Abstract
In countries around the world, sleep deprivation represents a widespread problem affecting school age children, teenagers and adults. Acute sleep deprivation and more chronic sleep restriction adversely affect individual health, impairing memory and cognitive performance as well as increasing the risk and progression of numerous diseases. In mammals, the hippocampus and hippocampus-dependent memory is vulnerable to the effects of acute sleep deprivation. Sleep deprivation induces changes in molecular signaling, gene expression and may cause changes in dendritic structure in neurons. Genome wide studies have shown that acute sleep deprivation alters gene transcription, although the pool of genes affected varies between brain regions. More recently, advances in research have drawn attention to differences in gene regulation between the level of the transcriptome compared with the pool of mRNA associated with ribosomes for protein translation following sleep deprivation. Thus, in addition to transcriptional changes, sleep deprivation also affects downstream processes to alter protein translation. In this review, we focus on the multiple levels through which acute sleep deprivation impacts gene regulation, highlighting potential post-transcriptional and translational processes that may be affected by sleep deprivation. Understanding the multiple levels of gene regulation impacted by sleep deprivation is essential for future development of therapeutics that may mitigate the effects of sleep loss.
Keywords: sleep deprivation, ribosome, memory, hippocampus, translation, gene expression
1. Introduction
Sleep deprivation represents a widespread public health problem in the United States as well as other countries worldwide (Hafner et al. 2017). In the U.S., more than half of school age children get insufficient sleep on a nightly basis (Tsao et al. 2021). In adolescents and teenagers, the problems of sleep deprivation affect approximately 70% of individuals (Center for Disease Control and Prevention, 2017 (Centers for Disease & Prevention 2009; Wheaton et al. 2018). In population models, approximately 27% of adults experience mild sleep restriction sleeping less than 7 hours nightly with an additional 18% sleeping less than 6 hours (Hafner et al. 2017). In addition to problems with chronic sleep restriction, approximately 70% of U.S. adults experience acute sleep deprivation at least one night per month with more than 30% of adults experiencing sleep deprivation on a regular basis (Centers for Disease & Prevention 2009). The pervasiveness of individuals experiencing regular sleep loss is seen worldwide. In the Netherlands, 43.2% of adults reported insufficient sleep in a national survey (Kerkhof 2017), while in Italy, 29.5% of adults reported insufficient sleep (Varghese et al. 2020). In the United Kingdom, approximately 16% of adults sleep less than six hours per night with another 19% receiving six to seven hours sleep per night (Hafner et al. 2017). In Australia, more than one-third of adults and half of teenagers experience inadequate sleep (Adams 2016; AIHW 2021). Many countries in Asia also have high proportions of individuals experiencing sleep deprivation with Japan often considered the world’s most sleep deprived country with 56% of adults in Japan estimated as receiving inadequate sleep (Hafner et al. 2017; Kakamu et al. 2021). Although studies of sleep from African countries are more limited, recent studies suggest that insufficient sleep and poor sleep quality is also a problem across age groups with studies highlighting sleep deprivation in older adults and in university students (Lankrew Ayalew 2022; Nsengimana et al. 2023; Stranges et al. 2012; Wang et al. 2019). Despite the growing number of population studies on sleep and sleep disorders across multiple countries, the question of whether the incidence of sleep deprivation among individuals is increasing can be difficult to answer as many large-scale studies rely upon self-reported or subjective sleep tests and the definitions of insufficient sleep can vary. However, the pervasiveness of sleep deprivation across age groups and societies necessitates increased understanding of the impacts of sleep loss.
Sleep deprivation results in significant decrements in cognition and adversely impacts individual health. Sleep disorders increase the incidence of metabolic diseases, cardiac diseases, cancer, as well as neuropsychiatric and mood disorders. Sleep and circadian rhythm disorders have also been linked to the increased incidence and accelerated progression of neurodegenerative diseases including Alzheimer’s disease (Sabia et al. 2021; Shi et al. 2018; Wang & Holtzman 2020; Wu et al. 2019). At a societal level, economic losses attributed to sleep loss due to lower workplace productivity, increased disease incidence and higher mortality risks, result in approximate annual costs in the U.S. between $280 to $411 billion with this range expected to increase to $318 to $456 billion by 2030 (Hafner et al. 2017). The cumulative costs for the economic impacts of sleep loss are predicted to be between 4 to 7 trillion dollars for the U.S. alone by 2030 (Hafner et al. 2017). In Japan, the economic impacts of sleep deprivation are estimated at greater than $138 billion annually. In Germany and the United Kingdom, economic losses due to sleep deprivation are between $50 - $60 billion annually (Hafner et al. 2017). In Australia, the annual economic costs for sleep deprivation are considered between $35 to 45 billion (Hillman et al. 2018; Streatfeild et al. 2021). Given the serious consequences of sleep loss to individuals and society, it is important to understand the behavioral, cellular and molecular consequences of sleep deprivation, particularly those processes affecting memory and performance.
2. Consequences of Sleep Deprivation
Sleep deprivation disrupts numerous physiological processes causing stress on many tissues and organs. In this section, we focus on the impacts of sleep deprivation on memory and the underlying molecular and neural changes induced by sleep deprivation (Figure 1).
Figure 1. Overview of the impacts of sleep deprivation on hippocampus dependent memory and examples of molecular pathways that are affected.
2.1. Memory
The adverse impacts of acute sleep deprivation on memory and performance are phylogenetically conserved with associative memory affected from invertebrates to humans. Table 1 provides examples of the impacts of sleep deprivation on memory across species. In Drosophila and honeybees, sleep deprivation results in deficits in memory consolidation ((Hussaini et al. 2009), particularly spatial memory in bees (Beyaert et al. 2012). Moreover, extended wake (sleep deprivation) in Drosophila decreases basal and induced neuronal activity (Bushey et al. 2015). In the marine mollusk, Aplysia californica, sleep deprivation prior to training inhibits both short and long-term associative memory (Krishnan et al. 2016a; Krishnan et al. 2016b). Sleep deprivation in zebrafish results in deficits in object discrimination (Pinheiro-da-Silva et al. 2017a), spatial learning (Pinheiro-da-Silva et al. 2017b), and conditioned avoidance (Pinheiro-da-Silva et al. 2018). In mammals, the effects of acute sleep deprivation are particularly apparent for hippocampus dependent memory (reviewed in (Havekes et al. 2015; Kreutzmann et al. 2015). Acute sleep deprivation inhibits the consolidation of hippocampus-dependent long-term memory for spatial object recognition, contextual fear conditioning, and spatial memory mice and rats (Graves et al. 2003; Guan et al. 2004; Hagewoud et al. 2010; Xu et al. 2010). In fact, sleep deprivation induces the use of alternate learning strategies to bypass hippocampus-dependent memory such as seen with spatial learning for food reward, although memory impairments are still observed in more complex reversal learning (Hagewoud et al. 2010). Furthermore, acute sleep deprivation inhibits the subsequent induction of hippocampal long-term potentiation (LTP) and heterosynaptic plasticity through synaptic tagging as seen in rodent studies (Vecsey et al. 2009; Vecsey et al. 2018; Wong et al. 2019). REM sleep deprivation also induces deficits in hippocampal LTP (Davis et al. 2003; McDermott et al. 2003). Three days of REM sleep deprivation in rats impairs hippocampus dependent memory with contextual fear conditioning, but had no effect on amygdala dependent cued fear conditioning (McDermott et al. 2003).
Table 1.
Examples of the impacts of sleep deprivation on memory from invertebrates to humans
| Findings related to memory | Method and length of sleep deprivation | Organism | References |
|---|---|---|---|
| Deficits in memory consolidation and spatial memory | Bees were intermittently sleep deprived (5 min sleep dep, 5 min rest) during the entire light and dark phase | Foraging honeybees (Apis mellifera carnica) |
Hussaini et al. 2009
Beyaert et al. 2012 |
| Decreases basal and induced neuronal activity | 6, 12, and 24 hours of sleep deprivation in rotating box | Drosophila | Bushey et al. 2015 |
| Inhibits short and long-term associative memory | 9 hours of sleep deprivation using context changes and tactile stimulation | Marine mollusk (Aplysia californica) |
Krishnan et al. 2016a
Krishnan et al. 2016b |
| Deficits in object discrimination, spatial learning and conditioned avoidance | 6 hours of sleep deprivation by extending the light phase or brief light pulses during the dark phase | Zebrafish (Danio rerio) |
Pinheiro-da-Silva et al. 2017a
Pinheiro-da-Silva et al. 2017b Pinheiro-da-Silva et al. 2018 |
| Impairs memory consolidation for contextual fear conditioning | 5 hours of sleep deprivation by gentle stroking | Mus musculus | Graves et al. 2003 |
| Impairs spatial working memory | 6 or 12 hours of sleep deprivation | Hagewoud et al. 2010 | |
| Impairs spatial learning and memory | 3 hours of sleep deprivation for 30 consecutive days by gentle touch with a brush | Xu et al. 2010 | |
| Inhibits the induction of hippocampal long-term potentiation and heterosynaptic plasticity | 5 hours of sleep deprivation by gentle handling |
Vecsey et al. 2009
Vecsey et al. 2018 Wong et al. 2019 |
|
| Inhibits the consolidation of hippocampus-dependent long-term memory for spatial memory | 6 hours of sleep deprivation by placing novel objects in the animal’s cage. | Rattus norvegicus | Guan et al. 2004 |
| Impairs hippocampus dependent memory contextual fear conditioning | REM sleep deprivation on single small platform in the middle of a water tank for 72 h. | McDermott et al. 2003 | |
| Decreases hippocampal activity during episodic memory training using visual stimuli, resulting in deficits in memory encoding | One night of sleep deprivation by waking activity (Internet, e-mail, short walks, reading and board games) | Homo sapiens | Yoo et al. 2007 |
| Alteration in the connectivity of hippocampal networks | Kaufmann et al. 2016 | ||
| Deficits in declarative memory (word-pair task) | Kuhn et al. 2016 | ||
| Neural network remodeling | Pesoli et al. 2022 | ||
| Decreases activation of prefrontal cortex, posterior parietal cortex and supplementary motor area | Extended sleep deprivation | Mu et al. 2005 |
In humans, fMRI studies have been used to evaluate the effects of sleep deprivation on brain activity. One night of sleep deprivation significantly decreases hippocampal activity during episodic memory training using visual stimuli, resulting in deficits in memory encoding (Yoo et al. 2007). Interestingly, while hippocampal activity necessary for memory was impaired, alternate networks were strengthened including coupling between the hippocampus and basic vigilance networks in the upper brainstem and thalamus (Yoo et al. 2007). More recent fMRI imaging assessments of hippocampal impairments following sleep deprivation found that sleep deprivation altered the connectivity of hippocampal networks (Kaufmann et al. 2016). These results are reminiscent of the bypass of hippocampal learning networks seen in rodent studies. One night of sleep deprivation also causes significant deficits in declarative memory using a word-pair task often used to assess hippocampus-cortical network function (Kuhn et al. 2016). Thus, in humans as well as in animal models, the hippocampus appears particularly vulnerable to the effects of sleep deprivation. Sleep deprivation also affects the cortex with decreases in LTP-like plasticity observed; however, baseline cortical excitability and synaptic strength were increased after sleep deprivation (Kuhn et al. 2016). fMRI studies have also shown that extended sleep deprivation decreases activation of parts of the prefrontal cortex, the posterior parietal cortex and the supplementary motor area following a working memory task (Mu et al. 2005). Studies using magnetoencephalography to evaluate functional connectivity found that brain networks underwent widespread remodeling after sleep deprivation, potentially as a mechanism to redistribute cognitive load (Pesoli et al. 2022). Sleep deprivation also reduces connectivity and induces alterations within the dorsal attention network, the visual network and the default mode network (Gao et al. 2015; Kaufmann et al. 2016).
2.2. Molecular Signaling
At the molecular level, probably the most well-studied pathway affected by sleep deprivation is the cAMP/PKA pathway. Acute sleep deprivation inhibits cAMP/PKA signaling in the hippocampus decreasing cAMP levels (Vecsey et al. 2009; Wong et al. 2019). Increasing cAMP levels through pharmacological inhibition of phosphodiesterases counters the effects of sleep deprivation restoring synaptic plasticity and enabling the molecular consolidation of long-term memory after sleep deprivation (Vecsey et al. 2009). Moreover, if cAMP is specifically increased only in excitatory neurons during sleep deprivation, the adverse impact of sleep deprivation on long-term spatial memory is ameliorated (Havekes et al. 2014). Downstream the cAMP/PKA signaling results in the phosphorylation of the transcription factor CREB, a transcription factor necessary for many forms of long-term memory. Acute sleep deprivation decreases basal levels of pCREB in the hippocampus as well as learning induced phosphorylation of CREB (Vecsey et al. 2009; Wong et al. 2019; Xu et al. 2010). The cAMP/PKA pathway is also targeted during chronic REM sleep deprivation. Multiple days of REM sleep deprivation depresses cAMP levels in the hippocampus and impairs long-term spatial memory in the Morris water maze (Maher et al. 2021). As with acute sleep deprivation, administration of phosphodiesterase inhibitors mitigates the impact of chronic sleep deprivation on spatial memory (Maher et al. 2021). The impact of sleep deprivation on cAMP/PKA signaling is phylogenetically conserved. In zebrafish, the expression of PKA and CREB are reduced after sleep deprivation (Lee et al. 2020). Moreover, after a fear conditioning avoidance task, sleep deprivation reduced the training induced nuclear increases in PKA and CREB and decreased CREB DNA binding (Lee et al. 2020).
Interestingly, cAMP/PKA signaling also functions in the regulation of wakefulness and sleep homeostasis. Reduction in PKA activity results in increased sleep fragmentation and decreased sleep rebound after sleep deprivation (Hellman et al. 2010). CREB also appears to function in wakefulness as the deletion of Creb1 in forebrain excitatory neurons increases sleep, yet significantly decreases rebound sleep following sleep deprivation (Wimmer et al. 2021). In Drosophila, the cAMP/PKA has been shown to play a key role in regulating sleep homeostasis (Hendricks et al. 2001). Thus, the cAMP/PKA pathway and CREB dependent transcription have evolutionarily conserved roles in the regulation of sleep/wake activity and homeostatic sleep needs.
Sleep deprivation also affects other kinase signaling pathways including the extracellular signal-regulated kinase (ERK) pathway. Six hours of total sleep deprivation has been shown to reduce ERK activity in the hippocampus of rats (Guan et al. 2004) and mice (Wong et al. 2019). Furthermore, chronic REM sleep deprivation also decreases hippocampal levels of phospho-ERK1/2 (Ravassard et al. 2009). Changes in ERK signaling are also seen after sleep deprivation in Drosophila (Vanderheyden et al. 2013). Thus, a significant impact of sleep deprivation appears to be through changes in MAPK signaling.
Brain derived neurotrophic factor (BDNF) is a key regulator of both presynaptic and post-synaptic plasticity, and is highly expressed in the hippocampus (Edelmann et al. 2014; Lin et al. 2018; Wang et al. 2022). Consequently, the effects of sleep deprivation on BDNF have also been well-studied in animal models and in humans (reviewed in (Rahmani et al. 2020). Early research found that acute REM sleep deprivation in rats decreased BDNF levels in the cerebellum and the brainstem, but not the hippocampus (Sei et al. 2000). However, total sleep deprivation reduces BDNF in hippocampus (Alkadhi & Alhaider 2016). Five hours of acute sleep deprivation in mice reduces both the pro-BDNF form that occurs prior to cleavage and mature BDNF in the hippocampus (Wong et al. 2019). Longer periods of sleep restriction also reduce BDNF RNA and protein levels (Guzman-Marin et al. 2006). When sleep deprivation occurs post-learning after training for hippocampus dependent learning in the Morris Water Maze, BDNF mRNA expression is decreased (Karabulut et al. 2019). Interestingly, the effects of sleep deprivation on BDNF signaling appear to occur through its low affinity receptor, the p75 neurotrophin receptor (p75NTR). The effects of acute sleep deprivation are alleviated with restoration seen in the levels of ERK phosphorylation, pCREB and BDNF in p75NTR knockout mice (Wong et al. 2019). In humans, one night of sleep deprivation decreases BDNF levels in plasma (Kuhn et al. 2016). The effects of acute and chronic sleep deprivation on cellular signaling mechanisms appear widespread, potentially affecting numerous downstream targets and multiple cellular processes (Table 2).
Table 2.
Sleep deprivation induces changes in molecular signaling
| Molecular changes | Method and length of sleep deprivation | Organism | References |
|---|---|---|---|
| Inhibits cAMP/PKA signaling in the hippocampus (decrease cAMP and pCREB levels). Reduces ERK activity and both pro-BDNF and mature BDNF in the hippocampus. |
5 hours of sleep deprivation by gentle handling | Mus musculus |
Vecsey et al. 2009
Xu et al. 2010 Wong et al. 2019 |
| Reduces cAMP levels in the hippocampus | REM sleep for 12 hours a day for six consecutive days | Maher et al. 2021 | |
| Reduction in PKA activity results in increased sleep fragmentation and decreased sleep | 6 hours of sleep deprivation by gentle handling | Hellman et al. 2010 | |
| Deletion of Creb in forebrain excitatory neurons increases sleep, but decreases rebound sleep | Wimmer et al. 2021 | ||
| BDNF mRNA expression is decreased | 3 hours of REM sleep deprivation post-learning (Morris Water Maze) | Karabulut et al. 2019 | |
| Reduction of PKA and CREB levels | One night sleep deprivation | Zebrafish (Danio rerio) | Lee et al. 2020 |
| cAMP/PKA regulates sleep homeostasis | 6 hours of sleep deprivation by mechanical stimuli | Drosophila | Hendricks et al. 2001 |
| Increased ERK signaling increases sleep; sleep deprivation increases ERK signaling potentially to induce rebound sleep | 12 hours of sleep deprivation | Vanderheyden et al. 2013 | |
| Reduction of ERK activity in the hippocampus | 6 hours of sleep deprivation | Sprague Dawley rat | Guan et al. 2004 |
| Decreases hippocampal levels of phospho-ERK1/2 | Chronic REM sleep deprivation for 72 hours on multiple platforms in a standard container filled with water | Sprague Dawley rat | Ravassard et al. 2009 |
| Reduction of BDNF RNA and protein levels | Treadmill method (3s on/12s off) for 48 hours | Guzman-Marin et al. 2006 | |
| Decreases BDNF levels in the cerebellum and the brainstem | 6 hours of REM sleep deprivation | Wistar rat | Sei et al. 2000 |
| Reduction of BDNF in the hippocampus | 24 hours of sleep deprivation in the “Modified Multiple Platforms” paradigm | Alkadhi & Alhaider 2016 | |
| Decreases BDNF levels in plasma | One night of total sleep deprivation with standardized activities | Homo sapiens | Kuhn et al. 2016 |
2.3. Morphological Changes
Although the impacts of sleep deprivation on behavior, memory, network connectivity and cellular signaling appear striking, the evidence is less clear regarding morphological changes in neurons (Table 3). Long-term memory and synaptic plasticity are associated with dynamic regulation of dendritic spine number and synapses. Sleep/wake cycles and the circadian clock regulate synapse number in invertebrates and vertebrate model systems resulting in changes in synaptic strength (Appelbaum et al. 2010; Bushey et al. 2011; Fernandez et al. 2008; Maret et al. 2011; Mehnert et al. 2007). In Drosophila, rhythmic variations are observed for the synapses of circadian pacemaker neurons with peaks occurring during the day and troughs at night for axon diameter, dendritic branching, synapse size and synapse number (Fernandez et al. 2008; Mehnert et al. 2007; Mehnert & Cantera 2011). Sleep/wake cycles also regulate synaptic transmission with changes in synapse number increased when flies are awake and decreased during sleep (Bushey et al. 2011). Similarly, in zebrafish, overall synapse number is reduced during the night, but interestingly, inhibitory synapse number is increased (Appelbaum et al. 2010; Elbaz et al. 2017). Dendritic spine number also appears to be regulated by sleep/wake cycles in adolescent mice with decreased spine number seen during sleep (Maret et al. 2011). Thus, synaptic transmission is regulated across species by sleep/wake cycles. More recent studies have also shown that some dendritic spines are reduced in size during sleep in mice, along with the elimination of some spines (de Vivo et al. 2017; Li et al. 2017). Thus, sleep regulates dendritic and spine morphology across species.
Table 3.
Examples of the impacts of sleep deprivation on neuronal structure
| Morphological findings | Method and length of sleep deprivation | Organism | References |
|---|---|---|---|
| Increases overall synapse number | 6 hours of sleep deprivation by vibration | Zebrafish | Appelbaum et al. 2010 |
| Decreases the number of inhibitory synapses | 24 hours of sleep deprivation with constant light | Elbaz et al. 2017 | |
| Golgi staining: Decreased spine number in the CA1 and dentate gyrus | 5 hours of sleep deprivation by gentle handling | Mus musculus |
Havekes et al. 2016
Raven et al. 2019 Wong et al. 2019 |
| Genetic fluorescent labeling: increased total spine number (thin spines) and spine volume. | Gisabella et al. 2020 | ||
| Branch-specific effects on spine density in the CA1 | Bolsius et al. 2022 | ||
| No effect on spine number or the shape of the dendritic tree in CA1 neurons – Animals group housed with enrichment prior to sleep deprivation | Brodin et al. 2022 | ||
| Branch specific reduction in dendritic spines in cortex | 7 hours of sleep deprivation by gentle handling after motor learning task | Yang et al. 2014 | |
| Reduction in training-induced new spines in adolescent mice | 7 hours of sleep deprivation by gentle handling after motor learning task | Adler et al. 2021 | |
| Decreased spine density in the dorsal hippocampus | 6 hours of sleep deprivation for 21 consecutive days | Huang et al. 2022 | |
| Synapses are strengthened when awake and weakened during sleep | Wake/sleep cycle | de Vivo et al. 2017 | |
| REM sleep prunes new dendritic spines in the motor cortex | Sleep | Li et al. 2017 | |
| Decreased dendritic spine number in the CA1 of the hippocampus | 24 hours of total sleep deprivation | Rattus norvegicus | Acosta-Pena et al. 2015 |
It has been more difficult to discern the effects of sleep deprivation on neuron structure and synaptic morphology, as considerable variation occurs depending upon the type of neuron and the brain region studied. In zebrafish, six hours of sleep deprivation, but not shorter periods, has been shown to increase overall synapse number (Appelbaum et al. 2010). However, more recent research found that sleep deprivation decreases the number of inhibitory synapses (Elbaz et al. 2017). Thus, inhibitory synapse number is increased during sleep, but in sleep deprivation, inhibitory neurotransmission is weakened. In mammalian research, differences between studies, may arise due to the techniques used to assess synapse number, the neurons examined or even the relative proximity of the dendrites to neuronal cell bodies. In mice, studies using Golgi staining to visualize dendritic branches and spines found that five hours of acute sleep deprivation decreases spine number in the CA1 and dentate gyrus (Havekes et al. 2016; Raven et al. 2019; Wong et al. 2019). In rats, twenty-four hours of sleep deprivation also decreased dendritic spine number in the CA1 of the hippocampus (Acosta-Pena et al. 2015). In contrast, recent research using genetic fluorescent labeling to visualize dendritic spines in excitatory CA1 neurons determined that five hours of sleep deprivation increased total spine number, primarily through an increase in thin spines, as well as an increase in spine volume (Gisabella et al. 2020). Although methodological differences in spine visualization may account for differences in spine measurements between these studies, it is more likely that differences in assessment of dendritic spines after sleep deprivation between these studies arise from positional variance in spine dynamics across dendritic branches. In-depth analysis of the effect of sleep deprivation on spine density found that sleep deprivation has branch specific effects in the CA1 (Bolsius et al. 2022). A recent study in which mice were group housed with enrichment prior to sleep deprivation found that a single session of acute sleep deprivation had no effect on spine number or the shape of the dendritic tree in CA1 neurons (Brodin et al. 2022). This study perhaps suggests that group housing and enrichment can aid in minimizing the effects of acute sleep deprivation. On the other hand, chronic sleep restriction of young mice for three weeks results in decreased spine density in the dorsal hippocampus (Huang et al. 2022). Branch specificity in spine dynamics and a reduction in spine number after sleep deprivation were previously observed in the primary motor cortex after learning (Yang et al. 2014). Animals trained for a motor learning task followed by seven hours of sleep deprivation showed a branch specific reduction in dendritic spines compared with animals that slept after training (Yang et al. 2014). Similarly, in adolescent mice, sleep after learning promotes spine formation and the development of new synapses in the primary motor cortex, while sleep deprivation reduces the number of learning-induced new spines (Adler et al. 2021).
3. Sleep Deprivation Targets Gene Regulation at Multiple Levels
In addition to impacts on cellular signaling, sleep deprivation also affects gene regulation at multiple levels including changes in chromatin accessibility, transcription, post-transcriptional stability of mRNA and translation. In the next sections, we survey some of the potential mechanisms through which sleep deprivation targets gene regulation. Although research using invertebrate models and non-mammalian vertebrates have substantially contributed to our understanding of molecular signaling, learning and memory, and the functions of sleep; for clarity, we will focus on studies from mammalian models in our discussion of the processes impacted by sleep deprivation in gene regulation.
3.1. Transcription
3.1.1. Tissue Specific Effects of Sleep Deprivation on Transcription
Early research investigating molecular differences between sleep and wake focused on transcriptional approaches and changes in gene expression using microarrays or RNA sequencing to examine differential gene expression in various brain regions with sleep/wake cycles and sleep deprivation. Gene transcription varies with sleep/wake cycles and the subset of regulated genes varies depending upon the brain region or cell type examined (Cirelli et al. 2004; Cirelli & Tononi 2000; Mackiewicz et al. 2007). Estimates suggest that up to 10% of cortical transcripts are regulated with sleep/wake cycles (Scarpa et al. 2018). In the cortex, multiple time points of gene expression data and modeling of transcriptome dynamics suggests that sleep/wake cycles, in particular the length of time awake, drive changes in gene expression (Hor et al. 2019). These studies suggest that changes in gene expression with sleep/wake cycles are intertwined with homeostatic sleep regulation.
Sleep deprivation appears to impact gene transcription with a high degree of regional and cellular specificity within the brain as evidenced by the considerable differences observed between the hippocampus and the cortex. For more in-depth information on the specific genes affected by acute sleep deprivation in the cortex and hippocampus, we refer the reader to the following studies (Cirelli & Tononi 1998; Gaine et al. 2021; Terao et al. 2003; Thompson et al. 2010; Vecsey et al. 2012). While studies of gene changes with sleep deprivation are interesting, on a practical note, there is considerable interest in understanding how these changes in gene expression can be reversed. Most of the studies on recovery sleep have examined gene expression after acute sleep deprivation. In the hippocampus, three hours of recovery sleep is sufficient to return many genes to baseline expression levels following acute sleep deprivation (Gaine et al. 2021; Vecsey et al. 2012). However, there are some genes which maintain aberrant expression even after recovery sleep including the mRNA splicing factor Srsf7 , the G protein coupled serotonin receptor Htr1a, a potassium voltage gated channel subfamily member Kcnv1 and the transcription factor Elk1 (Gaine et al. 2021; Vecsey et al. 2012). The functions of these genes affecting transcription, mRNA splicing and neurotransmission could potentially mediate the persistent effects of acute sleep deprivation. In the cortex, acute sleep deprivation induces differential expression of hundreds of genes, including core circadian clock genes (Gerstner et al. 2016; Hor et al. 2019; Ingiosi et al. 2019). Interestingly, as in the hippocampus, the amount of recovery sleep necessary for gene expression to return to baseline levels varies depending upon the gene examined (Gerstner et al. 2016; Ingiosi et al. 2019). Sleep deprivation also induces differences in gene regulation in non-brain tissues. In humans, two nights of total sleep deprivation induces gene dysregulation in the blood with 212 genes significantly affected, with most genes downregulated (Uyhelji et al. 2018).
3.1.2. Sleep deprivation Induces Epigenetic Changes
Gene transcription depends on the accessibility of the chromatin allowing transcription factors and the transcriptional machinery to bind to the DNA. Epigenetic modifications affecting chromatin structure and DNA accessibility can be affected by sleep deprivation, thus influencing gene transcription (reviewed in (Gaine et al. 2018). DNA methylation in upstream gene regulatory sequences represses gene transcription. In rodents, acute sleep deprivation was found to induce methylation changes in the promoters of more than 136 genes in the cerebral cortex, with both increased and decreased methylation observed (Massart et al. 2014). In humans, one night of sleep deprivation increases DNA methylation in the promoter regions of core circadian clock genes CRY1 and PER1 as seen in adipose tissue (Cedernaes et al. 2015). Acute sleep deprivation has also been shown to change DNA methylation for genes involved in fatty acid metabolism in humans (Skuladottir et al. 2016). A broader study of acute sleep deprivation and DNA methylation in humans using whole blood samples found that sleep deprivation changed methylation in 269 genes (Nilsson et al. 2016). A derived modification of methylation, DNA hydroxymethylation, is also seen in the mammalian brain, with hydroxymethylation occurring more downstream in the body of the gene. Hydroxymethylation has been associated with bidirectional gene regulation, with enhanced expression seen for some genes (Kriaucionis & Heintz 2009; Li & Liu 2011; Meng et al. 2015). Sleep deprivation affects hydroxymethylation in the cortex to a greater degree than methylation, with changes in DNA hydroxymethylation observed for 4697 genes (Massart et al. 2014). Interestingly, in cell cultures, cAMP signaling has been shown to enhance DNA hydroxymethylation providing another potential link through which cAMP/PKA may influence gene transcription (Camarena et al. 2017). Thus, in animal models and humans, acute sleep deprivation alters patterns of DNA methylation affecting gene transcription.
Histone acetylation is another epigenetic modification that has been examined after sleep deprivation, given its importance in memory formation. The acetylation of histone proteins to increase chromatin accessibility constitutes an important regulatory step necessary for the increased gene expression seen with the molecular consolidation of long-term memory and synaptic plasticity (Levenson et al. 2004; McQuown et al. 2011). Histone deacetylase inhibitors, such as Trichostatin A, facilitate hippocampus dependent long-term memory when weak training paradigms are used or even to reverse memory impairments in disease and stress models (Hawk et al. 2011; Pandey et al. 2015; Vargas-Lopez et al. 2016). Given the effects of acute and chronic sleep deprivation on hippocampus dependent long-term memory, researchers investigating histone acetylation found that five hours acute sleep deprivation in mice significantly decreases two prominent histone acetylation marks, H3K9 and H4K12 and significantly increases the abundance of the histone deacetylase, HDAC2 (Wong et al. 2020). Histone deacetylation and HDAC2 activity are associated with memory impairments (reviewed in (Pao & Tsai 2022). Treatment with an FDA approved histone deacetylase inhibitor restored histone acetylation levels and blocked the impairments in LTP and long-term memory induced by sleep deprivation (Wong et al. 2020). Moreover, inhibition of histone deacetylase inhibitors during chronic sleep deprivation also facilitates the consolidation of long-term memory (Duan et al. 2016). In the cerebral cortex, the effects of acute sleep deprivation on gene expression were correlated with chromatin accessibility for 24% of the genes examined (Hor et al. 2019). Changes in chromatin accessibility are detected quickly with sleep deprivation and for the most part, returned to baseline levels by 12 hours following sleep deprivation (Hor et al. 2019). Thus, it appears that one mechanism through which sleep deprivation may impact the molecular consolidation of memory is through chromosome accessibility and histone acetylation regulation of gene transcription.
3.2. Sleep Deprivation Alters mRNA Processing
Following mRNA transcription, newly formed mRNA transcripts undergo maturation and processing. In the nucleus, newly formed mRNA transcripts are subject to splicing and polyadenylation prior to nuclear export. Within the cytoplasm, mRNA stability and lifespan may also be regulated as well as the propensity for translation (reviewed in (Radhakrishnan & Green 2016). In the following sections, we discuss potential mechanisms in post-transcriptional processing that may be affected by sleep deprivation between transcription and translation and the evidence that suggests these processes are targeted (Figure 2).
Figure 2. Post-transcriptional processes impacted by sleep deprivation.
Sleep deprivation affects mRNA splicing, polyadenylation, and miRNA mediated degradation. Sleep deprivation also decreases mTOR signaling resulting in 4EBP mediated inhibition of translation.
3.2.1. Alternative Splicing and Polyadenylation
The human genome is estimated to contain 24,000 protein coding genes resulting in more than 100,000 proteins due to 95% of genes giving rise to multiple transcripts (Keren et al. 2010; Kim et al. 2018; Pan et al. 2008). Alternative splicing facilitates neuronal function by increasing the number of mRNA transcripts arising from a single gene, consequently increasing protein diversity. Additionally, alternative splicing may alter mRNA translation through changes in the stability of mRNA transcripts or decay rates. In neurons, mRNA splicing patterns are dynamically regulated to promote neural plasticity and synaptic function (Vuong et al. 2016). Disease related changes in pre-mRNA splicing and splicing pathologies have been associated with metabolic disorders, cancers, and neurodegenerative diseases including Alzheimer’s disease (reviewed in (Daguenet et al. 2015; Kim et al. 2018; Li et al. 2021a). Circadian regulation of alternative splicing has been shown for multiple tissues (Marcheva et al. 2020; McGlincy et al. 2012), suggesting that sleep/wake cycles may also affect alternative splicing. While alternative splicing of mRNA transcripts following sleep deprivation has not been well studied, technological advances and increased resolution in genome wide RNA sequencing will undoubtedly further our understanding of alternative splicing and sleep in the next few years.
The regulation of mRNA stability occurs, at least in part, via polyadenylation, the addition of adenosine monophosphates to the 3’ terminal end of the RNA transcript that augments RNA stability and increases translation efficiency (reviewed in (Charlesworth et al. 2013). As our understanding of the mechanisms and functions of polyadenylation have grown, disease associated changes in gene expression due to mis-steps in polyadenylation and deadenylation have been identified, underscoring the importance of polyadenylation in gene regulation (Dharmalingam et al. 2022; Ivshina et al. 2014). Circadian rhythms in polyadenylation and deadenylation have been shown in neurons and astrocytes (Gendreau et al. 2018; Gerstner et al. 2012; Kojima et al. 2012; Parnell et al. 2021). Changes in polyadenylation have also been associated with long-term potentiation in the hippocampus (Fontes et al. 2017). Potentially, if sleep deprivation disrupts the circadian clock or core clock genes, then subsequent changes in polyadenylation may affect RNA stability and gene transcription. Interestingly, two genes that are downregulated by sleep deprivation in the hippocampus, Cirbp and one Rbm3 isoform (Gaine et al. 2021) have been implicated in circadian gene expression through the regulation of polyadenylation (Liu et al. 2013). Decreased expression of Cirpb or Rbm3 resulted in the shortening of the 3’UTR for more than 100 genes and subsequent decreased circadian rhythms for many genes (Liu et al. 2013). Additionally, sleep deprivation increases the expression of Srsf7 in the hippocampus, a gene recently reported to have expression inversely correlated with polyA tail length in the 3’ UTR (Gaine et al. 2021; Schwich et al. 2021). However, the regulation of RNA processing and polyadenylation by sleep/wake cycles appears complex and may vary with brain regions. In the cortex and hypothalamus, the expression of polyadenylation factor 2 (Cpsf2) decreases during sleep, suggesting that polyadenylation may decrease with sleep (Mackiewicz et al. 2007). In humans, two nights of sleep deprivation has been shown to decrease the mRNA levels in the blood of cytoplasmic polyadenylation element binding protein CPEBP4, a gene involved in mediating polyA elongation and translation (Uyhelji et al. 2018). Although additional research needs to be done, it is clear that regulation of polyadenylation to affect mRNA stability represents a potential mechanism through which sleep deprivation could post-transcriptionally impact gene regulation.
3.2.2. mRNA stability: Degradation through miRNA
An important mechanism through which gene expression may be regulated is through microRNA (miRNA) mediated degradation of mRNA. miRNA sequences are comprised of 18–25 nucleotides that bind to the 3’ untranslated region (3’ UTR) of a gene resulting in a region of double stranded RNA that is then subject to cleavage and degradation or translational repression (Iwakawa & Tomari 2015; Kim & Pak 2020). There are approximately 2500 miRNAs in the human genome (Alles et al. 2019; Huang et al. 2020); miRTarBase, release 9.0), with studies estimating that more than 60% of protein coding mRNA transcripts are targeted by miRNAs (Friedman et al. 2009). miRNAs are dynamically regulated through transcription and degradation. The half-life of miRNAs can vary, although half-life is usually measured in hours (reviewed in (Kim & Pak 2020). Functionally, miRNAs can be targeted to dendrites and play a prominent role in synaptic plasticity (Kindler & Kreienkamp 2012; Schratt 2009; Song 2020; Steward & Worley 2001).
The regulation of sleep also appears dependent in part on miRNA regulation of gene expression. Pharmacological studies in rats using targeted miRNA inhibitors found that inhibition of miRNA-138 in the cortex suppressed total sleep duration and slow wave sleep during NREM, in particular during the animal’s normal rest phase (Davis et al. 2012). However, the effect of miRNAs on sleep depends upon the miRNA examined and presumably the mRNA transcripts that it targets. Injection of a pre form miR-132 (mimetic), a miRNA known to be involved in memory, decreased NREM sleep and lowered sleep intensity, although REM sleep was increased (Davis et al. 2011). Circadian and/or regulation by sleep/wake cycles occurs for many miRNAs including miR-132 (Aten et al. 2018; Cheng et al. 2007; Davis et al. 2012; Davis et al. 2011). miRNAs have also been shown to play a prominent role in regulating entrainment of the circadian clock in mammals as well as functioning in translational control affecting the period length of the circadian clock (Alvarez-Saavedra et al. 2011; Cheng et al. 2007; Park et al. 2020; Zhou et al. 2021). Circadian regulation of miRNA expression has been observed in plants, fungi, invertebrates and vertebrate animals (reviewed in (Parnell et al. 2021).
Changes in miRNA expression with sleep deprivation could mediate mRNA degradation, thereby affecting gene specific translation. In rats, eight hours of sleep deprivation resulted in changes in 50 miRNAs (Davis et al. 2007). Interestingly, as with gene expression, brain region specific differences were seen in the direction of miRNA regulation (Davis et al. 2007). In the prefrontal and somatosensory cortices, more than 90% of the differentially regulated miRNAs were significantly decreased. However, in the hippocampus the directionality was reversed with the significant upregulation of 49 miRNAs, of which 37 were decreased in the cortex such as miR-132 (Davis et al. 2007). This suggests that miRNAs in the hippocampus play a contributing role in the decreased gene expression seen after sleep deprivation.
During memory consolidation, miRNA regulation is bidirectional and depends upon the specific miRNA and its targets. Downregulation of some miRNAs permits upregulated gene-specific protein translation; whereas, the upregulation of other miRNAs facilitates the degradation of mRNA transcripts for genes that act as limits in memory formation (reviewed in (Aksoy-Aksel et al. 2014; Fiorenza & Barco 2016; Wei et al. 2017). The impairments in long-term memory seen in REM sleep deprivation following training in the Morris water maze are associated with changes in the hippocampal expression of miRNAs known to be related to memory including miR-132, miR-182 and miR-124 (Karabulut et al. 2019). With chronic REM sleep deprivation after Morris water maze training, miRNA-191a and miRNA-155 were found to have increased expression in the hippocampus correlated with impairments in long-term memory (Mohammadipoor-Ghasemabad et al. 2019). Similar to the dysregulation of miRNAs seen with sleep deprivation in rodent studies, one night total sleep deprivation in humans significantly increases expression of at least two miRNAs measured in blood samples (Weigend et al. 2019). Furthermore, in a multi-national European study of children and adolescents, miRNA profiles from blood plasma samples differed significantly for short sleepers compared with normal sleepers (Iacomino et al. 2020). This lends further evidence to the hypothesis that sleep deprivation post-transcriptionally affects gene expression through changes in miRNA regulation of mRNA stability. Interestingly, sleep disorders that increase sleep including narcolepsy induce miRNA dysregulation (Holm et al. 2014)). Thus, in humans and other mammals, miRNA regulation of mRNA transcripts affects sleep, while sleep loss induces region specific changes in miRNA expression, potentially providing a mechanism for sleep deprivation induced cognitive impairments.
To better understand how miRNA regulation may be involved in gene expression changes following sleep deprivation, we analyzed recent acute sleep deprivation studies for the transcriptome and translatome, the subset of mRNA associated with ribosomes, of the hippocampus (Gaine et al. 2021; Lyons et al. 2020). To identify conserved binding motifs for miRNAs, we analyzed mRNA transcripts for genes that were significantly downregulated in the translatome following sleep deprivation, but were not transcriptionally regulated. We used the ENSEMBL 3’UTR sequences obtained with the BioMart utility (www.ensembl.org/biomart/martview) to search for conserved miRNA binding motifs in the 3’UTR of transcripts downregulated by sleep deprivation in the translatome of hippocampal excitatory neurons ((Lyons et al. 2020), GEO series accession GSE156925). Conserved motifs were identified using unbiased motif discovery with the MEME suite with motifs subsequently compared with the miRNA binding site database using Tomtom as detailed (https://github.com/YannVRB/Review-Sleep-Deprivation-Journal-of-Neurochemistry.git). We found two miRNAs, miR-7045–3p and miR-6971–5p, with a high predicted probability for downregulating multiple genes that were downregulated with sleep deprivation in the translatome (Figure 3). There were 5 genes that can be downregulated by both miRNAs suggesting that miRNA regulation is a strong possibility for downregulating these genes. Interestingly, miR-6971 abundance exhibits circadian cycles in the liver with peak expression in the early subjective night (Wang et al. 2016). However, no research has been done on these miRNAs with sleep deprivation or circadian regulation in the brain.
Figure 3. Prediction analysis of miRNAs associated with downregulation of gene expression after sleep deprivation.
Analysis of genes down regulated in the translatome, but not the transcriptome, revealed predicted 3’ UTR binding sites for two miRNAs, miR-7045–3p and miR-6971–5p.
4. Impact of Sleep Deprivation on Protein Synthesis
Although the above processes contribute to altering the pool of mRNA transcripts available for translation after sleep deprivation, recent studies have shown considerable differences between the effects of acute sleep deprivation on the transcriptome compared to the translatome, particularly in the hippocampus (Delorme et al. 2021a; Delorme et al. 2021b; Gaine et al. 2021; Lyons et al. 2020; Puentes-Mestril et al. 2021). The disparity between gene regulation evident at the level of the hippocampal transcriptome and regulation in the translatome after acute sleep deprivation (Gaine et al. 2021; Lyons et al. 2020) suggests that additional translational mechanisms may also be affected by sleep deprivation.
4.1. Sleep Deprivation Alters Protein Abundance
During deep sleep, protein synthesis has been shown to increase in non-human primates in many regions of the brain, although variation occurs between brain regions (Nakanishi et al. 1997). In rodent models, rates of protein synthesis have also been shown to increase across the brain during slow wave sleep, although the same correlation was not observed during REM sleep (Ramm & Smith 1990). These studies suggest that areas particularly vulnerable to sleep deprivation may show altered rates of protein synthesis. Indeed, acute sleep deprivation has been found to decrease protein synthesis as well as altering mRNA stability (Lamon et al. 2021; Tudor et al. 2016). As with gene transcription, there are regional specific effects of sleep deprivation on the proteome. In mice, six hours of acute sleep deprivation decreases polyribosomal mRNA complexes in cortical tissue, presumably decreasing protein synthesis (Naidoo et al. 2005). More direct assessments of in vivo protein synthesis in brain tissue have been done using the incorporation of puromycin into growing polypeptide chains and subsequent quantification through western blotting, a technique known as ribopuromycylation or the SUnSET assay (Schmidt et al. 2009). Five hours of sleep deprivation was found to decrease in vivo protein synthesis by approximately 50% in the hippocampus (Tudor et al. 2016). In eight hour sleep deprivation experiments with subsequent analysis of synaptic proteins in the cerebral cortex of Wistar rats, the protein abundance of 78 proteins was altered after sleep deprivation (Gulyassy et al. 2022). In contrast to what has been shown for the hippocampus, almost 70% of cortical proteins were upregulated by sleep deprivation including upregulation of metabotropic GABA receptors. Interestingly this proteomic data suggested suppression of vesicle exocytosis and impaired endocytosis (Gulyassy et al. 2022). In the mouse forebrain as a whole, six hours of sleep deprivation altered the synaptic proteome and protein abundance (Bruning et al. 2019; Noya et al. 2019).
In humans, the effect of sleep deprivation on protein synthesis has been primarily investigated through studies of skeletal muscle mass, which is primarily dependent on protein synthesis (de Boer et al. 2007). In young adults, male and female, acute sleep deprivation decreases protein synthesis in muscle by 18% as measured in pre- and post- sleep deprivation muscle biopsies using 13C labeled phenylalanine (Lamon et al. 2021). However, in the same study, markers of protein degradation were not affected by acute sleep deprivation (Lamon et al. 2021). This research suggests that sleep deprivation targets protein synthesis, rather than protein degradation. Five days of partial sleep restriction in humans has also been shown to reduce myofibrillar protein synthesis (Saner et al. 2020). Similarly, four days of REM sleep deprivation in rats also caused decreased muscle mass (Dattilo et al. 2012; Monico-Neto et al. 2015). These studies raise the question of whether protein synthesis overall is affected or whether specific subsets of genes are targeted.
4.2. Sleep deprivation targets the initiation of translation
To better understand how sleep deprivation affects protein abundance and protein synthesis, it is necessary to look at the molecular mechanisms that underlie translation. Prior to the association of mRNA transcripts with ribosomes, upstream signaling through the target of rapamycin (mTOR) pathway provides a crucial level of regulation for cap-dependent translation. The mTOR complex phosphorylates two downstream targets, S6 kinase and eukaryotic initiation factor 4e binding protein (4EBP). Phosphorylation induces the activation of the serine/threonine S6 kinase resulting in the phosphorylation of downstream targets including ribosomal protein S6 (RPS6), a component of the 40S small ribosome subunit (reviewed in (Magnuson et al. 2012; Yi et al. 2021). RPS6 phosphorylation is dynamically regulated and can occur through multiple pathways which either activate S6 kinase or induce direct phosphorylation of RPS6 (Hutchinson et al. 2011; Yi et al. 2021). Phosphorylation of RPS6 affects mRNA binding to the ribosome, thus regulating translation. In a second pathway affecting translation, S6 kinase phosphorylates BMAL1 facilitating the interaction of BMAL1 with the translation initiation complex (Lipton et al. 2015; Marti et al. 2017). In the hippocampus, no change has been observed in the phosphorylation of S6 kinase after 5 hours of sleep deprivation (Tudor et al. 2016), suggesting that this is not a major pathway through which sleep deprivation impacts protein synthesis.
The second branch of the mTOR pathway is the phosphorylation of 4EBP2, the most prominent 4EBP in the brain (Banko et al. 2005). 4EBP2 regulates the initiation of cap-dependent translation by regulating the association of the eukaryotic initiation factors needed for the cap-binding protein complex. In its hypophosphorylated form, 4EBP2 binds to the initiation factor eIF4E preventing eIF4G from binding (Figure 2). mTOR phosphorylation of 4EBP causes the dissociation of 4EBP from the translation initiation factor eIF4E, thus allowing the subsequent association of eIF4E and eIF4G (reviewed in (Thoreen et al. 2012). eIF4E and eIF4G bind to eIF4A forming the complex that binds to the 5’cap of the mRNA transcript, allowing the initiation of translation (Ma & Blenis 2009). Acute sleep deprivation decreases activation of the mTORC complex resulting in decreased levels of phosphorylated 4EBP2 in the hippocampus (Tudor et al. 2016). Consequently, a decreased association of eIF4E and eIF4G is also seen in the hippocampus after sleep deprivation (Tudor et al. 2016). Thus, the mTOR-4EBP2 pathway appears to be a major target of sleep deprivation to decrease protein synthesis in the hippocampus. Interestingly, chronic sleep deprivation experiments in rats also found that sleep deprivation increased levels of hypophosphorylated (active) 4EBP and muscle atrophy (de Sa Souza et al. 2016), suggesting that the impact of sleep deprivation on protein synthesis through the mTOR-4EBP pathway affects multiple tissues.
eIF4E phosphorylation also promotes the initiation of translation (reviewed in (Amorim et al. 2018). Acute sleep deprivation in rats decreases levels of eIF4E phosphorylation in the dentate gyrus (Gronli et al. 2012). Similarly, two nights of total sleep deprivation decreases eiF4E expression, supporting the hypothesis that sleep deprivation decreases protein synthesis through suppression of translation initiation (Uyhelji et al. 2018). The initiation of translation is complex involving numerous initiation factors (reviewed in (Komar & Merrick 2020). The initiation of translation may also be regulated through an eIF2α dependent mechanism. eIF2α dependent translation has been shown important for local mRNA translation in dendrites and synaptic plasticity (reviewed in (Oliveira & Klann 2022). Dysregulation and accumulation of eIF2α phosphorylation has been observed in models of Alzheimer’s disease pathology and neurodegeneration (Chang et al. 2002a; Chang et al. 2002b). Six hours of acute sleep deprivation was found to double the level of phosphorylated eIF2α in the mouse cortex (Naidoo et al. 2005). However, in the hippocampus, acute sleep deprivation does not appear to affect eIF2α phosphorylation levels (Tudor et al. 2016). Thus, as with transcription, the mechanism through which sleep deprivation affects the initiation of translation also appears to vary depending upon region of the brain. Although much of the research to date has focused on the initiation of translation, sleep deprivation may also interfere with peptide elongation by impacting translation elongation factors. Phosphorylation of the eukaryotic elongation factor eEF2 inhibits the elongation of translation (reviewed in (Ballard et al. 2021). Acute sleep deprivation has been shown to increase eEF2 phosphorylation in the mouse hippocampus and prefrontal cortex (Gronli et al. 2012). Thus, in addition to the impacts on translation mediated by 4EBP, sleep deprivation also affects the abundance and modification of translation initiation factors.
4.3. Analysis of the translatome
In order to more specifically understand the genes and pathways through which sleep deprivation affects neural function and memory, it is important to analyze gene regulation at the level of the translatome. Development of new techniques for the isolation of mRNA transcripts associated with ribosomes has facilitated this type of analysis. Translating ribosome affinity purification and RNA sequencing (TRAP-Seq) with transgenic mouse lines or viral delivery of transgenes has enabled cell type specific isolation of the translatome (Dougherty 2017; Heiman et al. 2008; Nectow et al. 2017; Sanz et al. 2019; Sanz et al. 2009). Combination of TRAP-Seq approaches with specificity for ribosomes containing phosphorylated ribosomal protein S6 also been used for translatome profiling of activated neurons (Jiang et al. 2015; Knight et al. 2012). Research using TRAP-Seq techniques has been used to investigate gene regulation in hippocampus excitatory neurons following acute sleep deprivation (Delorme et al. 2021a; Lyons et al. 2020). These studies employed different periods and methods for sleep deprivation with Delorme and colleagues using 3 hours of sleep deprivation that included direct disturbance of the animal, while Lyons and colleagues used 5 hours of sleep deprivation that included only cage tapping or shaking (Delorme et al. 2021a; Lyons et al. 2020). Given these different approaches, comparative analysis of the resulting data sets may provide insight into the temporal profile of the changes in the translatome that occur during acute sleep deprivation. We compared the differential gene expression in hippocampal excitatory neurons from studies using 3 hours sleep deprivation (Delorme et al. 2021a) or 5 hours of sleep deprivation (Lyons et al. 2020) finding a common set of 51 differentially expressed genes (Figure 4a). The overlapping data set included transcription factors, members of the AMPK signaling pathway, and genes involved in RNA methylation, which suggests that additional genes may be regulated at later time points. Interestingly, there were a large number of genes that were exclusively regulated in one or the other data set. Whether these differences arose due to temporal regulation of the translatome during sleep deprivation or due to the different methods of sleep deprivation and prior housing remains an open question that warrants further investigation.
Figure 4. Comparative analysis of the translatome in hippocampal excitatory neurons after sleep deprivation and long-term potentiation.
a) Comparison of differentially expressed genes after 3 hours of sleep deprivation and 5 hours of sleep deprivation. b) Comparison of differentially expressed genes in the translatome after 5 hours of sleep deprivation with changes observed after LTP in the hippocampus. Genes highlighted in yellow have opposite directions of regulation between sleep deprivation and LTP.
Comparison of differentially regulated genes in the translatomes after sleep deprivation and synaptic plasticity (long-term potentiation) can potentially provide insight into specific mechanisms underlying the inhibition of memory consolidation by acute sleep deprivation. We compared differentially expressed genes in translatome of the excitatory neurons of the hippocampus after acute sleep deprivation (Lyons et al. 2020) with previously identified differentially expressed genes in the excitatory neurons of the CA1 and dentate gyrus of the hippocampus after the induction of long-term potentiation (Chen et al. 2017) in Figure 4b. As may be expected given the different effects of sleep deprivation and LTP on neural plasticity, the bulk of gene regulation in the translatome was exclusive to one data set or the other, although there were a few genes commonly regulated in both data sets that exhibited different directions of regulation. These results suggest that the changes in gene regulation in excitatory neurons induced by sleep deprivation constitute more than just opposite regulation of genes involved in synaptic plasticity. Five hours of acute sleep deprivation has also been shown to activate somatostatin expressing inhibitory neurons in the hippocampus, with the activation of these neurons appearing to be a mechanism through which sleep deprivation inhibits long-term memory (Delorme et al. 2021b). Thus, sleep deprivation affects gene regulation in both excitatory and inhibitory neurons of the hippocampus to inhibit synaptic plasticity and long-term memory consolidation.
5. Future Directions
The past twenty years has seen an explosion of research examining the impacts of sleep deprivation on cognitive performance, health and lifespan. Despite the evidence of adverse health impacts, acute and chronic sleep deprivation continue to be a significant public health problem in the United States and worldwide affecting children, teenagers and adults. The complexity of regional and cell-type specific effects in the brain following sleep deprivation make it clear that more research needs to be done to fully understand the molecular and cellular impacts of sleep deprivation on the brain. Although this is not an all-inclusive list, we identified several areas of research warranting further research for which increased understanding has the potential to enable advances for human health.
5.1. Molecular Changes during Recovery Sleep
The homeostatic need for sleep results in compensatory rebound sleep following sleep deprivation. The time needed for recovery following acute or prolonged sleep restriction can vary depending upon the physiological process or genes examined. Rebound sleep combined with a full night’s sleep following acute sleep deprivation is often insufficient to mitigate the adverse impacts of sleep deprivation as seen from studies across phyla. For example, in the marine mollusk Aplysia, the effects of acute sleep deprivation on short and long-term learning and memory persist for more than 24 hours (Krishnan et al. 2016a). In Drosophila, researchers using a model of Parkinson’s disease found that one night of acute sleep deprivation induced persistent deficits in short-term memory that lasted at least three days (Seugnet et al. 2009). In humans, some studies have reported that one long night of recovery sleep is sufficient to return performance scores to baseline following one night of acute sleep deprivation (Lamond et al. 2007). However, individuals with shorter periods of recovery sleep (6 or 8 hours) following acute sleep deprivation still exhibit performance deficits in neurobehavioral tasks (Lamond et al. 2007; Sallinen et al. 2004). For more challenging neurobehavioral tasks, two to three nights of recovery sleep were needed to restore performance following sleep deprivation (Ikegami et al. 2009). Recent research analyzing hippocampal connectivity and episodic memory found that two nights of recovery sleep following total sleep deprivation were sufficient to restore hippocampal connections, but not normal episodic memory (Chai et al. 2020). A few studies have investigated cellular changes during recovery following chronic sleep deprivation in rodent models and in humans. In mice, changes in neuroinflammation, microglial activation and neuronal degeneration in the hippocampus have been observed three weeks after seven days of REM sleep deprivation (Yin et al. 2017). In rats, three weeks of recovery sleep is insufficient to completely restore changes in the dendritic arborization of hippocampal CA3 that occur with three weeks of chronic sleep deprivation (Konakanchi et al. 2022). Thus, recovery from either acute sleep deprivation or chronic sleep restriction appears to be a lengthier process than previously envisioned.
The human brain displays some ability to adapt to mild chronic sleep loss to stabilize performance and attention at a reduced level; however, adaptation has not been observed during a seven day paradigm of severe sleep restriction with participants showing continuing declines in performance (Belenky et al. 2003). With mild sleep restriction (5 to 7 hours in bed), three days of recovery sleep was insufficient to return performance in psychomotor vigilance tasks to baseline levels restriction (Belenky et al. 2003). More recent research has shown that 7 days of recovery sleep following a 30% sleep reduction paradigm for ten days was insufficient to restore baseline performance on the Stroop test (Ochab et al. 2021). In individuals subjected to one week of sleep restriction followed by two night of recovery sleep, performance deficits in psychomotor vigilance tests persisted (Pejovic et al. 2013). Thus, for the millions of teenagers and adults in which chronic sleep loss occurs during the week, increased sleep on the weekends appears to be insufficient to restore cognitive performance to normal levels. Furthermore, researchers found that weekend recovery sleep was insufficient to account for the metabolic disturbances associated with sleep deprivation including changes in insulin sensitivity in healthy young adults (Depner et al. 2019). Interestingly, two days of recovery sleep following one week of mild sleep restriction does appear to normalize cortisol levels and cytokines (Pejovic et al. 2013). As a caveat, a recent study of one night total sleep deprivation followed by two nights of recovery sleep determined that although cortisol levels changed with sleep deprivation, they were not a marker of resilience or vulnerability to sleep deprivation based on multiple neurobehavioral tests (Yamazaki et al. 2021).
Molecular studies of rebound sleep have focused primarily on the identification of transcripts and proteins that changed following acute sleep deprivation and then returned to baseline within 2 – 3 hours of recovery sleep. For example, following 6 h sleep deprivation in mice, the phosphorylated glial protein NDRG2, a marker of sleep need, returns to baseline phosphorylation levels within 2 hours of recovery sleep (Suzuki et al. 2013). In transcriptome studies of the hippocampus following acute sleep deprivation, most genes, including immediate early genes and transcription factors examined return to baseline expression levels with three hours of recovery sleep with some notable exceptions (Gaine et al. 2021; Vecsey et al. 2012). In contrast, the splicing factor Srsf7 was found to reverse directions during recovery sleep supporting the hypothesis that sleep deprivation changes mRNA splicing patterns and recovery sleep may reduce alternate splicing (Gaine et al. 2021). However, studies for longer periods of recovery sleep have not been performed in the hippocampus. In the cortex, 2.5 h of recovery sleep also normalizes many of sleep deprivation associated changes in gene expression; however, some genes required up to 6 hours to return to basal levels (Gerstner et al. 2016). At the proteomic level, 78 synaptic proteins in the cerebral cortex were changed after 8 hours of sleep deprivation with 56 upregulated, while 39 synaptic proteins were altered following 16 hours of recovery sleep with 15 upregulated (Gulyassy et al. 2022). However, no intermediate time points in recovery sleep were examined. With the limited number of studies done examining the time course of gene regulation and protein synthesis during recovery from sleep deprivation, higher temporal resolution molecular studies of multiple brain regions need to be done to fully understand the recovery patterns in gene regulation after sleep deprivation.
5.2. Long-term Impact of Chronic Sleep Disturbances
In a recent study commissioned by the American Academy of Sleep Medicine, sleep disturbances in U.S. adults have increased during the Covid-19 pandemic with 56% of individuals surveyed reporting chronic sleep disturbances (AASM Sleep Prioritization Study March 2021). Future studies on the impacts of sleep loss need to include additional paradigm complexity including repeated bouts of acute sleep deprivation, the persistence of sleep fragmentation arising from sleep deprivation, chronic sleep loss, and the combinatorial effects of sleep deprivation and circadian dysfunction on memory and performance. Importantly, researchers should also consider the long-term effects of chronic sleep disturbances across age groups.
Models of chronic sleep restriction in rodents have shown that sleep restriction in adolescent and young adult animals results in long-lasting impairments in learning and memory. Chronic sleep restriction in pregnant dams results in impairments in hippocampus-dependent memory in the offspring including reductions in spatial memory in the Morris water maze, long-term potentiation and excitatory synaptic activity (Peng et al. 2016). Ten days of chronic sleep loss in adolescent rats leads to hippocampus-dependent memory impairments that are still evident after four weeks of recovery, although hippocampus independent memory was not similarly impacted (Howard & Hunter 2019). Chronic sleep restriction in adolescent mice also results in changes in synaptic structure impairing synaptic reorganization (Nagai et al. 2021). Recent research found that 12 weeks of chronic sleep restriction in young adult wild type mice induced persistent adverse impacts including a decrease in CA1 neurons and a reduction in hippocampal volume as well as increased tau phosphorylation twelve months later (Owen et al. 2021). Given the potential life-long consequences of chronic sleep loss in adolescents and young adults and the increasing problem of sleep deprivation in children and teenagers, more research needs to done at the molecular and cellular level to understand chronic sleep loss.
5.3. Sleep Disturbances with Aging
To date, most of the studies on sleep deprivation have been done using young animals or in young adults. However, changes in sleep patterns with aging or stress may affect memory and cognitive function. More research needs to be done on the molecular impacts of sleep disturbances with aging, particularly given the links between sleep disturbances and the increased progression of age-related diseases, including neurodegenerative diseases such as Alzheimer’s disease. Potentially, behavioral therapies or lifestyle changes may assist in ameliorating the cognitive impacts of sleep changes that occur with age. For example, exercise has been shown to improve memory and psychomotor performance in middle aged and elderly individuals, even increasing hippocampal volume (De la Rosa et al. 2019; Erickson et al. 2011). In rodent models, the intensity and duration of exercise appears to correlate with increased BDNF activation in the hippocampus and memory performance, although higher intensity exercise is needed for cortical activation (Aguiar et al. 2008; Cefis et al. 2019).
Disrupted sleep patterns and circadian rhythm disorders have been associated with an increased risk and incidence of Alzheimer’s disease and neurodegeneration in animal models and in humans. In humans, longitudinal studies of sleep in individuals starting in middle-age have shown that decreased sleep increases the risk of dementia by 30%, particularly Alzheimer’s disease (Sabia et al. 2021; Shi et al. 2018). At the molecular level, 12 weeks of chronic sleep restriction in a mouse tauopathy model accelerated the progression of pathologies including motor impairments and decreased neural volume in multiple brain regions (Zhu et al. 2018). Chronic sleep fragmentation has also been found to increase the deposition and accumulation of amyloid β with the severity of sleep fragmentation correlated to amyloid β deposition (Minakawa et al. 2017). Similarly, studies in humans have shown increases in Aβ plaque formation in individuals with poor sleep (Spira et al. 2014; Spira et al. 2013). Moreover, increased Aβ aggregation also increases sleep disturbances exacerbating disease progression (reviewed in (Chen et al. 2021; Sun et al. 2022). Sleep deprivation may, in part, increase the risk of neurological diseases through decreased clearing of metabolic waste via the glymphatic system (reviewed in (Achariyar et al. 2016; Bishir et al. 2020; Rasmussen et al. 2018). There have been many recent reviews discussing the interactions between sleep and circadian disorders and neurodegenerative diseases, particularly Alzheimer’s disease. For more information, (Vanderheyden et al. 2018; Wang & Holtzman 2020; Wu et al. 2019).
Although Alzheimer’s disease is the most studied neurodegenerative disease in regard to sleep issues, sleep disorders also are associated with other dementias and neurodegenerative diseases. Multiple studies have shown that individuals with Lewy body dementia experience even a greater percentage of sleep disruptions than Alzheimer’s patients, up to twice as much (Cagnin et al. 2017; Koren et al. 2023). Systematic reviews have found that approximately 90% of Lewy body dementia patients have at least one sleep disorder (Elder et al. 2022). Focusing on specific aspects of sleep disorders in dementia has allowed researchers to identify specific sleep disturbances associated with these conditions. For example, researchers have found that non-REM sleep with hypertonia may serve as a diagnostic biomarker for Lewy body dementia and Parkinson’s disease with dementia (Levendowski et al. 2022). Sleep deprivation and sleep disturbances have also been associated with Parkinson’s disease, with more than 60% of patients experiencing sleep issues (Louter et al. 2012; Maggi et al. 2023). In mice carrying a mutation in leucine-rich repeat kinase (Lrrk2), an autosomal dominant mutation associated with Parkinson’s disease, chronic sleep restriction increased the progression and severity of Parkinson pathologies (Liu et al. 2022b). Although the impact of specific gene changes that occur with sleep deprivation and disturbances in neurodegenerative diseases remains unknown, it is clear that more research needs done to understand the molecular mechanisms through which sleep disturbances increase the incidence or progression of neurodegenerative diseases. Given the increasing aging population in many countries including the U.S., more molecular and cellular research needs to be done to understand the effect of sleep disruption on age-related diseases and to identify potential mechanisms to alleviate these conditions.
5.4. Functions of Sleep and Systems Interactions
In addition to the connections between sleep and neuronal function, the functions of sleep are widespread interacting with multiple physiological systems including development, metabolism, the cardiovascular system, the immune system and muscle growth (reviewed in (Mullington et al. 2009; Tobaldini et al. 2017; Zielinski et al. 2016). Sleep deprivation damages multiple organs through oxidative stress and inflammation, including damage to the thyroid gland, liver, heart, and kidneys (Li et al. 2021b; Periasamy et al. 2015). In humans, sleep deprivation also affects immune system responses increasing inflammation (reviewed in (Mullington et al. 2009; Simpson & Dinges 2007). Systemic inflammatory markers are also increased following sleep deprivation in animal models as well as the activation of microglia (Liu et al. 2022a; Periasamy et al. 2015; Poroyko et al. 2016; Zhang et al. 2021). Thus, it is possible that indirect effects of sleep deprivation on other systems may affect neural function. Undoubtedly, future research will focus on system wide interactions of sleep.
5.5. Conclusion
Although sleep is generally thought of as a rhythmic daily behavior, it is clear that both acute and chronic sleep deprivation can induce changes in molecular signaling and gene expression that extend past the duration of the sleep loss. It is critical to understand the detailed molecular impacts of sleep deprivation as changes in gene expression underlie long-term memory and contribute to the increased incidence and exacerbation of neurodegenerative diseases. As the individual and societal factors contributing to sleep deprivation and sleep disturbances appear unlikely to abate, more research needs to be done to identify pathways and gene targets through which the impacts of sleep deprivation may be mitigated. The benefits of research on gene regulation and protein synthesis associated with sleep disturbances will also provide vital insight into gene dysregulation and identify ways in which to these molecular changes might be reversed. Increasing resilience to the adverse effects of sleep loss will have significant impacts on the human, societal and economic costs of sleep deprivation and sleep disturbances.
Acknowledgements
This work was supported by a multi-PI grant from the National Institutes of Health, Institute of Aging R01 AG062398 (TA and LCL). The funding agency had no role in the design of the study, data collection, analysis or interpretation, and no role in the writing of the manuscript. TA is also supported by the Roy J. Carver Chair in Neuroscience.
List of Abbreviations:
- 4EBP
Eukaryotic initiation factor 4e binding protein
- BDNF
Brain derived neurotrophic factor
- cAMP
Cyclic AMP, adenosine 3’,5’-cyclic monophosphate
- Cpebp4
Cytoplasmic polyadenylation element binding protein
- Cpsf2
Cleavage and polyadenylation factor 2
- CREB
cAMP response binding element
- CRY
Cryptochrome
- eIF
Eukaryotic initiation factor
- Elk1
ETS like Transcription Factor 1
- ERK
Extracellular signal-regulated kinase
- fMRI
Functional magnetic resonance imaging
- Hrt1a
5-hydroxytryptamine receptor 1a
- Kcnv1
Potassium voltage-gated channel subfamily V member 1
- LTP
Long-term potentiation
- miRNA
Micro RNA
- mRNA
Messenger RNA
- mTOR
Mammalian Target of Rapamycin
- NREM
Non rapid eye movement sleep
- pCREB
phosphorylated cyclic AMP response binding element
- PER1
Period circadian protein homolog 1
- PKA
Protein kinase A
- REM
Rapid eye movement sleep
- RNA
Ribonucleic acid
- RPS6
Ribosomal protein S6
- Srsf7
Serine and arginine rich splicing factor 7
- TRAP-Seq
Translating ribosome affinity purification and RNA sequencing
- UTR
Untranslated Region
Footnotes
Conflict of Interest
Ted Abel serves as a scientific advisor for Radius Health and Aditum Bio, and is a member of the Scientific Advisory Board of Embark Neuro. The authors declare that they have no other competing interests.
Data Availability Statement
Data analyzed in the current study is available at GEO series accession GSE156925 with the analysis pipeline at https://github.com/YannVRB/Review-Sleep-Deprivation-Journal-of-Neurochemistry.git.
References
- Achariyar TM, Li B, Peng W et al. (2016) Glymphatic distribution of CSF-derived apoE into brain is isoform specific and suppressed during sleep deprivation. Mol Neurodegener 11, 74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Acosta-Pena E, Camacho-Abrego I, Melgarejo-Gutierrez M, Flores G, Drucker-Colin R and Garcia-Garcia F. (2015) Sleep deprivation induces differential morphological changes in the hippocampus and prefrontal cortex in young and old rats. Synapse 69, 15–25. [DOI] [PubMed] [Google Scholar]
- Adams R, Appleton S, Taylor A, McEvoy D, Antic N. (2016) 2016 Sleep Health Survey of Australian Adults. Sleep Health Foundation. [Google Scholar]
- Adler A, Lai CSW, Yang G, Geron E, Bai Y. and Gan WB (2021) Sleep promotes the formation of dendritic filopodia and spines near learning-inactive existing spines. Proc Natl Acad Sci U S A 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aguiar AS Jr., Speck AE, Prediger RD, Kapczinski F. and Pinho RA (2008) Downhill training upregulates mice hippocampal and striatal brain-derived neurotrophic factor levels. J Neural Transm (Vienna) 115, 1251–1255. [DOI] [PubMed] [Google Scholar]
- AIHW (2021) Sleep problems as a risk factor for chronic conditions. Australian Institute of Health and Welfare. DOI: 10.25816/d2d7-p797 [DOI] [Google Scholar]
- Aksoy-Aksel A, Zampa F. and Schratt G. (2014) MicroRNAs and synaptic plasticity--a mutual relationship. Philos Trans R Soc Lond B Biol Sci 369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alkadhi KA and Alhaider IA (2016) Caffeine and REM sleep deprivation: Effect on basal levels of signaling molecules in area CA1. Mol Cell Neurosci 71, 125–131. [DOI] [PubMed] [Google Scholar]
- Alles J, Fehlmann T, Fischer U. et al. (2019) An estimate of the total number of true human miRNAs. Nucleic Acids Res 47, 3353–3364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alvarez-Saavedra M, Antoun G, Yanagiya A, Oliva-Hernandez R, Cornejo-Palma D, Perez-Iratxeta C, Sonenberg N. and Cheng HY (2011) miRNA-132 orchestrates chromatin remodeling and translational control of the circadian clock. Hum Mol Genet 20, 731–751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amorim IS, Lach G. and Gkogkas CG (2018) The Role of the Eukaryotic Translation Initiation Factor 4E (eIF4E) in Neuropsychiatric Disorders. Front Genet 9, 561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Appelbaum L, Wang G, Yokogawa T, Skariah GM, Smith SJ, Mourrain P. and Mignot E. (2010) Circadian and homeostatic regulation of structural synaptic plasticity in hypocretin neurons. Neuron 68, 87–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aten S, Hansen KF, Price KH, Wheaton K, Kalidindi A, Garcia A, Alzate-Correa D, Hoyt KR and Obrietan K. (2018) miR-132 couples the circadian clock to daily rhythms of neuronal plasticity and cognition. Learn Mem 25, 214–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ballard DJ, Peng HY, Das JK et al. (2021) Insights Into the Pathologic Roles and Regulation of Eukaryotic Elongation Factor-2 Kinase. Front Mol Biosci 8, 727863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banko JL, Poulin F, Hou L, DeMaria CT, Sonenberg N. and Klann E. (2005) The translation repressor 4E-BP2 is critical for eIF4F complex formation, synaptic plasticity, and memory in the hippocampus. J Neurosci 25, 9581–9590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Belenky G, Wesensten NJ, Thorne DR, Thomas ML, Sing HC, Redmond DP, Russo MB and Balkin TJ (2003) Patterns of performance degradation and restoration during sleep restriction and subsequent recovery: a sleep dose-response study. J Sleep Res 12, 1–12. [DOI] [PubMed] [Google Scholar]
- Beyaert L, Greggers U. and Menzel R. (2012) Honeybees consolidate navigation memory during sleep. J Exp Biol 215, 3981–3988. [DOI] [PubMed] [Google Scholar]
- Bishir M, Bhat A, Essa MM et al. (2020) Sleep Deprivation and Neurological Disorders. Biomed Res Int 2020, 5764017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bolsius YG, Meerlo P, Kas MJ, Abel T. and Havekes R. (2022) Sleep deprivation reduces the density of individual spine subtypes in a branch-specific fashion in CA1 neurons. J Sleep Res 31, e13438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brodin ATS, Gabulya S, Wellfelt K. and Karlsson TE (2022) Five Hours Total Sleep Deprivation Does Not Affect CA1 Dendritic Length or Spine Density. Front Synaptic Neurosci 14, 854160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bruning F, Noya SB, Bange T. et al. (2019) Sleep-wake cycles drive daily dynamics of synaptic phosphorylation. Science 366. [DOI] [PubMed] [Google Scholar]
- Bushey D, Tononi G. and Cirelli C. (2011) Sleep and synaptic homeostasis: structural evidence in Drosophila. Science 332, 1576–1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bushey D, Tononi G. and Cirelli C. (2015) Sleep- and wake-dependent changes in neuronal activity and reactivity demonstrated in fly neurons using in vivo calcium imaging. Proc Natl Acad Sci U S A 112, 4785–4790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cagnin A, Fragiacomo F, Camporese G, Turco M, Busse C, Ermani M. and Montagnese S. (2017) Sleep-Wake Profile in Dementia with Lewy Bodies, Alzheimer’s Disease, and Normal Aging. J Alzheimers Dis 55, 1529–1536. [DOI] [PubMed] [Google Scholar]
- Camarena V, Sant DW, Huff TC et al. (2017) cAMP signaling regulates DNA hydroxymethylation by augmenting the intracellular labile ferrous iron pool. Elife 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cedernaes J, Osler ME, Voisin S, Broman JE, Vogel H, Dickson SL, Zierath JR, Schioth HB and Benedict C. (2015) Acute Sleep Loss Induces Tissue-Specific Epigenetic and Transcriptional Alterations to Circadian Clock Genes in Men. J Clin Endocrinol Metab 100, E1255–1261. [DOI] [PubMed] [Google Scholar]
- Cefis M, Prigent-Tessier A, Quirie A, Pernet N, Marie C. and Garnier P. (2019) The effect of exercise on memory and BDNF signaling is dependent on intensity. Brain Struct Funct 224, 1975–1985. [DOI] [PubMed] [Google Scholar]
- Centers for Disease, C. and Prevention (2009) Perceived insufficient rest or sleep among adults - United States, 2008. MMWR Morb Mortal Wkly Rep 58, 1175–1179. [PubMed] [Google Scholar]
- Chai Y, Fang Z, Yang FN et al. (2020) Two nights of recovery sleep restores hippocampal connectivity but not episodic memory after total sleep deprivation. Sci Rep 10, 8774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang RC, Suen KC, Ma CH, Elyaman W, Ng HK and Hugon J. (2002a) Involvement of double-stranded RNA-dependent protein kinase and phosphorylation of eukaryotic initiation factor-2alpha in neuronal degeneration. J Neurochem 83, 1215–1225. [DOI] [PubMed] [Google Scholar]
- Chang RC, Wong AK, Ng HK and Hugon J. (2002b) Phosphorylation of eukaryotic initiation factor-2alpha (eIF2alpha) is associated with neuronal degeneration in Alzheimer’s disease. Neuroreport 13, 2429–2432. [DOI] [PubMed] [Google Scholar]
- Charlesworth A, Meijer HA and de Moor CH (2013) Specificity factors in cytoplasmic polyadenylation. Wiley Interdiscip Rev RNA 4, 437–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen K, Lv Y, Long X, Liu W. and Zhou J. (2021) The Association between Insomnia-Like Sleep Pattern Changes and Cognitive Dysfunction: Possible Mechanism and Therapeutic Strategy. Curr Alzheimer Res 18, 858–875. [DOI] [PubMed] [Google Scholar]
- Chen PB, Kawaguchi R, Blum C, Achiro JM, Coppola G, O’Dell TJ and Martin KC (2017) Mapping Gene Expression in Excitatory Neurons during Hippocampal Late-Phase Long-Term Potentiation. Front Mol Neurosci 10, 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng HY, Papp JW, Varlamova O. et al. (2007) microRNA modulation of circadian-clock period and entrainment. Neuron 54, 813–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cirelli C, Gutierrez CM and Tononi G. (2004) Extensive and divergent effects of sleep and wakefulness on brain gene expression. Neuron 41, 35–43. [DOI] [PubMed] [Google Scholar]
- Cirelli C. and Tononi G. (1998) Differences in gene expression between sleep and waking as revealed by mRNA differential display. Brain Res Mol Brain Res 56, 293–305. [DOI] [PubMed] [Google Scholar]
- Cirelli C. and Tononi G. (2000) Gene expression in the brain across the sleep-waking cycle. Brain Res 885, 303–321. [DOI] [PubMed] [Google Scholar]
- Daguenet E, Dujardin G. and Valcarcel J. (2015) The pathogenicity of splicing defects: mechanistic insights into pre-mRNA processing inform novel therapeutic approaches. EMBO Rep 16, 1640–1655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dattilo M, Antunes HK, Medeiros A, Monico-Neto M, Souza Hde S, Lee KS, Tufik S. and de Mello MT (2012) Paradoxical sleep deprivation induces muscle atrophy. Muscle Nerve 45, 431–433. [DOI] [PubMed] [Google Scholar]
- Davis CJ, Bohnet SG, Meyerson JM and Krueger JM (2007) Sleep loss changes microRNA levels in the brain: a possible mechanism for state-dependent translational regulation. Neurosci Lett 422, 68–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis CJ, Clinton JM and Krueger JM (2012) MicroRNA 138, let-7b, and 125a inhibitors differentially alter sleep and EEG delta-wave activity in rats. J Appl Physiol (1985) 113, 1756–1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis CJ, Clinton JM, Taishi P, Bohnet SG, Honn KA and Krueger JM (2011) MicroRNA 132 alters sleep and varies with time in brain. J Appl Physiol (1985) 111, 665–672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis CJ, Harding JW and Wright JW (2003) REM sleep deprivation-induced deficits in the latency-to-peak induction and maintenance of long-term potentiation within the CA1 region of the hippocampus. Brain Res 973, 293–297. [DOI] [PubMed] [Google Scholar]
- de Boer MD, Selby A, Atherton P. et al. (2007) The temporal responses of protein synthesis, gene expression and cell signalling in human quadriceps muscle and patellar tendon to disuse. J Physiol 585, 241–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De la Rosa A, Solana E, Corpas R, Bartres-Faz D, Pallas M, Vina J, Sanfeliu C. and Gomez-Cabrera MC (2019) Long-term exercise training improves memory in middle-aged men and modulates peripheral levels of BDNF and Cathepsin B. Sci Rep 9, 3337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Sa Souza H, Antunes HKM, Dattilo M, Lee KS, Monico-Neto M, de Campos Giampa SQ, Phillips SM, Tufik S. and de Mello MT (2016) Leucine supplementation is anti-atrophic during paradoxical sleep deprivation in rats. Amino Acids 48, 949–957. [DOI] [PubMed] [Google Scholar]
- de Vivo L, Bellesi M, Marshall W, Bushong EA, Ellisman MH, Tononi G. and Cirelli C. (2017) Ultrastructural evidence for synaptic scaling across the wake/sleep cycle. Science 355, 507–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Delorme J, Wang L, Kodoth V, Wang Y, Ma J, Jiang S. and Aton SJ (2021a) Hippocampal neurons’ cytosolic and membrane-bound ribosomal transcript profiles are differentially regulated by learning and subsequent sleep. Proc Natl Acad Sci U S A 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Delorme J, Wang L, Kuhn FR et al. (2021b) Sleep loss drives acetylcholine- and somatostatin interneuron-mediated gating of hippocampal activity to inhibit memory consolidation. Proc Natl Acad Sci U S A 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Depner CM, Melanson EL, Eckel RH et al. (2019) Ad libitum Weekend Recovery Sleep Fails to Prevent Metabolic Dysregulation during a Repeating Pattern of Insufficient Sleep and Weekend Recovery Sleep. Curr Biol 29, 957–967 e954. [DOI] [PubMed] [Google Scholar]
- Dharmalingam P, Mahalingam R, Yalamanchili HK, Weng T, Karmouty-Quintana H, Guha A. and R AT (2022) Emerging roles of alternative cleavage and polyadenylation (APA) in human disease. J Cell Physiol 237, 149–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dougherty JD (2017) The Expanding Toolkit of Translating Ribosome Affinity Purification. J Neurosci 37, 12079–12087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duan R, Liu X, Wang T, Wu L, Gao X. and Zhang Z. (2016) Histone Acetylation Regulation in Sleep Deprivation-Induced Spatial Memory Impairment. Neurochem Res 41, 2223–2232. [DOI] [PubMed] [Google Scholar]
- Edelmann E, Lessmann V. and Brigadski T. (2014) Pre- and postsynaptic twists in BDNF secretion and action in synaptic plasticity. Neuropharmacology 76 Pt C, 610–627. [DOI] [PubMed] [Google Scholar]
- Elbaz I, Zada D, Tovin A, Braun T, Lerer-Goldshtein T, Wang G, Mourrain P. and Appelbaum L. (2017) Sleep-Dependent Structural Synaptic Plasticity of Inhibitory Synapses in the Dendrites of Hypocretin/Orexin Neurons. Mol Neurobiol 54, 6581–6597. [DOI] [PubMed] [Google Scholar]
- Elder GJ, Lazar AS, Alfonso-Miller P. and Taylor JP (2022) Sleep disturbances in Lewy body dementia: A systematic review. Int J Geriatr Psychiatry 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erickson KI, Voss MW, Prakash RS et al. (2011) Exercise training increases size of hippocampus and improves memory. Proc Natl Acad Sci U S A 108, 3017–3022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fernandez MP, Berni J. and Ceriani MF (2008) Circadian remodeling of neuronal circuits involved in rhythmic behavior. PLoS Biol 6, e69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fiorenza A. and Barco A. (2016) Role of Dicer and the miRNA system in neuronal plasticity and brain function. Neurobiol Learn Mem 135, 3–12. [DOI] [PubMed] [Google Scholar]
- Fontes MM, Guvenek A, Kawaguchi R. et al. (2017) Activity-Dependent Regulation of Alternative Cleavage and Polyadenylation During Hippocampal Long-Term Potentiation. Sci Rep 7, 17377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Friedman RC, Farh KK, Burge CB and Bartel DP (2009) Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 19, 92–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaine ME, Bahl E, Chatterjee S, Michaelson JJ, Abel T. and Lyons LC (2021) Altered hippocampal transcriptome dynamics following sleep deprivation. Mol Brain 14, 125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaine ME, Chatterjee S. and Abel T. (2018) Sleep Deprivation and the Epigenome. Front Neural Circuits 12, 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao L, Bai L, Zhang Y. et al. (2015) Frequency-dependent changes of local resting oscillations in sleep-deprived brain. PLoS One 10, e0120323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gendreau KL, Unruh BA, Zhou C. and Kojima S. (2018) Identification and Characterization of Transcripts Regulated by Circadian Alternative Polyadenylation in Mouse Liver. G3 (Bethesda) 8, 3539–3548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gerstner JR, Koberstein JN, Watson AJ, Zapero N, Risso D, Speed TP, Frank MG and Peixoto L. (2016) Removal of unwanted variation reveals novel patterns of gene expression linked to sleep homeostasis in murine cortex. BMC Genomics 17, 727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gerstner JR, Vanderheyden WM, LaVaute T, Westmark CJ, Rouhana L, Pack AI, Wickens M. and Landry CF (2012) Time of day regulates subcellular trafficking, tripartite synaptic localization, and polyadenylation of the astrocytic Fabp7 mRNA. J Neurosci 32, 1383–1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gisabella B, Scammell T, Bandaru SS and Saper CB (2020) Regulation of hippocampal dendritic spines following sleep deprivation. J Comp Neurol 528, 380–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Graves LA, Heller EA, Pack AI and Abel T. (2003) Sleep deprivation selectively impairs memory consolidation for contextual fear conditioning. Learn Mem 10, 168–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gronli J, Dagestad G, Milde AM, Murison R. and Bramham CR (2012) Post-transcriptional effects and interactions between chronic mild stress and acute sleep deprivation: regulation of translation factor and cytoplasmic polyadenylation element-binding protein phosphorylation. Behav Brain Res 235, 251–262. [DOI] [PubMed] [Google Scholar]
- Guan Z, Peng X. and Fang J. (2004) Sleep deprivation impairs spatial memory and decreases extracellular signal-regulated kinase phosphorylation in the hippocampus. Brain Res 1018, 38–47. [DOI] [PubMed] [Google Scholar]
- Gulyassy P, Todorov-Volgyi K, Toth V, Gyorffy BA, Puska G, Simor A, Juhasz G, Drahos L. and Kekesi KA (2022) The Effect of Sleep Deprivation and Subsequent Recovery Period on the Synaptic Proteome of Rat Cerebral Cortex. Mol Neurobiol 59, 1301–1319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guzman-Marin R, Ying Z, Suntsova N, Methippara M, Bashir T, Szymusiak R, Gomez-Pinilla F. and McGinty D. (2006) Suppression of hippocampal plasticity-related gene expression by sleep deprivation in rats. J Physiol 575, 807–819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hafner M, Stepanek M, Taylor J, Troxel WM and van Stolk C. (2017) Why Sleep Matters-The Economic Costs of Insufficient Sleep: A Cross-Country Comparative Analysis. Rand Health Q 6, 11. [PMC free article] [PubMed] [Google Scholar]
- Hagewoud R, Havekes R, Novati A, Keijser JN, Van der Zee EA and Meerlo P. (2010) Sleep deprivation impairs spatial working memory and reduces hippocampal AMPA receptor phosphorylation. J Sleep Res 19, 280–288. [DOI] [PubMed] [Google Scholar]
- Havekes R, Bruinenberg VM, Tudor JC, Ferri SL, Baumann A, Meerlo P. and Abel T. (2014) Transiently increasing cAMP levels selectively in hippocampal excitatory neurons during sleep deprivation prevents memory deficits caused by sleep loss. J Neurosci 34, 15715–15721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Havekes R, Meerlo P. and Abel T. (2015) Animal studies on the role of sleep in memory: from behavioral performance to molecular mechanisms. Curr Top Behav Neurosci 25, 183–206. [DOI] [PubMed] [Google Scholar]
- Havekes R, Park AJ, Tudor JC et al. (2016) Sleep deprivation causes memory deficits by negatively impacting neuronal connectivity in hippocampal area CA1. Elife 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hawk JD, Florian C. and Abel T. (2011) Post-training intrahippocampal inhibition of class I histone deacetylases enhances long-term object-location memory. Learn Mem 18, 367–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heiman M, Schaefer A, Gong S. et al. (2008) A translational profiling approach for the molecular characterization of CNS cell types. Cell 135, 738–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hellman K, Hernandez P, Park A. and Abel T. (2010) Genetic evidence for a role for protein kinase A in the maintenance of sleep and thalamocortical oscillations. Sleep 33, 19–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hendricks JC, Williams JA, Panckeri K, Kirk D, Tello M, Yin JC and Sehgal A. (2001) A non-circadian role for cAMP signaling and CREB activity in Drosophila rest homeostasis. Nat Neurosci 4, 1108–1115. [DOI] [PubMed] [Google Scholar]
- Hillman D, Mitchell S, Streatfeild J, Burns C, Bruck D. and Pezzullo L. (2018) The economic cost of inadequate sleep. Sleep 41. [DOI] [PubMed] [Google Scholar]
- Holm A, Bang-Berthelsen CH, Knudsen S, Kornum BR, Modvig S, Jennum P. and Gammeltoft S. (2014) miRNA profiles in plasma from patients with sleep disorders reveal dysregulation of miRNAs in narcolepsy and other central hypersomnias. Sleep 37, 1525–1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hor CN, Yeung J, Jan M, Emmenegger Y, Hubbard J, Xenarios I, Naef F. and Franken P. (2019) Sleep-wake-driven and circadian contributions to daily rhythms in gene expression and chromatin accessibility in the murine cortex. Proc Natl Acad Sci U S A 116, 25773–25783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howard KA and Hunter AS (2019) Immediate and long-lasting cognitive consequences of adolescent chronic sleep restriction. Behav Neurosci 133, 461–466. [DOI] [PubMed] [Google Scholar]
- Huang B, Sun B, Yang R. et al. (2022) Long-lasting effects of postweaning sleep deprivation on cognitive function and social behaviors in adult mice. Neuropharmacology 215, 109164. [DOI] [PubMed] [Google Scholar]
- Huang HY, Lin YC, Li J. et al. (2020) miRTarBase 2020: updates to the experimentally validated microRNA-target interaction database. Nucleic Acids Res 48, D148–D154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hussaini SA, Bogusch L, Landgraf T. and Menzel R. (2009) Sleep deprivation affects extinction but not acquisition memory in honeybees. Learn Mem 16, 698–705. [DOI] [PubMed] [Google Scholar]
- Hutchinson JA, Shanware NP, Chang H. and Tibbetts RS (2011) Regulation of ribosomal protein S6 phosphorylation by casein kinase 1 and protein phosphatase 1. J Biol Chem 286, 8688–8696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iacomino G, Lauria F, Russo P. et al. (2020) Circulating miRNAs are associated with sleep duration in children/adolescents: Results of the I.Family Study. Exp Physiol 105, 347–356. [DOI] [PubMed] [Google Scholar]
- Ikegami K, Ogyu S, Arakomo Y, Suzuki K, Mafune K, Hiro H. and Nagata S. (2009) Recovery of cognitive performance and fatigue after one night of sleep deprivation. J Occup Health 51, 412–422. [DOI] [PubMed] [Google Scholar]
- Ingiosi AM, Schoch H, Wintler T. et al. (2019) Shank3 modulates sleep and expression of circadian transcription factors. Elife 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ivshina M, Lasko P. and Richter JD (2014) Cytoplasmic polyadenylation element binding proteins in development, health, and disease. Annu Rev Cell Dev Biol 30, 393–415. [DOI] [PubMed] [Google Scholar]
- Iwakawa HO and Tomari Y. (2015) The Functions of MicroRNAs: mRNA Decay and Translational Repression. Trends Cell Biol 25, 651–665. [DOI] [PubMed] [Google Scholar]
- Jiang Y, Gong NN, Hu XS, Ni MJ, Pasi R. and Matsunami H. (2015) Molecular profiling of activated olfactory neurons identifies odorant receptors for odors in vivo. Nat Neurosci 18, 1446–1454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kakamu T, Hidaka T, Masuishi Y, Kasuga H, Endo S, Sakurazawa M, Munakata Y, Tajimi K. and Fukushima T. (2021) Effect of occupation on sleep duration among daytime Japanese workers: A cross-sectional study. Medicine (Baltimore) 100, e28123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karabulut S, Korkmaz Bayramov K, Bayramov R, Ozdemir F, Topaloglu T, Ergen E, Yazgan K, Taskiran AS and Golgeli A. (2019) Effects of post-learning REM sleep deprivation on hippocampal plasticity-related genes and microRNA in mice. Behav Brain Res 361, 7–13. [DOI] [PubMed] [Google Scholar]
- Kaufmann T, Elvsashagen T, Alnaes D. et al. (2016) The brain functional connectome is robustly altered by lack of sleep. Neuroimage 127, 324–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Keren H, Lev-Maor G. and Ast G. (2010) Alternative splicing and evolution: diversification, exon definition and function. Nat Rev Genet 11, 345–355. [DOI] [PubMed] [Google Scholar]
- Kerkhof GA (2017) Epidemiology of sleep and sleep disorders in The Netherlands. Sleep Med 30, 229–239. [DOI] [PubMed] [Google Scholar]
- Kim CK and Pak TR (2020) miRNA degradation in the mammalian brain. Am J Physiol Cell Physiol 319, C624–C629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim HK, Pham MHC, Ko KS, Rhee BD and Han J. (2018) Alternative splicing isoforms in health and disease. Pflugers Arch 470, 995–1016. [DOI] [PubMed] [Google Scholar]
- Kindler S. and Kreienkamp HJ (2012) Dendritic mRNA targeting and translation. Adv Exp Med Biol 970, 285–305. [DOI] [PubMed] [Google Scholar]
- Knight ZA, Tan K, Birsoy K, Schmidt S, Garrison JL, Wysocki RW, Emiliano A, Ekstrand MI and Friedman JM (2012) Molecular profiling of activated neurons by phosphorylated ribosome capture. Cell 151, 1126–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kojima S, Sher-Chen EL and Green CB (2012) Circadian control of mRNA polyadenylation dynamics regulates rhythmic protein expression. Genes Dev 26, 2724–2736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Komar AA and Merrick WC (2020) A Retrospective on eIF2A-and Not the Alpha Subunit of eIF2. Int J Mol Sci 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Konakanchi S, Raavi V, Ml HK and Shankar Ms V. (2022) Effect of chronic sleep deprivation and sleep recovery on hippocampal CA3 neurons, spatial memory and anxiety-like behavior in rats. Neurobiol Learn Mem 187, 107559. [DOI] [PubMed] [Google Scholar]
- Koren T, Fisher E, Webster L, Livingston G. and Rapaport P. (2023) Prevalence of sleep disturbances in people with dementia living in the community: A systematic review and meta-analysis. Ageing Res Rev 83, 101782. [DOI] [PubMed] [Google Scholar]
- Kreutzmann JC, Havekes R, Abel T. and Meerlo P. (2015) Sleep deprivation and hippocampal vulnerability: changes in neuronal plasticity, neurogenesis and cognitive function. Neuroscience 309, 173–190. [DOI] [PubMed] [Google Scholar]
- Kriaucionis S. and Heintz N. (2009) The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain. Science 324, 929–930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krishnan HC, Gandour CE, Ramos JL, Wrinkle MC, Sanchez-Pacheco JJ and Lyons LC (2016a) Acute Sleep Deprivation Blocks Short- and Long-Term Operant Memory in Aplysia. Sleep 39, 2161–2171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krishnan HC, Noakes EJ and Lyons LC (2016b) Chronic sleep deprivation differentially affects short and long-term operant memory in Aplysia. Neurobiol Learn Mem 134 Pt B, 349–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuhn M, Wolf E, Maier JG et al. (2016) Sleep recalibrates homeostatic and associative synaptic plasticity in the human cortex. Nat Commun 7, 12455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lamon S, Morabito A, Arentson-Lantz E. et al. (2021) The effect of acute sleep deprivation on skeletal muscle protein synthesis and the hormonal environment. Physiol Rep 9, e14660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lamond N, Jay SM, Dorrian J, Ferguson SA, Jones C. and Dawson D. (2007) The dynamics of neurobehavioural recovery following sleep loss. J Sleep Res 16, 33–41. [DOI] [PubMed] [Google Scholar]
- Lankrew Ayalew TW, B.G., Haile KE (2022) Prevalence and associated factors of sleep deprivation among Haramaya University students, Ethiopia, 2021: cross-sectional study. Sleep Science Practice 6. [Google Scholar]
- Lee Y, Park J, Kim SM, Nam K, Lee W, Cho H. and Han IO (2020) Sleep deprivation impairs learning and memory by decreasing protein O-GlcNAcylation in the brain of adult zebrafish. FASEB J 34, 853–864. [DOI] [PubMed] [Google Scholar]
- Levendowski DJ, Walsh CM, Boeve BF et al. (2022) Non-REM sleep with hypertonia in Parkinsonian Spectrum Disorders: A pilot investigation. Sleep Med 100, 501–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levenson JM, O’Riordan KJ, Brown KD, Trinh MA, Molfese DL and Sweatt JD (2004) Regulation of histone acetylation during memory formation in the hippocampus. J Biol Chem 279, 40545–40559. [DOI] [PubMed] [Google Scholar]
- Li D, McIntosh CS, Mastaglia FL, Wilton SD and Aung-Htut MT (2021a) Neurodegenerative diseases: a hotbed for splicing defects and the potential therapies. Transl Neurodegener 10, 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li W. and Liu M. (2011) Distribution of 5-hydroxymethylcytosine in different human tissues. J Nucleic Acids 2011, 870726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li W, Ma L, Yang G. and Gan WB (2017) REM sleep selectively prunes and maintains new synapses in development and learning. Nat Neurosci 20, 427–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Zhang W, Liu M, Zhang Q, Lin Z, Jia M, Liu D. and Lin L. (2021b) Imbalance of Autophagy and Apoptosis Induced by Oxidative Stress May Be Involved in Thyroid Damage Caused by Sleep Deprivation in Rats. Oxid Med Cell Longev 2021, 5645090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin PY, Kavalali ET and Monteggia LM (2018) Genetic Dissection of Presynaptic and Postsynaptic BDNF-TrkB Signaling in Synaptic Efficacy of CA3-CA1 Synapses. Cell Rep 24, 1550–1561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lipton JO, Yuan ED, Boyle LM et al. (2015) The Circadian Protein BMAL1 Regulates Translation in Response to S6K1-Mediated Phosphorylation. Cell 161, 1138–1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu H, Huang X, Li Y, Xi K, Han Y, Mao H, Ren K, Wang W. and Wu Z. (2022a) TNF signaling pathway-mediated microglial activation in the PFC underlies acute paradoxical sleep deprivation-induced anxiety-like behaviors in mice. Brain Behav Immun 100, 254–266. [DOI] [PubMed] [Google Scholar]
- Liu X, Yu H, Wang Y, Li S, Cheng C, Al-Nusaif M. and Le W. (2022b) Altered Motor Performance, Sleep EEG, and Parkinson’s Disease Pathology Induced by Chronic Sleep Deprivation in Lrrk2(G2019S) Mice. Neurosci Bull 38, 1170–1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y, Hu W, Murakawa Y, Yin J, Wang G, Landthaler M. and Yan J. (2013) Cold-induced RNA-binding proteins regulate circadian gene expression by controlling alternative polyadenylation. Sci Rep 3, 2054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Louter M, Aarden WC, Lion J, Bloem BR and Overeem S. (2012) Recognition and diagnosis of sleep disorders in Parkinson’s disease. J Neurol 259, 2031–2040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lyons LC, Chatterjee S, Vanrobaeys Y, Gaine ME and Abel T. (2020) Translational changes induced by acute sleep deprivation uncovered by TRAP-Seq. Mol Brain 13, 165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma XM and Blenis J. (2009) Molecular mechanisms of mTOR-mediated translational control. Nat Rev Mol Cell Biol 10, 307–318. [DOI] [PubMed] [Google Scholar]
- Mackiewicz M, Shockley KR, Romer MA et al. (2007) Macromolecule biosynthesis: a key function of sleep. Physiol Genomics 31, 441–457. [DOI] [PubMed] [Google Scholar]
- Maggi G, Vitale C, Cerciello F. and Santangelo G. (2023) Sleep and wakefulness disturbances in Parkinson’s disease: A meta-analysis on prevalence and clinical aspects of REM sleep behavior disorder, excessive daytime sleepiness and insomnia. Sleep Med Rev 68, 101759. [DOI] [PubMed] [Google Scholar]
- Magnuson B, Ekim B. and Fingar DC (2012) Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks. Biochem J 441, 1–21. [DOI] [PubMed] [Google Scholar]
- Maher A, El Sayed N, Nafea H. and Gad MZ (2021) Rolipram rescues memory consolidation deficits caused by sleep deprivation: Implication of the cAMP/PKA and cAMP/Epac pathways. CNS Neurol Disord Drug Targets. [DOI] [PubMed] [Google Scholar]
- Marcheva B, Perelis M, Weidemann BJ et al. (2020) A role for alternative splicing in circadian control of exocytosis and glucose homeostasis. Genes Dev 34, 1089–1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maret S, Faraguna U, Nelson AB, Cirelli C. and Tononi G. (2011) Sleep and waking modulate spine turnover in the adolescent mouse cortex. Nat Neurosci 14, 1418–1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marti AR, Patil S, Mrdalj J, Meerlo P, Skrede S, Pallesen S, Pedersen TT, Bramham CR and Gronli J. (2017) No Escaping the Rat Race: Simulated Night Shift Work Alters the Time-of-Day Variation in BMAL1 Translational Activity in the Prefrontal Cortex. Front Neural Circuits 11, 70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Massart R, Freyburger M, Suderman M. et al. (2014) The genome-wide landscape of DNA methylation and hydroxymethylation in response to sleep deprivation impacts on synaptic plasticity genes. Transl Psychiatry 4, e347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McDermott CM, LaHoste GJ, Chen C, Musto A, Bazan NG and Magee JC (2003) Sleep deprivation causes behavioral, synaptic, and membrane excitability alterations in hippocampal neurons. J Neurosci 23, 9687–9695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McGlincy NJ, Valomon A, Chesham JE, Maywood ES, Hastings MH and Ule J. (2012) Regulation of alternative splicing by the circadian clock and food related cues. Genome Biol 13, R54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McQuown SC, Barrett RM, Matheos DP et al. (2011) HDAC3 is a critical negative regulator of long-term memory formation. J Neurosci 31, 764–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mehnert KI, Beramendi A, Elghazali F, Negro P, Kyriacou CP and Cantera R. (2007) Circadian changes in Drosophila motor terminals. Dev Neurobiol 67, 415–421. [DOI] [PubMed] [Google Scholar]
- Mehnert KI and Cantera R. (2011) Circadian rhythms in the morphology of neurons in Drosophila. Cell Tissue Res 344, 381–389. [DOI] [PubMed] [Google Scholar]
- Meng H, Cao Y, Qin J, Song X, Zhang Q, Shi Y. and Cao L. (2015) DNA methylation, its mediators and genome integrity. Int J Biol Sci 11, 604–617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Minakawa EN, Miyazaki K, Maruo K, Yagihara H, Fujita H, Wada K. and Nagai Y. (2017) Chronic sleep fragmentation exacerbates amyloid beta deposition in Alzheimer’s disease model mice. Neurosci Lett 653, 362–369. [DOI] [PubMed] [Google Scholar]
- Mohammadipoor-Ghasemabad L, Sangtarash MH, Sheibani V, Sasan HA and Esmaeili-Mahani S. (2019) Hippocampal microRNA-191a-5p Regulates BDNF Expression and Shows Correlation with Cognitive Impairment Induced by Paradoxical Sleep Deprivation. Neuroscience 414, 49–59. [DOI] [PubMed] [Google Scholar]
- Monico-Neto M, Giampa SQ, Lee KS et al. (2015) Negative energy balance induced by paradoxical sleep deprivation causes multicompartmental changes in adipose tissue and skeletal muscle. Int J Endocrinol 2015, 908159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mu Q, Mishory A, Johnson KA, Nahas Z, Kozel FA, Yamanaka K, Bohning DE and George MS (2005) Decreased brain activation during a working memory task at rested baseline is associated with vulnerability to sleep deprivation. Sleep 28, 433–446. [DOI] [PubMed] [Google Scholar]
- Mullington JM, Haack M, Toth M, Serrador JM and Meier-Ewert HK (2009) Cardiovascular, inflammatory, and metabolic consequences of sleep deprivation. Prog Cardiovasc Dis 51, 294–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nagai H, de Vivo L, Marshall W, Tononi G. and Cirelli C. (2021) Effects of Severe Sleep Disruption on the Synaptic Ultrastructure of Young Mice. eNeuro 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Naidoo N, Giang W, Galante RJ and Pack AI (2005) Sleep deprivation induces the unfolded protein response in mouse cerebral cortex. J Neurochem 92, 1150–1157. [DOI] [PubMed] [Google Scholar]
- Nakanishi H, Sun Y, Nakamura RK et al. (1997) Positive correlations between cerebral protein synthesis rates and deep sleep in Macaca mulatta. Eur J Neurosci 9, 271–279. [DOI] [PubMed] [Google Scholar]
- Nectow AR, Moya MV, Ekstrand MI et al. (2017) Rapid Molecular Profiling of Defined Cell Types Using Viral TRAP. Cell Rep 19, 655–667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nilsson EK, Bostrom AE, Mwinyi J. and Schioth HB (2016) Epigenomics of Total Acute Sleep Deprivation in Relation to Genome-Wide DNA Methylation Profiles and RNA Expression. OMICS 20, 334–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Noya SB, Colameo D, Bruning F. et al. (2019) The forebrain synaptic transcriptome is organized by clocks but its proteome is driven by sleep. Science 366. [DOI] [PubMed] [Google Scholar]
- Nsengimana A, Mugabo E, Niyonsenga J, Hategekimana JC, Biracyaza E, Mutarambirwa R, Ngabo E. and Nduwayezu R. (2023) Sleep quality among undergraduate medical students in Rwanda: a comparative study. Sci Rep 13, 265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ochab JK, Szwed J, Oles K. et al. (2021) Observing changes in human functioning during induced sleep deficiency and recovery periods. PLoS One 16, e0255771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oliveira MM and Klann E. (2022) eIF2-dependent translation initiation: Memory consolidation and disruption in Alzheimer’s disease. Semin Cell Dev Biol 125, 101–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Owen JE, Zhu Y, Fenik P, Zhan G, Bell P, Liu C. and Veasey S. (2021) Late-in-life neurodegeneration after chronic sleep loss in young adult mice. Sleep 44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan Q, Shai O, Lee LJ, Frey BJ and Blencowe BJ (2008) Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat Genet 40, 1413–1415. [DOI] [PubMed] [Google Scholar]
- Pandey K, Sharma KP and Sharma SK (2015) Histone deacetylase inhibition facilitates massed pattern-induced synaptic plasticity and memory. Learn Mem 22, 514–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pao PC and Tsai LH (2022) Histone Deacetylases 1 and 2 in Memory Function. ACS Chem Neurosci 13, 848–858. [DOI] [PubMed] [Google Scholar]
- Park I, Kim D, Kim J, Jang S, Choi M, Choe HK, Choe Y. and Kim K. (2020) microRNA-25 as a novel modulator of circadian Period2 gene oscillation. Exp Mol Med 52, 1614–1626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parnell AA, De Nobrega AK and Lyons LC (2021) Translating around the clock: Multi-level regulation of post-transcriptional processes by the circadian clock. Cell Signal 80, 109904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pejovic S, Basta M, Vgontzas AN et al. (2013) Effects of recovery sleep after one work week of mild sleep restriction on interleukin-6 and cortisol secretion and daytime sleepiness and performance. Am J Physiol Endocrinol Metab 305, E890–896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peng Y, Wang W, Tan T, He W, Dong Z, Wang YT and Han H. (2016) Maternal sleep deprivation at different stages of pregnancy impairs the emotional and cognitive functions, and suppresses hippocampal long-term potentiation in the offspring rats. Mol Brain 9, 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Periasamy S, Hsu DZ, Fu YH and Liu MY (2015) Sleep deprivation-induced multi-organ injury: role of oxidative stress and inflammation. EXCLI J 14, 672–683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pesoli M, Rucco R, Liparoti M. et al. (2022) A night of sleep deprivation alters brain connectivity and affects specific executive functions. Neurol Sci 43, 1025–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pinheiro-da-Silva J, Silva PF, Nogueira MB and Luchiari AC (2017a) Sleep deprivation effects on object discrimination task in zebrafish (Danio rerio). Anim Cogn 20, 159–169. [DOI] [PubMed] [Google Scholar]
- Pinheiro-da-Silva J, Tran S. and Luchiari AC (2018) Sleep deprivation impairs cognitive performance in zebrafish: A matter of fact? Behav Processes 157, 656–663. [DOI] [PubMed] [Google Scholar]
- Pinheiro-da-Silva J, Tran S, Silva PF and Luchiari AC (2017b) Good night, sleep tight: The effects of sleep deprivation on spatial associative learning in zebrafish. Pharmacol Biochem Behav 159, 36–47. [DOI] [PubMed] [Google Scholar]
- Poroyko VA, Carreras A, Khalyfa A. et al. (2016) Chronic Sleep Disruption Alters Gut Microbiota, Induces Systemic and Adipose Tissue Inflammation and Insulin Resistance in Mice. Sci Rep 6, 35405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Puentes-Mestril C, Delorme J, Wang L, Donnelly M, Popke D, Jiang S. and Aton SJ (2021) Sleep Loss Drives Brain Region-Specific and Cell Type-Specific Alterations in Ribosome-Associated Transcripts Involved in Synaptic Plasticity and Cellular Timekeeping. J Neurosci 41, 5386–5398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Radhakrishnan A. and Green R. (2016) Connections Underlying Translation and mRNA Stability. J Mol Biol 428, 3558–3564. [DOI] [PubMed] [Google Scholar]
- Rahmani M, Rahmani F. and Rezaei N. (2020) The Brain-Derived Neurotrophic Factor: Missing Link Between Sleep Deprivation, Insomnia, and Depression. Neurochem Res 45, 221–231. [DOI] [PubMed] [Google Scholar]
- Ramm P. and Smith CT (1990) Rates of cerebral protein synthesis are linked to slow wave sleep in the rat. Physiol Behav 48, 749–753. [DOI] [PubMed] [Google Scholar]
- Rasmussen MK, Mestre H. and Nedergaard M. (2018) The glymphatic pathway in neurological disorders. Lancet Neurol 17, 1016–1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ravassard P, Pachoud B, Comte JC et al. (2009) Paradoxical (REM) sleep deprivation causes a large and rapidly reversible decrease in long-term potentiation, synaptic transmission, glutamate receptor protein levels, and ERK/MAPK activation in the dorsal hippocampus. Sleep 32, 227–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raven F, Meerlo P, Van der Zee EA, Abel T. and Havekes R. (2019) A brief period of sleep deprivation causes spine loss in the dentate gyrus of mice. Neurobiol Learn Mem 160, 83–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sabia S, Fayosse A, Dumurgier J, van Hees VT, Paquet C, Sommerlad A, Kivimaki M, Dugravot A. and Singh-Manoux A. (2021) Association of sleep duration in middle and old age with incidence of dementia. Nat Commun 12, 2289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sallinen M, Harma M, Akila R, Holm A, Luukkonen R, Mikola H, Muller K. and Virkkala J. (2004) The effects of sleep debt and monotonous work on sleepiness and performance during a 12-h dayshift. J Sleep Res 13, 285–294. [DOI] [PubMed] [Google Scholar]
- Saner NJ, Lee MJ, Pitchford NW et al. (2020) The effect of sleep restriction, with or without high-intensity interval exercise, on myofibrillar protein synthesis in healthy young men. J Physiol 598, 1523–1536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanz E, Bean JC, Carey DP, Quintana A. and McKnight GS (2019) RiboTag: Ribosomal Tagging Strategy to Analyze Cell-Type-Specific mRNA Expression In Vivo. Curr Protoc Neurosci 88, e77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanz E, Yang L, Su T, Morris DR, McKnight GS and Amieux PS (2009) Cell-type-specific isolation of ribosome-associated mRNA from complex tissues. Proc Natl Acad Sci U S A 106, 13939–13944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scarpa JR, Jiang P, Gao VD et al. (2018) Cross-species systems analysis identifies gene networks differentially altered by sleep loss and depression. Sci Adv 4, eaat1294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmidt EK, Clavarino G, Ceppi M. and Pierre P. (2009) SUnSET, a nonradioactive method to monitor protein synthesis. Nat Methods 6, 275–277. [DOI] [PubMed] [Google Scholar]
- Schratt G. (2009) microRNAs at the synapse. Nat Rev Neurosci 10, 842–849. [DOI] [PubMed] [Google Scholar]
- Schwich OD, Blumel N, Keller M. et al. (2021) SRSF3 and SRSF7 modulate 3’UTR length through suppression or activation of proximal polyadenylation sites and regulation of CFIm levels. Genome Biol 22, 82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sei H, Saitoh D, Yamamoto K, Morita K. and Morita Y. (2000) Differential effect of short-term REM sleep deprivation on NGF and BDNF protein levels in the rat brain. Brain Res 877, 387–390. [DOI] [PubMed] [Google Scholar]
- Seugnet L, Galvin JE, Suzuki Y, Gottschalk L. and Shaw PJ (2009) Persistent short-term memory defects following sleep deprivation in a drosophila model of Parkinson disease. Sleep 32, 984–992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi L, Chen SJ, Ma MY, Bao YP, Han Y, Wang YM, Shi J, Vitiello MV and Lu L. (2018) Sleep disturbances increase the risk of dementia: A systematic review and meta-analysis. Sleep Med Rev 40, 4–16. [DOI] [PubMed] [Google Scholar]
- Simpson N. and Dinges DF (2007) Sleep and inflammation. Nutr Rev 65, S244–252. [DOI] [PubMed] [Google Scholar]
- Skuladottir GV, Nilsson EK, Mwinyi J. and Schioth HB (2016) One-night sleep deprivation induces changes in the DNA methylation and serum activity indices of stearoyl-CoA desaturase in young healthy men. Lipids Health Dis 15, 137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song M. (2020) miRNAs-dependent regulation of synapse formation and function. Genes Genomics 42, 837–845. [DOI] [PubMed] [Google Scholar]
- Spira AP, Chen-Edinboro LP, Wu MN and Yaffe K. (2014) Impact of sleep on the risk of cognitive decline and dementia. Curr Opin Psychiatry 27, 478–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spira AP, Gamaldo AA, An Y. et al. (2013) Self-reported sleep and beta-amyloid deposition in community-dwelling older adults. JAMA Neurol 70, 1537–1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steward O. and Worley PF (2001) A cellular mechanism for targeting newly synthesized mRNAs to synaptic sites on dendrites. Proc Natl Acad Sci U S A 98, 7062–7068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stranges S, Tigbe W, Gomez-Olive FX, Thorogood M. and Kandala NB (2012) Sleep problems: an emerging global epidemic? Findings from the INDEPTH WHO-SAGE study among more than 40,000 older adults from 8 countries across Africa and Asia. Sleep 35, 1173–1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Streatfeild J, Smith J, Mansfield D, Pezzullo L. and Hillman D. (2021) The social and economic cost of sleep disorders. Sleep 44. [DOI] [PubMed] [Google Scholar]
- Sun YY, Wang Z, Zhou HY and Huang HC (2022) Sleep-Wake Disorders in Alzheimer’s Disease: A Review. ACS Chem Neurosci 13, 1467–1478. [DOI] [PubMed] [Google Scholar]
- Suzuki A, Sinton CM, Greene RW and Yanagisawa M. (2013) Behavioral and biochemical dissociation of arousal and homeostatic sleep need influenced by prior wakeful experience in mice. Proc Natl Acad Sci U S A 110, 10288–10293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terao A, Greco MA, Davis RW, Heller HC and Kilduff TS (2003) Region-specific changes in immediate early gene expression in response to sleep deprivation and recovery sleep in the mouse brain. Neuroscience 120, 1115–1124. [DOI] [PubMed] [Google Scholar]
- Thompson CL, Wisor JP, Lee CK et al. (2010) Molecular and anatomical signatures of sleep deprivation in the mouse brain. Front Neurosci 4, 165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thoreen CC, Chantranupong L, Keys HR, Wang T, Gray NS and Sabatini DM (2012) A unifying model for mTORC1-mediated regulation of mRNA translation. Nature 485, 109–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tobaldini E, Costantino G, Solbiati M, Cogliati C, Kara T, Nobili L. and Montano N. (2017) Sleep, sleep deprivation, autonomic nervous system and cardiovascular diseases. Neurosci Biobehav Rev 74, 321–329. [DOI] [PubMed] [Google Scholar]
- Tsao HS, Gjelsvik A, Sojar S. and Amanullah S. (2021) Sounding the Alarm on Sleep: A Negative Association Between Inadequate Sleep and Flourishing. J Pediatr 228, 199–207 e193. [DOI] [PubMed] [Google Scholar]
- Tudor JC, Davis EJ, Peixoto L. et al. (2016) Sleep deprivation impairs memory by attenuating mTORC1-dependent protein synthesis. Sci Signal 9, ra41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uyhelji HA, Kupfer DM, White VL, Jackson ML, Van Dongen HPA and Burian DM (2018) Exploring gene expression biomarker candidates for neurobehavioral impairment from total sleep deprivation. BMC Genomics 19, 341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vanderheyden WM, Gerstner JR, Tanenhaus A, Yin JC and Shaw PJ (2013) ERK phosphorylation regulates sleep and plasticity in Drosophila. PLoS One 8, e81554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vanderheyden WM, Lim MM, Musiek ES and Gerstner JR (2018) Alzheimer’s Disease and Sleep-Wake Disturbances: Amyloid, Astrocytes, and Animal Models. J Neurosci 38, 2901–2910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vargas-Lopez V, Lamprea MR and Munera A. (2016) Histone deacetylase inhibition abolishes stress-induced spatial memory impairment. Neurobiol Learn Mem 134 Pt B, 328–338. [DOI] [PubMed] [Google Scholar]
- Varghese NE, Lugo A, Ghislandi S, Colombo P, Pacifici R. and Gallus S. (2020) Sleep dissatisfaction and insufficient sleep duration in the Italian population. Sci Rep 10, 17943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vecsey CG, Baillie GS, Jaganath D. et al. (2009) Sleep deprivation impairs cAMP signalling in the hippocampus. Nature 461, 1122–1125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vecsey CG, Huang T. and Abel T. (2018) Sleep deprivation impairs synaptic tagging in mouse hippocampal slices. Neurobiol Learn Mem 154, 136–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vecsey CG, Peixoto L, Choi JH et al. (2012) Genomic analysis of sleep deprivation reveals translational regulation in the hippocampus. Physiol Genomics 44, 981–991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vuong CK, Black DL and Zheng S. (2016) The neurogenetics of alternative splicing. Nat Rev Neurosci 17, 265–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C. and Holtzman DM (2020) Bidirectional relationship between sleep and Alzheimer’s disease: role of amyloid, tau, and other factors. Neuropsychopharmacology 45, 104–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C, Liu J, Li Z. et al. (2019) Predictor of sleep difficulty among community dwelling older populations in 2 African settings. Medicine (Baltimore) 98, e17971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang CS, Kavalali ET and Monteggia LM (2022) BDNF signaling in context: From synaptic regulation to psychiatric disorders. Cell 185, 62–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, Fan Z, Zhao M, Li J, Lu M, Liu W, Ying H, Liu M. and Yan J. (2016) Oscillating primary transcripts harbor miRNAs with circadian functions. Sci Rep 6, 21598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei CW, Luo T, Zou SS and Wu AS (2017) Research progress on the roles of microRNAs in governing synaptic plasticity, learning and memory. Life Sci 188, 118–122. [DOI] [PubMed] [Google Scholar]
- Weigend S, Holst SC, Meier J, Brock M, Kohler M. and Landolt HP (2019) Prolonged Waking and Recovery Sleep Affect the Serum MicroRNA Expression Profile in Humans. Clocks Sleep 1, 75–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wheaton AG, Jones SE, Cooper AC and Croft JB (2018) Short Sleep Duration Among Middle School and High School Students - United States, 2015. MMWR Morb Mortal Wkly Rep 67, 85–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wimmer ME, Cui R, Blackwell JM and Abel T. (2021) Cyclic AMP response element-binding protein is required in excitatory neurons in the forebrain to sustain wakefulness. Sleep 44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wong LW, Chong YS, Wong WLE and Sajikumar S. (2020) Inhibition of Histone Deacetylase Reinstates Hippocampus-Dependent Long-Term Synaptic Plasticity and Associative Memory in Sleep-Deprived Mice. Cereb Cortex 30, 4169–4182. [DOI] [PubMed] [Google Scholar]
- Wong LW, Tann JY, Ibanez CF and Sajikumar S. (2019) The p75 Neurotrophin Receptor Is an Essential Mediator of Impairments in Hippocampal-Dependent Associative Plasticity and Memory Induced by Sleep Deprivation. J Neurosci 39, 5452–5465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu H, Dunnett S, Ho YS and Chang RC (2019) The role of sleep deprivation and circadian rhythm disruption as risk factors of Alzheimer’s disease. Front Neuroendocrinol 54, 100764. [DOI] [PubMed] [Google Scholar]
- Xu ZQ, Gao CY, Fang CQ, Zhou HD and Jiang XJ (2010) The mechanism and characterization of learning and memory impairment in sleep-deprived mice. Cell Biochem Biophys 58, 137–140. [DOI] [PubMed] [Google Scholar]
- Yamazaki EM, Antler CA, Casale CE, MacMullen LE, Ecker AJ and Goel N. (2021) Cortisol and C-Reactive Protein Vary During Sleep Loss and Recovery but Are Not Markers of Neurobehavioral Resilience. Front Physiol 12, 782860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang G, Lai CS, Cichon J, Ma L, Li W. and Gan WB (2014) Sleep promotes branch-specific formation of dendritic spines after learning. Science 344, 1173–1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yi YW, You KS, Park JS, Lee SG and Seong YS (2021) Ribosomal Protein S6: A Potential Therapeutic Target against Cancer? Int J Mol Sci 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yin M, Chen Y, Zheng H, Pu T, Marshall C, Wu T. and Xiao M. (2017) Assessment of mouse cognitive and anxiety-like behaviors and hippocampal inflammation following a repeated and intermittent paradoxical sleep deprivation procedure. Behav Brain Res 321, 69–78. [DOI] [PubMed] [Google Scholar]
- Yoo SS, Hu PT, Gujar N, Jolesz FA and Walker MP (2007) A deficit in the ability to form new human memories without sleep. Nat Neurosci 10, 385–392. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Xie B, Chen X, Zhang J. and Yuan S. (2021) A key role of gut microbiota-vagus nerve/spleen axis in sleep deprivation-mediated aggravation of systemic inflammation after LPS administration. Life Sci 265, 118736. [DOI] [PubMed] [Google Scholar]
- Zhou L, Miller C, Miraglia LJ, Romero A, Mure LS, Panda S. and Kay SA (2021) A genome-wide microRNA screen identifies the microRNA-183/96/182 cluster as a modulator of circadian rhythms. Proc Natl Acad Sci U S A 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu Y, Zhan G, Fenik P, Brandes M, Bell P, Francois N, Shulman K. and Veasey S. (2018) Chronic Sleep Disruption Advances the Temporal Progression of Tauopathy in P301S Mutant Mice. J Neurosci 38, 10255–10270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zielinski MR, McKenna JT and McCarley RW (2016) Functions and Mechanisms of Sleep. AIMS Neurosci 3, 67–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data analyzed in the current study is available at GEO series accession GSE156925 with the analysis pipeline at https://github.com/YannVRB/Review-Sleep-Deprivation-Journal-of-Neurochemistry.git.




