1
|
Lin YS, Lange D, Baur DM, Foerges A, Chu C, Li C, Elmenhorst EM, Neumaier B, Bauer A, Aeschbach D, Landolt HP, Elmenhorst D. Repeated caffeine intake suppresses cerebral grey matter responses to chronic sleep restriction in an A 1 adenosine receptor-dependent manner: a double-blind randomized controlled study with PET-MRI. Sci Rep 2024; 14:12724. [PMID: 38830861 PMCID: PMC11148136 DOI: 10.1038/s41598-024-61421-8] [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: 12/27/2023] [Accepted: 05/03/2024] [Indexed: 06/05/2024] Open
Abstract
Evidence has shown that both sleep loss and daily caffeine intake can induce changes in grey matter (GM). Caffeine is frequently used to combat sleepiness and impaired performance caused by insufficient sleep. It is unclear (1) whether daily use of caffeine could prevent or exacerbate the GM alterations induced by 5-day sleep restriction (i.e. chronic sleep restriction, CSR), and (2) whether the potential impact on GM plasticity depends on individual differences in the availability of adenosine receptors, which are involved in mediating effects of caffeine on sleep and waking function. Thirty-six healthy adults participated in this double-blind, randomized, controlled study (age = 28.9 ± 5.2 y/; F:M = 15:21; habitual level of caffeine intake < 450 mg; 29 homozygous C/C allele carriers of rs5751876 of ADORA2A, an A2A adenosine receptor gene variant). Each participant underwent a 9-day laboratory visit consisting of one adaptation day, 2 baseline days (BL), 5-day sleep restriction (5 h time-in-bed), and a recovery day (REC) after an 8-h sleep opportunity. Nineteen participants received 300 mg caffeine in coffee through the 5 days of CSR (CAFF group), while 17 matched participants received decaffeinated coffee (DECAF group). We examined GM changes on the 2nd BL Day, 5th CSR Day, and REC Day using magnetic resonance imaging and voxel-based morphometry. Moreover, we used positron emission tomography with [18F]-CPFPX to quantify the baseline availability of A1 adenosine receptors (A1R) and its relation to the GM plasticity. The results from the voxel-wise multimodal whole-brain analysis on the Jacobian-modulated T1-weighted images controlled for variances of cerebral blood flow indicated a significant interaction effect between caffeine and CSR in four brain regions: (a) right temporal-occipital region, (b) right dorsomedial prefrontal cortex (DmPFC), (c) left dorsolateral prefrontal cortex (DLPFC), and (d) right thalamus. The post-hoc analyses on the signal intensity of these GM clusters indicated that, compared to BL, GM on the CSR day was increased in the DECAF group in all clusters but decreased in the thalamus, DmPFC, and DLPFC in the CAFF group. Furthermore, lower baseline subcortical A1R availability predicted a larger GM reduction in the CAFF group after CSR of all brain regions except for the thalamus. In conclusion, our data suggest an adaptive GM upregulation after 5-day CSR, while concomitant use of caffeine instead leads to a GM reduction. The lack of consistent association with individual A1R availability may suggest that CSR and caffeine affect thalamic GM plasticity predominantly by a different mechanism. Future studies on the role of adenosine A2A receptors in CSR-induced GM plasticity are warranted.
Collapse
Affiliation(s)
- Yu-Shiuan Lin
- Centre for Chronobiology, University Psychiatric Clinics Basel, Wilhelm Kleinstr. 27, 4002, Basel, Switzerland.
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland.
- Athinoula. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachussetts General Hospital, Harvard Medical School, Boston, USA.
| | - Denise Lange
- Department of Sleep and Human Factors, Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Diego Manuel Baur
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Sleep & Health Zurich, University Center of Competence, University of Zurich, Zurich, Switzerland
| | - Anna Foerges
- Institute of Neuroscience and Medicine, INM-2, Forschungszentrum Jülich, Wilhelm-Johnen-Strasse, 52428, Jülich, North Rhine-Westphalia, Germany
- Department of Neurophysiology, Institute of Zoology (Bio-II), RWTH Aachen University, Aachen, Germany
| | - Congying Chu
- Institute of Neuroscience and Medicine, INM-2, Forschungszentrum Jülich, Wilhelm-Johnen-Strasse, 52428, Jülich, North Rhine-Westphalia, Germany
| | - Changhong Li
- Institute of Neuroscience and Medicine, INM-2, Forschungszentrum Jülich, Wilhelm-Johnen-Strasse, 52428, Jülich, North Rhine-Westphalia, Germany
| | - Eva-Maria Elmenhorst
- Department of Sleep and Human Factors, Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
- Institute for Occupational, Social, and Environmental Medicine, RWTH Aachen University, Aachen, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine, INM-5, Forschungszentrum Jülich, Jülich, Germany
| | - Andreas Bauer
- Institute of Neuroscience and Medicine, INM-2, Forschungszentrum Jülich, Wilhelm-Johnen-Strasse, 52428, Jülich, North Rhine-Westphalia, Germany
| | - Daniel Aeschbach
- Department of Sleep and Human Factors, Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Bonn, Germany
| | - Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Sleep & Health Zurich, University Center of Competence, University of Zurich, Zurich, Switzerland
| | - David Elmenhorst
- Institute of Neuroscience and Medicine, INM-2, Forschungszentrum Jülich, Wilhelm-Johnen-Strasse, 52428, Jülich, North Rhine-Westphalia, Germany.
- Multimodal Neuroimaging Group, Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
2
|
Terzi A, Ngo KJ, Mourrain P. Phylogenetic conservation of the interdependent homeostatic relationship of sleep regulation and redox metabolism. J Comp Physiol B 2024; 194:241-252. [PMID: 38324048 PMCID: PMC11233307 DOI: 10.1007/s00360-023-01530-4] [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: 07/28/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 02/08/2024]
Abstract
Sleep is an essential and evolutionarily conserved process that affects many biological functions that are also strongly regulated by cellular metabolism. The interdependence between sleep homeostasis and redox metabolism, in particular, is such that sleep deprivation causes redox metabolic imbalances in the form of over-production of ROS. Likewise (and vice versa), accumulation of ROS leads to greater sleep pressure. Thus, it is theorized that one of the functions of sleep is to act as the brain's "antioxidant" at night by clearing oxidation built up from daily stress of the active day phase. In this review, we will highlight evidence linking sleep homeostasis and regulation to redox metabolism by discussing (1) the bipartite role that sleep-wake neuropeptides and hormones have in redox metabolism through comparing cross-species cellular and molecular mechanisms, (2) the evolutionarily metabolic changes that accompanied the development of sleep loss in cavefish, and finally, (3) some of the challenges of uncovering the cellular mechanism underpinning how ROS accumulation builds sleep pressure and cellularly, how this pressure is cleared.
Collapse
Affiliation(s)
- Aslihan Terzi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Keri J Ngo
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
| | - Philippe Mourrain
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- INSERM 1024, Ecole Normale Supérieure, Paris, France.
| |
Collapse
|
3
|
Kompotis K, Mang GM, Hubbard J, Jimenez S, Emmenegger Y, Polysopoulos C, Hor CN, Wigger L, Hébert SS, Mongrain V, Franken P. Cortical miR-709 links glutamatergic signaling to NREM sleep EEG slow waves in an activity-dependent manner. Proc Natl Acad Sci U S A 2024; 121:e2220532121. [PMID: 38207077 PMCID: PMC10801902 DOI: 10.1073/pnas.2220532121] [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: 12/09/2022] [Accepted: 11/29/2023] [Indexed: 01/13/2024] Open
Abstract
MicroRNAs (miRNAs) are key post-transcriptional regulators of gene expression that have been implicated in a plethora of neuronal processes. Nevertheless, their role in regulating brain activity in the context of sleep has so far received little attention. To test their involvement, we deleted mature miRNAs in post-mitotic neurons at two developmental ages, i.e., in early adulthood using conditional Dicer knockout (cKO) mice and in adult mice using an inducible conditional Dicer cKO (icKO) line. In both models, electroencephalographic (EEG) activity was affected and the response to sleep deprivation (SD) altered; while the rapid-eye-movement sleep (REMS) rebound was compromised in both, the increase in EEG delta (1 to 4 Hz) power during non-REMS (NREMS) was smaller in cKO mice and larger in icKO mice compared to controls. We subsequently investigated the effects of SD on the forebrain miRNA transcriptome and found that the expression of 48 miRNAs was affected, and in particular that of the activity-dependent miR-709. In vivo inhibition of miR-709 in the brain increased EEG power during NREMS in the slow-delta (0.75 to 1.75 Hz) range, particularly after periods of prolonged wakefulness. Transcriptome analysis of primary cortical neurons in vitro revealed that miR-709 regulates genes involved in glutamatergic neurotransmission. A subset of these genes was also affected in the cortices of sleep-deprived, miR-709-inhibited mice. Our data implicate miRNAs in the regulation of EEG activity and indicate that miR-709 links neuronal activity during wakefulness to brain synchrony during sleep through the regulation of glutamatergic signaling.
Collapse
Affiliation(s)
- Konstantinos Kompotis
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, ZurichCH-8057, Switzerland
| | - Géraldine M. Mang
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| | - Jeffrey Hubbard
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| | - Sonia Jimenez
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| | - Yann Emmenegger
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| | - Christos Polysopoulos
- Department of Biostatistics, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, ZurichCH-8057, Switzerland
| | - Charlotte N. Hor
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| | - Leonore Wigger
- Genomic Technologies Facility, Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| | - Sébastien S. Hébert
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Axe Neurosciences, Québec, QCG1V 4G2, Canada
- Département de psychiatrie et de neurosciences, Faculté de médecine, Université Laval, Québec, QCG1V 0A6, Canada
| | - Valérie Mongrain
- Department of Neuroscience, Université de Montréal, Montréal, QCH3T 1J4, Canada
- Centre de recherche, Centre hospitalier de l’Université de Montréal, Montréal, QCH2X 0A9, Canada
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, QCH4J 1C5, Canada
| | - Paul Franken
- Center for Integrative Genomics, University of Lausanne, LausanneCH-1015, Switzerland
| |
Collapse
|
4
|
Xue J, Li B, Huang B, Feng H, Li X, Liang S, Yuan F, Wang S, Shi H, Shao J, Shi Y. Sex-dependent and long-lasting effects of adolescent sleep deprivation on social behaviors in adult mice. Pharmacol Biochem Behav 2023; 232:173657. [PMID: 37804868 DOI: 10.1016/j.pbb.2023.173657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Increasing evidence indicates that sleep deprivation (SD) can exert multiple negative effects on neuronal circuits, resulting in memory impairment, depression, and anxiety, among other consequences. The long-term effects of SD during early life on behavioral phenotypes in adulthood are still poorly understood. In this study, we investigated the long-lasting effects of SD in adolescence on social behaviors, including empathic ability and social dominance, and the role of the gut microbiota in these processes, using a series of behavioral paradigms in mice combined with 16S rRNA gene pyrosequencing. Behavioral assay results showed that SD in adolescence significantly reduced the frequency of licking, the total time spent licking, and the time spent sniffing during the emotional contagion test in male mice, effects that were not observed in female mice. These findings indicated that SD in adolescence exerts long-term, negative effects on empathic ability in mice and that this effect is sex-dependent. In contrast, SD in adolescence had no significant effect on locomotor activities, social dominance but decreased social interaction in male mice in adulthood. Meanwhile, 16S rRNA gene pyrosequencing results showed that gut microbial richness and diversity were significantly altered in adult male mice subjected to SD in adolescence. Our data provide direct evidence that SD in youth can induce alterations in empathic ability in adult male mice, which may be associated with changes in the gut microbiota. These findings highlight the long-lasting effects of sleep loss in adolescence on social behaviors in adulthood and the role played by the brain-gut axis.
Collapse
Affiliation(s)
- Jiping Xue
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Bingyu Li
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Boya Huang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Hao Feng
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Xinrui Li
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Shihao Liang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Fang Yuan
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Sheng Wang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medicinal University, Shijiazhuang 050017, China; Nursing School, Hebei Medicinal University, Shijiazhuang 050031, China.
| | - Juan Shao
- Department of Senile Disease, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China.
| | - Yun Shi
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
5
|
Milman NE, Tinsley CE, Raju RM, Lim MM. Loss of sleep when it is needed most - Consequences of persistent developmental sleep disruption: A scoping review of rodent models. Neurobiol Sleep Circadian Rhythms 2023; 14:100085. [PMID: 36567958 PMCID: PMC9768382 DOI: 10.1016/j.nbscr.2022.100085] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Sleep is an essential component of development. Developmental sleep disruption (DSD) impacts brain maturation and has been associated with significant consequences on socio-emotional development. In humans, poor sleep during infancy and adolescence affects neurodevelopmental outcomes and may be a risk factor for the development of autism spectrum disorder (ASD) or other neuropsychiatric illness. Given the wide-reaching and enduring consequences of DSD, identifying underlying mechanisms is critical to best inform interventions with translational capacity. In rodents, studies have identified some mechanisms and neural circuits by which DSD causes later social, emotional, sensorimotor, and cognitive changes. However, these studies spanned methodological differences, including different developmental timepoints for both sleep disruption and testing, different DSD paradigms, and even different rodent species. In this scoping review on DSD in rodents, we synthesize these various studies into a cohesive framework to identify common neural mechanisms underlying DSD-induced dysfunction in brain and behavior. Ultimately, this review serves the goal to inform the generation of novel translational interventions for human developmental disorders featuring sleep disruption.
Collapse
Affiliation(s)
- Noah E.P. Milman
- Oregon Health and Science University, Dept. of Behavioral and Systems Neuroscience, Portland, OR, 97214, USA
- Veterans Affairs Portland Health Care System, Portland, OR, 97214, USA
| | - Carolyn E. Tinsley
- Oregon Health and Science University, Dept. of Behavioral and Systems Neuroscience, Portland, OR, 97214, USA
- Veterans Affairs Portland Health Care System, Portland, OR, 97214, USA
| | - Ravikiran M. Raju
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Miranda M. Lim
- Oregon Health and Science University, Dept. of Behavioral and Systems Neuroscience, Portland, OR, 97214, USA
- Veterans Affairs Portland Health Care System, Portland, OR, 97214, USA
| |
Collapse
|
6
|
Flores CC, Loschky SS, Marshall W, Spano GM, Massaro Cenere M, Tononi G, Cirelli C. Identification of ultrastructural signatures of sleep and wake in the fly brain. Sleep 2022; 45:zsab235. [PMID: 35554595 PMCID: PMC9113029 DOI: 10.1093/sleep/zsab235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/12/2021] [Indexed: 11/14/2022] Open
Abstract
The cellular consequences of sleep loss are poorly characterized. In the pyramidal neurons of mouse frontal cortex, we found that mitochondria and secondary lysosomes occupy a larger proportion of the cytoplasm after chronic sleep restriction compared to sleep, consistent with increased cellular burden due to extended wake. For each morphological parameter, the within-animal variance was high, suggesting that the effects of sleep and sleep loss vary greatly among neurons. However, the analysis was based on 4-5 mice/group and a single section/cell. Here, we applied serial block-face scanning electron microscopy to identify signatures of sleep and sleep loss in the Drosophila brain. Stacks of images were acquired and used to obtain full 3D reconstructions of the cytoplasm and nucleus of 263 Kenyon cells from adult flies collected after a night of sleep (S) or after 11 h (SD11) or 35 h (SD35) of sleep deprivation (9 flies/group). Relative to S flies, SD35 flies showed increased density of dark clusters of chromatin and Golgi apparata and a trend increase in the percent of cell volume occupied by mitochondria, consistent with increased need for energy and protein supply during extended wake. Logistic regression models could assign each neuron to the correct experimental group with good accuracy, but in each cell, nuclear and cytoplasmic changes were poorly correlated, and within-fly variance was substantial in all experimental groups. Together, these results support the presence of ultrastructural signatures of sleep and sleep loss but underscore the complexity of their effects at the single-cell level.
Collapse
Affiliation(s)
- Carlos C Flores
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Sophia S Loschky
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - William Marshall
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Mathematics and Statistics, Brock University, St. Catharines, ON, Canada
| | | | | | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
7
|
Wang L, Aton SJ. Perspective - ultrastructural analyses reflect the effects of sleep and sleep loss on neuronal cell biology. Sleep 2022; 45:zsac047. [PMID: 35554582 PMCID: PMC9113019 DOI: 10.1093/sleep/zsac047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/05/2022] [Indexed: 11/13/2022] Open
Abstract
Recent electron microscopic analyses of neurons in the Drosophila and rodent brain demonstrate that acute or chronic sleep loss can alter the structures of various organelles, including mitochondria, nucleus, and Golgi apparatus. Here, we discuss these findings in the context of biochemical findings from the sleep deprived brain, to clarify how these morphological changes may related to altered organelle function. We discuss how, taken together, the available data suggest that sleep loss (particularly chronic sleep loss) disrupts such fundamental cellular processes as transcription, translation, intracellular transport, and metabolism. A better understanding of these effects will have broad implications for understanding the biological importance of sleep, and the relationship of sleep loss to neuropathology.
Collapse
Affiliation(s)
- Lijing Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Sara J Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Sleep deficiency as a driver of cellular stress and damage in neurological disorders. Sleep Med Rev 2022; 63:101616. [PMID: 35381445 PMCID: PMC9177816 DOI: 10.1016/j.smrv.2022.101616] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/23/2022]
Abstract
Neurological disorders encompass an extremely broad range of conditions, including those that present early in development and those that progress slowly or manifest with advanced age. Although these disorders have distinct underlying etiologies, the activation of shared pathways, e.g., integrated stress response (ISR) and the development of shared phenotypes (sleep deficits) may offer clues toward understanding some of the mechanistic underpinnings of neurologic dysfunction. While it is incontrovertibly complex, the relationship between sleep and persistent stress in the brain has broad implications in understanding neurological disorders from development to degeneration. The convergent nature of the ISR could be a common thread linking genetically distinct neurological disorders through the dysregulation of a core cellular homeostasis pathway.
Collapse
|
9
|
Li RR, Yan J, Chen H, Zhang WW, Hu YB, Zhang J, Hu ZA, Xiong Y, Yao ZX, Hu B. Sleep Deprivation Impairs Learning-Induced Increase in Hippocampal Sharp Wave Ripples and Associated Spike Dynamics during Recovery Sleep. Cereb Cortex 2021; 32:824-838. [PMID: 34383018 DOI: 10.1093/cercor/bhab247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 11/14/2022] Open
Abstract
Sleep deprivation (SD) causes deficits in off-line memory consolidation, but the underlying network oscillation mechanisms remain unclear. Hippocampal sharp wave ripple (SWR) oscillations play a critical role in off-line memory consolidation. Therefore, we trained mice to learn a hippocampus-dependent trace eyeblink conditioning (tEBC) task and explored the influence of 1.5-h postlearning SD on hippocampal SWRs and related spike dynamics during recovery sleep. We found an increase in hippocampal SWRs during postlearning sleep, which predicted the consolidation of tEBC in conditioned mice. In contrast, sleep-deprived mice showed a loss of tEBC learning-induced increase in hippocampal SWRs during recovery sleep. Moreover, the sleep-deprived mice exhibited weaker reactivation of tEBC learning-associated pyramidal cells in hippocampal SWRs during recovery sleep. In line with these findings, tEBC consolidation was impaired in sleep-deprived mice. Furthermore, sleep-deprived mice showed augmented fast excitation from pyramidal cells to interneurons and enhanced participation of interneurons in hippocampal SWRs during recovery sleep. Among various interneurons, parvalbumin-expressing interneurons specifically exhibited overexcitation during hippocampal SWRs. Our findings suggest that altered hippocampal SWRs and associated spike dynamics during recovery sleep may be candidate network oscillation mechanisms underlying SD-induced memory deficits.
Collapse
Affiliation(s)
- Rong-Rong Li
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Jie Yan
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Hao Chen
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Wei-Wei Zhang
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yu-Bo Hu
- Department of Orthopaedics, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jie Zhang
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Zhi-An Hu
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yan Xiong
- Department of Orthopaedics, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zhong-Xiang Yao
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Bo Hu
- Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China.,Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Army Medical University, Chongqing 400038, China
| |
Collapse
|
10
|
Novozhilova M, Mishchenko T, Kondakova E, Lavrova T, Gavrish M, Aferova S, Franceschi C, Vedunova M. Features of age-related response to sleep deprivation: in vivo experimental studies. Aging (Albany NY) 2021; 13:19108-19126. [PMID: 34320466 PMCID: PMC8386558 DOI: 10.18632/aging.203372] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/17/2021] [Indexed: 12/23/2022]
Abstract
Insomnia is currently considered one of the potential triggers of accelerated aging. The frequency of registered sleep-wake cycle complaints increases with age and correlates with the quality of life of elderly people. Nevertheless, whether insomnia is actually an age-associated process or whether it acts as an independent stress-factor that activates pathological processes, remains controversial. In this study, we analyzed the effects of long-term sleep deprivation modeling on the locomotor and orienting-exploratory activity, spatial learning abilities and working memory of C57BL/6 female mice of different ages. We also evaluated the modeled stress influence on morphological changes in brain tissue, the functional activity of the mitochondrial apparatus of nerve cells, and the level of DNA methylation and mRNA expression levels of the transcription factor HIF-1α (Hif1) and age-associated molecular marker PLIN2. Our findings point to the age-related adaptive capacity of female mice to the long-term sleep deprivation influence. For young (1.5 months) mice, the modeled sleep deprivation acts as a stress factor leading to weight loss against the background of increased food intake, the activation of animals' locomotor and exploratory activity, their mnestic functions, and molecular and cellular adaptive processes ensuring animal resistance both to stress and risk of accelerated aging development. Sleep deprivation in adult (7-9 months) mice is accompanied by an increase in body weight against the background of active food intake, increased locomotor and exploratory activity, gross disturbances in mnestic functions, and decreased adaptive capacity of brain cells, that potentially increasing the risk of pathological reactions and neurodegenerative processes.
Collapse
Affiliation(s)
- Maria Novozhilova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Tatiana Mishchenko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Elena Kondakova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Tatiana Lavrova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Maria Gavrish
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Svetlana Aferova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Claudio Franceschi
- Institute of Information Technologies, Mathematics and Mechanics (ITMM), National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Maria Vedunova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| |
Collapse
|
11
|
Effects of Severe Sleep Disruption on the Synaptic Ultrastructure of Young Mice. eNeuro 2021; 8:ENEURO.0077-21.2021. [PMID: 34193511 PMCID: PMC8287877 DOI: 10.1523/eneuro.0077-21.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/07/2021] [Accepted: 06/16/2021] [Indexed: 11/21/2022] Open
Abstract
There is molecular, electrophysiological, and ultrastructural evidence that a net increase in synaptic strength occurs in many brain circuits during spontaneous wake (SW) or short sleep deprivation, reflecting ongoing learning. Sleep leads instead to a broad but selective weakening of many forebrain synapses, thus preventing synaptic saturation and decreasing the energy cost of synaptic activity. Whether synaptic potentiation can persist or further increase after long sleep deprivation is unknown. Whether synaptic renormalization can occur during chronic sleep restriction (CSR) is also unknown. Here, we addressed these questions by measuring an established ultrastructural measure of synaptic strength, the axon-spine interface (ASI), in the primary motor cortex (M1) of (1) one-month-old adolescent mice CSR using a paradigm that decreases NREM and REM sleep by two/thirds; (2) in two-week-old mouse pups sleep deprived for 15 h, or allowed afterward to recover for 16 h. Both groups were compared with mice of the same age that were asleep or awake for a few hours (both sexes). The ASI size of CSR mice (n = 3) was comparable to that measured after SW or short sleep deprivation and larger than after sleep (n = 4/group). In pups, the ASI size increased after short sleep loss (n = 3) relative to sleep (n = 4), fell below sleep levels after long sleep deprivation (n = 4), and remained low after recovery (n = 3). Long sleep deprived pups also lost some weight. These results suggest that (1) severe sleep restriction is incompatible with synaptic renormalization; (2) very young mice cannot maintain high synaptic strength during prolonged wake.
Collapse
|
12
|
Sanjari Moghaddam H, Mohammadi E, Dolatshahi M, Mohebi F, Ashrafi A, Khazaie H, Aarabi MH. White matter microstructural abnormalities in primary insomnia: A systematic review of diffusion tensor imaging studies. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110132. [PMID: 33049323 DOI: 10.1016/j.pnpbp.2020.110132] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/12/2020] [Accepted: 10/06/2020] [Indexed: 02/01/2023]
Abstract
Primary insomnia (PI), the most common sleep disorder, is primarily characterized by difficulties in initiating and maintaining sleep and deficits in daytime functioning. Study of white matter (WM) connections of the brain might provide valuable information regarding the underlying neural mechanism of PI. Diffusion tensor imaging (DTI) provides non-invasive access to the microstructural and network properties of brain WM, and thus, a great opportunity to quantitatively and sensitively study the human brain. The current literature of PI does not provide a consistent explanation of the etiology and pathology of the disorder; thus, we searched PubMed, EMBASE, and SCOPUS for DTI studies that compared WM integrity or network organization between PI patients and healthy controls to integrate all existing literature as an insight for further studies, and, hopefully, effective prevention and management of the disorder. English peer-reviewed full-text publications that investigated the diffusion indices of PI patients or those with insomnia symptoms compared with a group of healthy controls were included. We included 11 studies and extracted the data for qualitative synthesis. Except for one study, all studies were rated as high-quality, based on quality assessment. In aggregation, a total of 541 patients with PI and 679 healthy controls were included in this study. Our review of DTI studies suggests that WM disruptions in PI are better characterized in the context of neural networks. Frontostriatal, frontothalamic, and corticocortiscal networks, as well as the limbic system, seem to be the main neural networks with microstructural and network alterations in patients with PI. To illustrate, different parts of corona radiate and internal capsule within the corticosubcortical networks and superior longitudinal fasciculus within the corticocortical networks showed altered microstructural properties in PI patients. In view of the fact that the findings from individual studies are heterogeneous, it is difficult to derive consistent findings from the results, and the divergence of the findings must not be disregarded. Thus, this study is a starting point for future studies, and its implications for etiology and pathogenesis of insomnia should be regarded cautiously.
Collapse
Affiliation(s)
| | - Esmaeil Mohammadi
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Dolatshahi
- Faculty of medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnam Mohebi
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Agaah Ashrafi
- Department of Psychiatry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Habibolah Khazaie
- Sleep Disorders Research Center, Department of Psychiatry, Kermanshah University of Medical Sciences (KUMS), Kermanshah, Iran
| | | |
Collapse
|
13
|
Disrupted frontostriatal connectivity in primary insomnia: a DTI study. Brain Imaging Behav 2021; 15:2524-2531. [PMID: 33651331 DOI: 10.1007/s11682-021-00454-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/28/2020] [Accepted: 01/10/2021] [Indexed: 12/20/2022]
Abstract
Dysfunction of the sleep-wake transition is considered to be associated with the pathology of patients with primary insomnia (PI). Previous animal study had reported that brain circuits between the striatum and cortex can regulate sleep-wake transitions. So far, few studies have systematically explored the structural connectivity of the striatum-centered circuits and their potential roles in patients with PI. In this study, we chosen the striatum as the seed and 10 priori target regions as masks to assess the structural connectivity by using seed-based classification with a diffusion tensor imaging (DTI) probabilistic tractography method. Track strengths of the striatum-centered circuits were compared between 22 patients with PI (41.27 ± 9.21 years) and 30 healthy controls (HC) (35.2 ± 8.14 years). Pittsburgh Sleep Quality Index (PSQI) was used to measure the sleep quality in all participants. Lower track strengths (left striatum- anterior cingulate cortex (ACC), left striatum- dorsal anterior cingulate cortex (dACC), left striatum-Hippocampus, and right striatum-Hippocampus) were observed in patients with PI compared to HC. Additionally, the lower track strengths of brain circuits mentioned above were negatively correlated with PSQI. Taken together, our findings revealed the lower tract strength of frontostriatal circuits in patients with PI and HC, which provided the implications of the system-level structural connections of frontostriatal circuits in the pathology of PI. We suggested that the track strengths of the frontostriatal circuits calculated from DTI can be the potential neuroimaging biomarkers of the sleep quality in patients with PI.
Collapse
|
14
|
Tuan LH, Tsao CY, Lee LJH, Lee LJ. Voluntary exercise ameliorates synaptic pruning deficits in sleep-deprived adolescent mice. Brain Behav Immun 2021; 93:96-110. [PMID: 33358980 DOI: 10.1016/j.bbi.2020.12.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/17/2020] [Accepted: 12/16/2020] [Indexed: 11/26/2022] Open
Abstract
Adolescence is a critical period for brain development and adequate sleep during this period is essential for physical function and mental health. Emerging evidence has detailed the neurological impacts of sleep insufficiency on adolescents, as was unveiled by our previous study, microglia, one of the crucial contributors to synaptic pruning, is functionally disrupted by lack of sleep. Here, we provided evidence featuring the protective effect and the underlying mechanisms of voluntary exercise (VE) on microglial functions in an adolescent 72 h sleep deprivation (SD) model. We identified that the aberrant hippocampal neuronal activity and impaired short-term memory performance in sleep-deprived mice were prevented by 11 days of VE. VE significantly normalized the SD-induced dendritic spine increment and maintained the microglial phagocytic ability in sleep-deprived mice. Moreover, we found that the amendment of the noradrenergic signal in the central nervous system may explain the preventative effects of VE on the abnormalities of microglial and neuronal functions caused by SD. These data suggested that VE may confer protection to the microglia-mediated synaptic pruning in the sleep-deprived adolescent brains. Therefore, physical exercise could be a beneficial health practice for the adolescents that copes the adverse influence of inevitable sleep insufficiency.
Collapse
Affiliation(s)
- Li-Heng Tuan
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC
| | - Chih-Yu Tsao
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC
| | - Lukas Jyuhn-Hsiarn Lee
- Division of Environmental Health and Occupational Medicine, National Health Research Institutes, Miaoli, Taiwan, ROC
| | - Li-Jen Lee
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC; Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan, ROC; Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
15
|
Frank MG. The Ontogenesis of Mammalian Sleep: Form and Function. CURRENT SLEEP MEDICINE REPORTS 2020; 6:267-279. [PMID: 33816063 PMCID: PMC8014960 DOI: 10.1007/s40675-020-00190-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE OF REVIEW To present an up-to-date review and synthesis of findings about perinatal sleep development and function. I discuss landmark events in sleep ontogenesis, evidence that sleep promotes brain development and plasticity, and experimental considerations in this topic. RECENT FINDINGS Mammalian sleep undergoes dramatic changes in expression and regulation during perinatal development. This includes a progressive decrease in rapid-eye-movement (REM) sleep time, corresponding increases in nonREM sleep and wake time, and the appearance of mature sleep regulatory processes (homeostatic and circadian). These developmental events coincide with periods of rapid brain maturation and heightened synaptic plasticity. The latter involve an initial experience-independent phase, when circuit development is guided by spontaneous activity, and later occurring critical periods, when these circuits are shaped by experience. SUMMARY These ontogenetic changes suggest important interactions between sleep and brain development. More specifically, sleep may promote developmental programs of synaptogenesis and synaptic pruning and influence the opening and closing of critical periods of brain plasticity.
Collapse
Affiliation(s)
- Marcos G Frank
- Washington State University Spokane, Elson S. Floyd College of Medicine, Pharmaceutical and Biomedical Science Building 213, 412 E. Spokane Falls Blvd
| |
Collapse
|
16
|
Zhang J, Zhang L, Chang Y, Gu Q, Zhang J, Zhu Z, Qian Z, Wei C, Liu Z, Ren W, Han J. The Endocannabinoid System Contributes to Memory Deficits Induced by Rapid-eye-movement Sleep Deprivation in Adolescent Mice. Neuroscience 2020; 433:174-183. [PMID: 32198011 DOI: 10.1016/j.neuroscience.2020.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 02/07/2023]
Abstract
Sleep loss or insomnia is among the contributing factors of cognitive deficit, the underlying mechanisms of which remain largely elusive. The endocannabinoid (eCB) system plays a role in sleep, while it is unknown if it is involved in the regulation of memory retrieval by sleep deprivation. In addition, it still controversial how rapid-eye-movement sleep deprivation (REMSD) affects the spatial memory of adolescent mice. Here, we found that 24-h REMSD impairs spatial memory retrieval of adolescent mice in an object-place recognition task, which was rescued by NESS0327, a neutral cannabinoid receptor 1 (CB1R) antagonist. Mechanistically, REMSD induced eCB-mediated short-term and long-term synaptic plasticity changing including depolarization-induced suppression of inhibition (DSI) in the pyramidal neurons of the hippocampus, in which long-term synaptic plasticity changing was rescued by NESS0327. REMSD downregulated monoacylglycerol lipase, a hydrolase for the endocannabinoid 2-arachidonoylglycerol (2-AG), suggesting the involvement of eCB accumulation and the consequent synaptic plasticity in REMSD-elicited memory impairment in adolescent mice. These findings shed light on the role of sleep disorders in learning and memory deficit of adolescents.
Collapse
Affiliation(s)
- Jinming Zhang
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Lizi Zhang
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China; College of Life Sciences, Shaanxi Normal University, China
| | - Yuan Chang
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Qiaofen Gu
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Junmin Zhang
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Zhou Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Zhaoqiang Qian
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Chunling Wei
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Zhiqiang Liu
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Wei Ren
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China
| | - Jing Han
- Key Lab of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, China.
| |
Collapse
|
17
|
Karthikeyan R, Cardinali DP, Shakunthala V, Spence DW, Brown GM, Pandi-Perumal SR. Understanding the role of sleep and its disturbances in Autism spectrum disorder. Int J Neurosci 2020; 130:1033-1046. [PMID: 31903819 DOI: 10.1080/00207454.2019.1711377] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Several studies have established a positive relationship between sleep difficulties and symptomatology in ASD children. The rationale for this review is to describe and discuss the sleep difficulties, which are one of the significant complications associated with autism spectrum disorder (ASD). PURPOSE Many types of sleep disorders have been reported in ASD individuals, but still lack a comprehensive study and in-depth analysis. Despite the contribution of sleep problems to the overall symptoms of ASD, the symptoms of disturbed sleep experienced by many affected patients have only recently started to receive attention from clinicians and family members. MATERIALS AND METHODS This narrative overview has been prepared based on searching standard research databases with specific keywords; b. Additional search was made using the bibliographies of the retrieved articles; and c. author's collection of relevant peer-reviewed articles. Once selected, manuscripts are then compared and summarized based on the author's perspective. Results are based on a qualitative rather than a quantitative level. RESULTS This article highlights the role of sleep in the brain and neural development of children and emphasizes that the intensity of sleep problems is associated with an increased occurrence of ASD symptoms. It also suggests the significance of treating sleep problems in ASD individuals. CONCLUSIONS The review provides broader perspectives and a better understanding of sleep problems in pathophysiology, mechanism, and management with respect to ASD individuals. Finally, the implications for clinical practice and future agendas have also been discussed.
Collapse
Affiliation(s)
- Ramanujam Karthikeyan
- Department of Animal Behavior & Physiology, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Daniel P Cardinali
- Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Venkat Shakunthala
- Department of Zoology, University of Mysuru, Manasagangotri, Karnataka, India
| | - David Warren Spence
- Independent Researcher, Department of Sleep Medicine, Toronto, Ontario, Canada
| | - Gregory M Brown
- Molecular Brain Science, Centre for Addiction and Mental Health, Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
18
|
Mukherjee KK, Lee AY, Zhu L, Darvas M, Ladiges W. Sleep-deprived cognitive impairment in aging mice is alleviated by rapamycin. AGING PATHOBIOLOGY AND THERAPEUTICS 2019; 1:5-9. [PMID: 35083443 PMCID: PMC8789090 DOI: 10.31491/apt.2019.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Sleep deprivation-induced cognitive impairment is a major health concern and an age-related risk factor for dementia. There is an urgent need to develop ways of preventing the adverse neurological effects of sleep deprivation, but current preclinical animal models of short-term sleep deprivation are not well described. METHODS C57BL6 mice of varying ages were sleep deprived for 4 hours a day for 4 days, and then tested with a Box maze navigation task. RESULTS Sleep deprived mice at young, middle and older ages showed learning impairment that varied by strain and gender. In general, females were more sensitive to sleep deprivation than males. To determine whether sleep deprivation-induced learning impairment would respond to therapeutic intervention, an independent cohort of mice was treated with rapamycin daily during the 4 days of sleep deprivation. Mice that were sleep deprived and treated with rapamycin showed significant improvement in learning time suggesting that the cognitive impairment might be associated in part with molecular and cellular mechanisms targeted by rapamycin. CONCLUSIONS The observations from this study suggest that aging mice would be productive models to study pathobiology and therapeutic intervention of cognitive impairment triggered by age-related sleeping disorders in people.
Collapse
Affiliation(s)
- Kishore K. Mukherjee
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle WA 98104, USA
| | - Amanda Y. Lee
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle WA 98104, USA
| | - Lida Zhu
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle WA 98104, USA
| | - Martin Darvas
- Department of Pathology, School of Medicine, University of Washington, Seattle WA 98104, USA
| | - Warren Ladiges
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle WA 98104, USA
| |
Collapse
|
19
|
Harkness JH, Bushana PN, Todd RP, Clegern WC, Sorg BA, Wisor JP. Sleep disruption elevates oxidative stress in parvalbumin-positive cells of the rat cerebral cortex. Sleep 2019; 42:5145871. [PMID: 30371896 DOI: 10.1093/sleep/zsy201] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Indexed: 11/13/2022] Open
Abstract
We used a novel automated sleep disruption (SD) apparatus to determine the impact of SD on sleep and molecular markers of oxidative stress in parvalbumin (PV) neurons in the rat prefrontal cortex (PFC). Rats were subjected to two 6 hr SD sessions from zeitgeber time (ZT) 0 to ZT6, one by the gentle handling method and the other by an automated agitator running the length of the rat's home cage floor (a novel SD method). The same rats were later subjected to a 12 hr SD session from ZT0 to ZT12. Sleep was disrupted with both methods, although rats slept less during gentle handling than during the automated condition. Immediately after both SD sessions, rats displayed compensatory sleep characterized by elevated slow-wave activity. We measured in the prelimbic prefrontal cortex (prelimbic PFC; 6 and 12 hr SD) and orbital frontal cortex (12 hr SD) the intensity of the oxidative stress marker, 8-oxo-2'-deoxyguanosine (8-oxo-dG) as well as the staining intensity of PV and the PV cell-associated perineuronal net marker, Wisteria floribunda agglutinin (WFA). In the prelimbic PFC, 6 hr SD increased the intensity of 8-oxo-dG, PV, and WFA. After 12 hr SD, the intensity of 8-oxo-dG was elevated in all neurons. PV intensity was elevated only in neurons colabeled with 8-oxo-dG or WFA, and no changes were found in WFA intensity. We conclude that in association with SD-induced sleep drive, PV neurons in the prelimbic PFC exhibit oxidative stress.
Collapse
Affiliation(s)
- John H Harkness
- Department of Integrative Physiology and Neuroscience, Translational Addiction Research Center, Washington State University, Vancouver, WA
| | - Priyanka N Bushana
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Spokane, WA
| | - Ryan P Todd
- Department of Integrative Physiology and Neuroscience, Translational Addiction Research Center, Washington State University, Vancouver, WA
| | - William C Clegern
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Spokane, WA
| | - Barbara A Sorg
- Department of Integrative Physiology and Neuroscience, Translational Addiction Research Center, Washington State University, Vancouver, WA
| | - Jonathan P Wisor
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Spokane, WA
| |
Collapse
|
20
|
Tuan LH, Lee LJ. Microglia-mediated synaptic pruning is impaired in sleep-deprived adolescent mice. Neurobiol Dis 2019; 130:104517. [PMID: 31229687 DOI: 10.1016/j.nbd.2019.104517] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 05/10/2019] [Accepted: 06/19/2019] [Indexed: 01/26/2023] Open
Abstract
The detrimental effects of sleep insufficiency have been extensively explored. However, only a few studies have addressed this issue in adolescents. In the present study, we examined and compared the effects of 72 h paradoxical sleep deprivation (SD) on adolescent (5 weeks old) and adult (~12 weeks old) mice. Following 72 h of SD, induced by a modified multiple-platform method, mice were subjected to behavioral, histological and neurochemical examinations. In both adolescent and adult mice, SD adversely affected short-term memory in a novel object recognition test. Compared with normal-sleep controls, sleep-deprived adolescent mice had an increased density of excitatory synapses in the granule cells of the dentate gyrus, but no such pattern was observed in the adult group. The engulfment of postsynaptic components within the microglia after SD was reduced in adolescents but not in adults, suggesting an impaired microglia-mediated synaptic pruning in adolescent SD mice. Possible contributing factors included the decreases in CX3CR1, CD11b and P2Y12, closely associated with the synaptic pruning via microglial phagocytosis. In adult SD mice, microglia-associated inflammatory reactions were noted. In sum, sleep deprivation induces age-dependent microglial reactions in adolescent and adult mice, respectively; yet results in similar defects in short-term recognition memory. Sufficient sleep is indispensable for adolescents and adults.
Collapse
Affiliation(s)
- Li-Heng Tuan
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Li-Jen Lee
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC; Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC; Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
21
|
Bellesi M, Haswell JD, de Vivo L, Marshall W, Roseboom PH, Tononi G, Cirelli C. Myelin modifications after chronic sleep loss in adolescent mice. Sleep 2019; 41:4850494. [PMID: 29741724 DOI: 10.1093/sleep/zsy034] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Indexed: 01/28/2023] Open
Abstract
Study Objectives Previous studies found that sleep loss can suppress the expression of genes implicated in myelination and can have adverse effects on oligodendrocyte precursor cells. On the other hand, sleep may favor myelination by promoting the expression of genes involved in its formation and maintenance. Albeit limited, these results suggest that sleep loss can have detrimental effects on the formation and maintenance of myelin. Methods Here, we tested this hypothesis by evaluating ultrastructural modifications of myelin in two brain regions (corpus callosum and lateral olfactory tract) of mice exposed to different periods of sleep loss, from a few hours of sleep deprivation to ~5 days of chronic sleep restriction. In addition, we measured the internodal length-the distance between consecutive nodes of Ranvier along the axon-and plasma corticosterone levels. Results We find that g-ratio-the ratio of the diameter of the axon itself to the outer diameter of the myelinated fiber-increases after chronic sleep loss. This effect is mediated by a reduction in myelin thickness and is not associated with changes in the internodal length. Relative to sleep, plasma corticosterone levels increase after acute sleep deprivation, but show only a trend to increase after chronic sleep loss. Conclusions Chronic sleep loss may negatively affect myelin.
Collapse
Affiliation(s)
- Michele Bellesi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI.,Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | | | - Luisa de Vivo
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - William Marshall
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | | | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
22
|
|
23
|
Wu Y, Liu M, Zeng S, Ma X, Yan J, Lin C, Xu G, Li G, Yin Y, Fu S, Hua K, Li C, Wang T, Li C, Jiang G. Abnormal Topology of the Structural Connectome in the Limbic Cortico-Basal-Ganglia Circuit and Default-Mode Network Among Primary Insomnia Patients. Front Neurosci 2018; 12:860. [PMID: 30532688 PMCID: PMC6266325 DOI: 10.3389/fnins.2018.00860] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022] Open
Abstract
Purpose: Primary insomnia (PI) is the second most common mental disorder. However, the topologic alterations in structural brain connectome in patients with PI remain largely unknown. Methods: A total of 44 PI patients and 46 age-, gender-, and education level matched healthy control (HC) participants were recruited in this study. Diffusion tensor imaging (DTI) and resting state MRI were used to construct structural connectome for each participant, and the network parameters were employed by non-parametric permutations to evaluate the significant differences between the two groups. Relationships between abnormal network metrics and clinical characteristics, including the disease duration, the Pittsburgh Sleep Quality Index (PSQI), the Insomnia Severity Index (ISI), the Self-Rating Anxiety Scale (SAS), and the Self-Rating Depression Scale (SDS), were investigated with Spearman's correlation analysis in PI patients. Results: PI patients demonstrated small-world architecture with lower global (P = 0.005) and local (P = 0.035) efficiencies compared with the HC group. The unique hub nodal properties in PI patients were mainly in the right limbic cortico-basal-ganglia circuit. Five disrupted subnetworks in PI patients were observed in the limbic cortico-basal-ganglia circuit and left default-mode networks (DMN) (P < 0.05, NBS corrected). Moreover, most unique hub nodal properties in the right limbic cortico-basal-ganglia circuit were significantly correlated with disease duration, and clinical characteristics (SAS, SDS, ISI scores) in PI processing. Conclusion: These findings suggested the abnormal anatomical network architecture may be closely linked to clinical characteristics in PI. The study provided novel insights into the neural substrates underlying symptoms and neurophysiologic mechanisms of PI.
Collapse
Affiliation(s)
- Yunfan Wu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Mengchen Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Shaoqing Zeng
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiaofen Ma
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Jianhao Yan
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Chulan Lin
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Guang Xu
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Guomin Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yi Yin
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shishun Fu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Kelei Hua
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Chao Li
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Tianyue Wang
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Cheng Li
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Guihua Jiang
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
24
|
Bellesi M, de Vivo L, Koebe S, Tononi G, Cirelli C. Sleep and Wake Affect Glycogen Content and Turnover at Perisynaptic Astrocytic Processes. Front Cell Neurosci 2018; 12:308. [PMID: 30254569 PMCID: PMC6141665 DOI: 10.3389/fncel.2018.00308] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/23/2018] [Indexed: 12/26/2022] Open
Abstract
Astrocytic glycogen represents the only form of glucose storage in the brain, and one of the outcomes of its breakdown is the production of lactate that can be used by neurons as an alternative energetic substrate. Since brain metabolism is higher in wake than in sleep, it was hypothesized that glycogen stores are depleted during wake and replenished during sleep. Furthermore, it was proposed that glycogen depletion leads to the progressive increase in adenosine levels during wake, providing a homeostatic signal that reflects the buildup of sleep pressure. However, previous studies that measured glycogen dynamics across the sleep/wake cycle obtained inconsistent results, and only measured glycogen in whole tissue. Since most energy in the brain is used to sustain synaptic activity, here we employed tridimensional electron microscopy to quantify glycogen content in the astrocytic processes surrounding the synapse. We studied axon-spine synapses in the frontal cortex of young mice after ~7 h of sleep, 7–8 h of spontaneous or forced wake, or 4.5 days of sleep restriction. Relative to sleep, all wake conditions increased the number of glycogen granules around the synapses to a similar extent. However, progressively longer periods of wake were associated with progressively smaller glycogen granules, suggesting increased turnover. Despite the increased number of granules, in all wake conditions the estimated amount of glucose within the granules was lower than in sleep, indicating that sleep may favor glucose storage. Finally, chronic sleep restriction moved glycogen granules closer to the synaptic cleft. Thus, both short and long wake lead to increased glycogen turnover around cortical synapses, whereas sleep promotes glycogen accumulation.
Collapse
Affiliation(s)
- Michele Bellesi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States.,Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | - Luisa de Vivo
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States
| | - Samuel Koebe
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States
| | - Giulio Tononi
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
25
|
Allada R, Cirelli C, Sehgal A. Molecular Mechanisms of Sleep Homeostasis in Flies and Mammals. Cold Spring Harb Perspect Biol 2017; 9:a027730. [PMID: 28432135 PMCID: PMC5538413 DOI: 10.1101/cshperspect.a027730] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Sleep is homeostatically regulated with sleep pressure accumulating with the increasing duration of prior wakefulness. Yet, a clear understanding of the molecular components of the homeostat, as well as the molecular and cellular processes they sense and control to regulate sleep intensity and duration, remain a mystery. Here, we will discuss the cellular and molecular basis of sleep homeostasis, first focusing on the best homeostatic sleep marker in vertebrates, slow wave activity; second, moving to the molecular genetic analysis of sleep homeostasis in the fruit fly Drosophila; and, finally, discussing more systemic aspects of sleep homeostasis.
Collapse
Affiliation(s)
- Ravi Allada
- Department of Neurobiology, Northwestern University, Evanston, Ilinois 60208
| | - Chiara Cirelli
- Department of Psychiatry, University of Wisconsin-Madison, Madison, Wisconsin 53719
| | - Amita Sehgal
- Department of Neuroscience, Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pennsylvania 19104-6058
| |
Collapse
|
26
|
Sleep Loss Promotes Astrocytic Phagocytosis and Microglial Activation in Mouse Cerebral Cortex. J Neurosci 2017; 37:5263-5273. [PMID: 28539349 DOI: 10.1523/jneurosci.3981-16.2017] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/23/2017] [Accepted: 04/13/2017] [Indexed: 11/21/2022] Open
Abstract
We previously found that Mertk and its ligand Gas6, astrocytic genes involved in phagocytosis, are upregulated after acute sleep deprivation. These results suggested that astrocytes may engage in phagocytic activity during extended wake, but direct evidence was lacking. Studies in humans and rodents also found that sleep loss increases peripheral markers of inflammation, but whether these changes are associated with neuroinflammation and/or activation of microglia, the brain's resident innate immune cells, was unknown. Here we used serial block-face scanning electron microscopy to obtain 3D volume measurements of synapses and surrounding astrocytic processes in mouse frontal cortex after 6-8 h of sleep, spontaneous wake, or sleep deprivation (SD) and after chronic (∼5 d) sleep restriction (CSR). Astrocytic phagocytosis, mainly of presynaptic components of large synapses, increased after both acute and chronic sleep loss relative to sleep and wake. MERTK expression and lipid peroxidation in synaptoneurosomes also increased to a similar extent after short and long sleep loss, suggesting that astrocytic phagocytosis may represent the brain's response to the increase in synaptic activity associated with prolonged wake, clearing worn components of heavily used synapses. Using confocal microscopy, we then found that CSR but not SD mice show morphological signs of microglial activation and enhanced microglial phagocytosis of synaptic elements, without obvious signs of neuroinflammation in the CSF. Because low-level sustained microglia activation can lead to abnormal responses to a secondary insult, these results suggest that chronic sleep loss, through microglia priming, may predispose the brain to further damage.SIGNIFICANCE STATEMENT We find that astrocytic phagocytosis of synaptic elements, mostly of presynaptic origin and in large synapses, is upregulated already after a few hours of sleep deprivation and shows a further significant increase after prolonged and severe sleep loss, suggesting that it may promote the housekeeping of heavily used and strong synapses in response to the increased neuronal activity of extended wake. By contrast, chronic sleep restriction but not acute sleep loss activates microglia, promotes their phagocytic activity, and does so in the absence of overt signs of neuroinflammation, suggesting that like many other stressors, extended sleep disruption may lead to a state of sustained microglia activation, perhaps increasing the brain's susceptibility to other forms of damage.
Collapse
|
27
|
Murata E, Mohri I, Kato-Nishimura K, Iimura J, Ogawa M, Tachibana M, Ohno Y, Taniike M. Evaluation of behavioral change after adenotonsillectomy for obstructive sleep apnea in children with autism spectrum disorder. RESEARCH IN DEVELOPMENTAL DISABILITIES 2017; 65:127-139. [PMID: 28514706 DOI: 10.1016/j.ridd.2017.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/17/2017] [Accepted: 04/20/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND AND OBJECTIVE Obstructive sleep apnea (OSA) may affect daily cognitive functioning in children. The aims of our study were two-fold. The first aim was to detect, using the Child Behavior Checklist (CBCL), whether adenotonsillectomy (AT) for the treatment of OSA improved the behavior of children with autism spectrum disorder (ASD). The second aim was to identify characteristics for behavioral improvement following the treatment of OSA in these children with ASD. METHODS The behaviors of ASD children aged 5-14 years diagnosed as having OSA (n=30) were evaluated using CBCL before and after AT. CBCL evaluation of ASD children without OSA at two time points with the same interval served as a control (n=24). We statistically examined the two groups. In addition, we conducted a paired t-test to assess changes in CBCL Tscores between the improved group and unchanged/deteriorated group to identify characteristics that may affect behavioral changes following OSA treatment. RESULTS After AT, T-scores of the CBCL scales were significantly improved in the OSA group, but no change was observed in the control. A paired t-test revealed that the improved group had significantly higher scores on the CBCL pre-AT than the unchanged/deteriorated group in ASD children with OSA after OSA treatment. CONCLUSIONS Behavioral problems were significantly improved following AT in ASD children with OSA. Early detection and treatment of children with OSA is essential to prevent behavioral problems and to support mental development.
Collapse
Affiliation(s)
- Emi Murata
- Department of Child Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Ikuko Mohri
- Department of Child Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan; Department of Pediatrics, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan.
| | - Kumi Kato-Nishimura
- Department of Child Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan; Ota Memorial Sleep Center, 1-50 Nisshincho, Kawasaki, Kawasaki-ku, Kanagawa, 2100024, Japan
| | - Jiro Iimura
- Department of Otorhinolaryngology, Ota General Hospital, 1-50 Nisshincho, Kawasaki, Kawasaki-ku, Kanagawa, 2100024, Japan
| | - Makoto Ogawa
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Masaya Tachibana
- Department of Child Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan; Department of Pediatrics, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Yuko Ohno
- Department of Mathematical Health Science, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| | - Masako Taniike
- Department of Child Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan; Department of Pediatrics, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 5650871, Japan
| |
Collapse
|
28
|
Zhao Z, Zhao X, Veasey SC. Neural Consequences of Chronic Short Sleep: Reversible or Lasting? Front Neurol 2017; 8:235. [PMID: 28620347 PMCID: PMC5449441 DOI: 10.3389/fneur.2017.00235] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 05/12/2017] [Indexed: 12/22/2022] Open
Abstract
Approximately one-third of adolescents and adults in developed countries regularly experience insufficient sleep across the school and/or work week interspersed with weekend catch up sleep. This common practice of weekend recovery sleep reduces subjective sleepiness, yet recent studies demonstrate that one weekend of recovery sleep may not be sufficient in all persons to fully reverse all neurobehavioral impairments observed with chronic sleep loss, particularly vigilance. Moreover, recent studies in animal models demonstrate persistent injury to and loss of specific neuron types in response to chronic short sleep (CSS) with lasting effects on sleep/wake patterns. Here, we provide a comprehensive review of the effects of chronic sleep disruption on neurobehavioral performance and injury to neurons, astrocytes, microglia, and oligodendrocytes and discuss what is known and what is not yet established for reversibility of neural injury. Recent neurobehavioral findings in humans are integrated with animal model research examining long-term consequences of sleep loss on neurobehavioral performance, brain development, neurogenesis, neurodegeneration, and connectivity. While it is now clear that recovery of vigilance following short sleep requires longer than one weekend, less is known of the impact of CSS on cognitive function, mood, and brain health long term. From work performed in animal models, CSS in the young adult and short-term sleep loss in critical developmental windows can have lasting detrimental effects on neurobehavioral performance.
Collapse
Affiliation(s)
- Zhengqing Zhao
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiangxiang Zhao
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Sigrid C Veasey
- Center for Sleep and Circadian Neurobiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
29
|
Teicher MH, Ohashi K, Khan A, Hernandez Garcia LC, Klengel T, Anderson CM, Silveri MM. Does sleep disruption mediate the effects of childhood maltreatment on brain structure? Eur J Psychotraumatol 2017; 8:1450594. [PMID: 29844885 PMCID: PMC5965034 DOI: 10.1080/20008198.2018.1450594] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 02/27/2018] [Indexed: 02/01/2023] Open
Abstract
Background: Childhood maltreatment is associated with alterations in morphology of stress susceptible brain regions. Maltreatment is also known to markedly increase risk for psychopathology and to have an enduring disruptive effect on sleep. Objective: To determine whether abnormalities in sleep continuity have effects on brain morphometry and to evaluate the extent to which sleep impairments mediate the effects of maltreatment on brain structure. Method: Maltreatment and Abuse Chronology of Exposure (MACE) scale ratings, actigraph-assessed sleep and 3T MRI were obtained on N = 37 18-19-year-old participants recruited from the community (N = 34 with neuroimaging). Results: Fourteen participants had no history of maltreatment while N = 23 were exposed, on average, to 4.7 types of maltreatment. Multiplicity of maltreatment was strongly associated with reduced sleep efficiency, increased wake after sleep onset time and number/duration of awakenings, which were independent of effects of maltreatment on depression and anxiety. The most important predictors of impaired sleep were exposure to parental non-verbal emotional abuse at 9-10 years of age. Reduced sleep efficiency correlated with reduced grey matter volume in hippocampus including CA1 subfield, molecular layer and dentate gyrus as well as inferior frontal gyrus and insula. Sleep mediated 39-46% of the effects of maltreatment on volume of hippocampal structures and inferior frontal gyrus. Conclusions: Actigraph-assessed sleep is disrupted in maltreated late teens and mediates a significant portion of the effects of maltreatment on hippocampal volume. Studies are needed to assess whether efforts to enhance sleep in maltreated children can pre-empt or ameliorate neurobiological consequences of maltreatment.
Collapse
Affiliation(s)
- Martin H Teicher
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.,Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| | - Kyoko Ohashi
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.,Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| | - Alaptagin Khan
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.,Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| | - Laura C Hernandez Garcia
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.,Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| | - Torsten Klengel
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.,Neurobiology of Fear Laboratory, McLean Hospital, Belmont, MA, USA
| | - Carl M Anderson
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.,Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA.,Brain Imaging Center, McLean Hospital, Belmont, MA, USA
| | - Marisa M Silveri
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.,Brain Imaging Center, McLean Hospital, Belmont, MA, USA.,Neurodevelopmental Laboratory on Addictions and Mental Health, McLean Hospital, BelmontMA, USA
| |
Collapse
|
30
|
Contribution of sleep to the repair of neuronal DNA double-strand breaks: evidence from flies and mice. Sci Rep 2016; 6:36804. [PMID: 27830758 PMCID: PMC5103291 DOI: 10.1038/srep36804] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 10/21/2016] [Indexed: 12/22/2022] Open
Abstract
Exploration of a novel environment leads to neuronal DNA double-strand breaks (DSBs). These DSBs are generated by type 2 topoisomerase to relieve topological constrains that limit transcription of plasticity-related immediate early genes. If not promptly repaired, however, DSBs may lead to cell death. Since the induction of plasticity-related genes is higher in wake than in sleep, we asked whether it is specifically wake associated with synaptic plasticity that leads to DSBs, and whether sleep provides any selective advantage over wake in their repair. In flies and mice, we find that enriched wake, more than simply time spent awake, induces DSBs, and their repair in mice is delayed or prevented by subsequent wake. In both species the repair of irradiation-induced neuronal DSBs is also quicker during sleep, and mouse genes mediating the response to DNA damage are upregulated in sleep. Thus, sleep facilitates the repair of neuronal DSBs.
Collapse
|
31
|
Effects of Chronic Sleep Restriction during Early Adolescence on the Adult Pattern of Connectivity of Mouse Secondary Motor Cortex. eNeuro 2016; 3:eN-NWR-0053-16. [PMID: 27351022 PMCID: PMC4913218 DOI: 10.1523/eneuro.0053-16.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/14/2016] [Accepted: 04/18/2016] [Indexed: 11/29/2022] Open
Abstract
Cortical circuits mature in stages, from early synaptogenesis and synaptic pruning to late synaptic refinement, resulting in the adult anatomical connection matrix. Because the mature matrix is largely fixed, genetic or environmental factors interfering with its establishment can have irreversible effects. Sleep disruption is rarely considered among those factors, and previous studies have focused on very young animals and the acute effects of sleep deprivation on neuronal morphology and cortical plasticity. Adolescence is a sensitive time for brain remodeling, yet whether chronic sleep restriction (CSR) during adolescence has long-term effects on brain connectivity remains unclear. We used viral-mediated axonal labeling and serial two-photon tomography to measure brain-wide projections from secondary motor cortex (MOs), a high-order area with diffuse projections. For each MOs target, we calculated the projection fraction, a combined measure of passing fibers and axonal terminals normalized for the size of each target. We found no homogeneous differences in MOs projection fraction between mice subjected to 5 days of CSR during early adolescence (P25–P30, ≥50% decrease in daily sleep, n=14) and siblings that slept undisturbed (n=14). Machine learning algorithms, however, classified animals at significantly above chance levels, indicating that differences between the two groups exist, but are subtle and heterogeneous. Thus, sleep disruption in early adolescence may affect adult brain connectivity. However, because our method relies on a global measure of projection density and was not previously used to measure connectivity changes due to behavioral manipulations, definitive conclusions on the long-term structural effects of early CSR require additional experiments.
Collapse
|