2022; 187:114399. [PMID: 35716898 DOI: 10.1016/j.addr.2022.114399] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/29/2022] [Accepted: 06/10/2022] [Indexed: 11/25/2022]
Abstract
Optogenetics has received wide attention in biomedical fields because of itsadvantages in temporal precision and spatial resolution. Beyond contributions to important advances in fundamental research, optogenetics is inspiring a shift towards new methods of improving human well-being and treating diseases. Soft, flexible and biocompatible systems using µLEDs as a light source have been introduced to realize brain-compatible optogenetic implants, but there are still many technical challenges to overcome before their human applications. In this review, we address progress in the development of implantable µLED probes and recent achievements in (i) device engineering design, (ii) driving power, (iii) multifunctionality and (iv) closed-loop systems. (v) Expanded optogenetic applications based on remarkable advances in µLED implants will also be discussed.
Collapse
Affiliation(s)
- Jae Hee Lee
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Sinjeong Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | - Keon Jae Lee
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| |
Collapse
|
98
|
Demidova A, Kahl E, Fendt M. Orexin deficiency affects sensorimotor gating and its amphetamine-induced impairment. Prog Neuropsychopharmacol Biol Psychiatry 2022; 116:110517. [PMID: 35101602 DOI: 10.1016/j.pnpbp.2022.110517] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/17/2022]
Abstract
The orexin neuropeptides have an important role in the regulation of the sleep/wake cycle and foraging, as well as in reward processing and emotions. Furthermore, recent research implicates the orexin system in different behavioral endophenotypes of neuropsychiatric diseases such as social avoidance and cognitive flexibility. Utilizing orexin-deficient mice, the present study tested the hypothesis that orexin is involved in two further mouse behavioral endophenotypes of neuropsychiatric disorders, i.e., sensorimotor gating and amphetamine sensitivity. The data revealed that orexin-deficient mice expressed a deficit in sensorimotor gating, measured by prepulse inhibition of the startle response. Amphetamine treatment impaired prepulse inhibition in wildtype and heterozygous orexin-deficient mice, but had no effects in homozygous orexin-deficient mice. Furthermore, locomotor activity and center time in the open field was not affected by orexin deficiency but was similarly increased or decreased, respectively, by amphetamine treatment in all genotypes. These data indicate that the orexin system modulates prepulse inhibition and is involved in mediating amphetamine's effect on prepulse inhibition. Future studies should investigate whether pharmacological manipulations of the orexin system can be used to treat neuropsychiatric diseases associated with deficits in sensorimotor gating, such as schizophrenia or attention deficit hyperactivity disorder.
Collapse
Affiliation(s)
- Alexandrina Demidova
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany; Psychology Master Program, Otto-von-Guericke University Magdeburg, Germany
| | - Evelyn Kahl
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany; Psychology Master Program, Otto-von-Guericke University Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto-von-Guericke University Magdeburg, Germany.
| |
Collapse
|
99
|
Abstract
Behavioral states naturally alternate between wakefulness and the sleep phases rapid eye movement and nonrapid eye movement sleep. Waking and sleep states are complex processes that are elegantly orchestrated by spatially fine-tuned neurochemical changes of neurotransmitters and neuromodulators including glutamate, acetylcholine, γ-aminobutyric acid, norepinephrine, dopamine, serotonin, histamine, hypocretin, melanin concentrating hormone, adenosine, and melatonin. However, as highlighted in this brief overview, no single neurotransmitter or neuromodulator, but rather their complex interactions within organized neuronal ensembles, regulate waking and sleep states. The neurochemical pathways presented here are aimed to provide a conceptual framework for the understanding of the effects of currently used sleep medications.
Collapse
Affiliation(s)
- Sebastian C Holst
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, Basel 4070, Switzerland.
| | - Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zürich, Winterthurerstrasse 190, Zürich 8057, Switzerland; Zürich Center for Interdisciplinary Sleep Research (ZiS), University of Zürich, Zürich, Switzerland
| |
Collapse
|
100
|
Li S, de Lecea L. The brake matters: Hyperexcitable arousal circuits in sleep fragmentation with age. Clin Transl Med 2022; 12:e900. [PMID: 35696605 PMCID: PMC9191867 DOI: 10.1002/ctm2.900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 11/07/2022] Open
Affiliation(s)
- Shi‐Bin Li
- Department of Psychiatry and Behavioral SciencesStanford University School of MedicineStanfordCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford UniversityStanfordCaliforniaUSA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral SciencesStanford University School of MedicineStanfordCaliforniaUSA
- Wu Tsai Neurosciences InstituteStanford UniversityStanfordCaliforniaUSA
| |
Collapse
|