Skip to main content
F1000Research logoLink to F1000Research
. 2020 Mar 27;9:F1000 Faculty Rev-214. [Version 1] doi: 10.12688/f1000research.22028.1

Recent advances in understanding the genetics of sleep

Maxime Jan 1, Bruce F O'Hara 2, Paul Franken 1,a
PMCID: PMC7104869  PMID: 32274013

Abstract

Sleep is a ubiquitous and complex behavior in both its manifestation and regulation. Despite its essential role in maintaining optimal performance, health, and well-being, the genetic mechanisms underlying sleep remain poorly understood. Here, we review the forward genetic approaches undertaken in the last four years to elucidate the genes and gene pathways affecting sleep and its regulation. Despite an increasing number of studies and mining large databases, a coherent picture on “sleep” genes has yet to emerge. We highlight the results achieved by using unbiased genetic screens mainly in humans, mice, and fruit flies with an emphasis on normal sleep and make reference to lessons learned from the circadian field.

Keywords: Sleep, Sleep Regulation, Genetics, Genetic Screen, Forward genetics, GWAS, GRP

Introduction

Through genetic analyses of sleep, we hope to learn about the molecular substrates of this remarkable behavior and its pathologies. Sleep is remarkable because we spend a considerable portion of our lives in this state (>24 years for most of us) yet we know little about its likely numerous functions. Clearly, sleep is important as lack of sleep, poor-quality sleep, or sleep disorders greatly impact performance and well-being and may lead to poor health. Genetics can be used to approach a variety of questions about sleep regulation, sleep function, abnormal sleep, and the consequences of poor or insufficient sleep. For example, research aimed at discovering the gene variants altering susceptibility to the deleterious effects of sleep loss and the molecular pathways activated by sleep loss are two areas of great interest, from both a basic scientific understanding and a clinical point of view.

With perhaps some envy, sleep geneticists look at the astounding progress made in the closely related circadian rhythms research field. Results from mutagenesis screens in flies and mice for circadian phenotypes 1, 2, mostly variations in circadian period length, led to the discovery of a core set of interacting genes (that is, “clock genes”) that pace the clock 3 and to a Nobel prize in 2017 ( www.nobelprize.org/prizes/medicine/2017/prize-announcement/). With this promise, mutagenesis screens have been applied to the problem of sleep, and although important new insights have been gained, a core set of genes regulating sleep (apart from those involved in sleep’s circadian modulation) remains to be identified. Perhaps one reason hampering breakthrough progress matching that in the circadian field is that sleep is a circuit-driven behavior whereas circadian rhythms rely on cell-autonomous processes and thus are more tractable in terms of identifying key players through genetic dissection. Moreover, sleep is not one phenotype but a multi-faceted behavior starting off with two very different sleep states—rapid eye movement (REM) sleep and non-REM (NREM) sleep—that seem preserved across vertebrates 4 and that each have their distinct (neuro)physiology, regulation, and functions. In a recent study, we quantified well over 300 sleep-related phenotypes in a genetic reference population (GRP) of mice 5. These phenotypes included time spent in the various sleep-wake states, their distribution over the day, and the frequency- and amplitude-specific characteristics of the electroencephalogram (EEG), all under undisturbed baseline conditions and after a period of enforced waking. The median heritability in this comprehensive sleep-wake phenome was high (h 2 = 0.68), and although a number of phenotypes were highly correlated, many among them proved to be independent sleep traits each with their distinct polygenic mode of inheritance. It is therefore unrealistic to aim at finding core “sleep genes”; instead, we expect to uncover sets of genes or gene pathways each modulating specific aspects of sleep. Such genes could affect sleep traits directly or could do so indirectly through early (or late) processes of neurodevelopment and neuroplasticity. In addition to its phenotypic complexity, sleep varies with life history and environment and can differ greatly among animal species according to their ecological niche. Therefore, genetic analysis of sleep in species other than the established model species that have been used thus far could help delineate conserved genetic pathways regulation sleep 6.

With this in mind, this brief overview of the progress in the field will focus on the recent efforts to discover the genetic underpinnings of sleep in various species and mainly concern flies, mice, and humans with some reference to zebrafish and Caenorhabditis elegans work. Genetics is a broad term and is used differently among fields; for example, we will have to ignore optogenetics although this technique has revolutionized sleep research in the last 15 years in dissecting its underlying neuro-circuitry 7, 8. Similarly, we will not be comprehensive in reporting on the ever-growing list of candidate genes that have been genetically targeted to study their specific role in aspects of sleep, although much has been learned using this approach. This “gene-to-phenotype” approach is referred to as reverse genetics. In reply, classic genetic approaches (from phenotype to gene), which include the mutagenesis screens mentioned above, have become known as forward genetic approaches. However, these methods can clearly overlap and the distinction depends on scale; a large screen of knockout mice for known single genes could be considered a large-scale reverse genetic approach but is really more similar to a mutagenesis screen, as we will discuss below. Here, progress in the last four years (2016–2019) using unbiased gene discovery approaches will be reviewed with a strong emphasis on studies focusing on “normal” sleep and its regulation as opposed to sleep-related disorders.

Forward genetic screens for induced genetic variants

In forward genetic screens, changes in gene function are induced more or less randomly 9 throughout the genome. Mutations affecting gene function have been made through the use of chemicals—for example, N-ethyl-N-nitrosourea (ENU) or ethyl methanesulfonate (EMS)—or, in the fruit fly, Drosophila melanogaster through insertion of transposable P elements. Screens for the effects of overexpression of genes can be achieved by making use of the GAL4-upstream activation sequence (GAL4-UAS) system 10 or through injection of plasmids containing inducible overexpression vectors 11. Phenotyping of a large number of animals will identify lines with aberrant phenotypes caused by single genes.

The earliest sleep screen was performed in Drosophila 12. In that study, about 9,000 mutagenized lines were phenotyped for extreme short sleep time. Flies carrying loss-of-function mutations for Shaker, a gene encoding a voltage-dependent potassium channel, were found to sleep exceptionally little. Surprisingly, after screening several thousand additional mutagenized lines, a later Drosophila sleep screen identified the gene Sleepless, which proved to be a regulator of Shaker 13. Together, the two studies suggest neuronal membrane excitability as a core feature of homeostatic sleep drive 13, 14.

In mammals, the first mutagenesis sleep screen was published in 2016 15. ENU was used to induce random point mutations as 8,000 mice were screened for sleep abnormalities. A mutation in a single phosphorylation site in the protein kinase Sik3 was found to increasehomeostatic sleep drive concomitantly with increased time spent in NREM sleep 16. SIK3 was found to associate specifically with phosphoproteins for which the phosphorylation state was found to be sleep-wake–driven, suggesting that the sleep homeostatic process represents a phosphorylation (during wakefulness)/dephosphorylation (during NREM sleep) cycle 17, 18. Moreover, in the same screen, a missense mutation in Nalcn, a voltage-independent and non-selective cation channel, reduced time spent in REM sleep, and a point mutation in Cacna1a, encoding a voltage-dependent calcium channel, reduced wakefulness by about 70 minutes per day 19. While other genes discovered with this screen are likely to be in the pipeline, the yield thus far seems disappointing given the large number of mice phenotyped.

In C. elegans, forward genetic screens for sleep have been performed as well. In these nematodes, periods of sleep can be observed at the transition between developmental stages (developmentally timed sleep) or sleep can be induced by stress (stress-induced sleep) 20. Upon stress, the release of the neuropeptide Flp-13 (FMRFamide-like peptide-13) was shown to mediate the sleep-promoting effect 21. In an EMS mutagenesis screen in Flp-13–overexpressing worms for genes modifying the sleep-promoting effect of Flp-13, its receptor Dmsr-1, encoding a G protein–coupled receptor, was identified 22, 23. The G-protein that is coupled to Dmsr-1 may be the G i/o alpha subunit Goa-1, which was identified in another mutagenesis screen for genes regulating developmentally timed sleep, along with Gpb-2, encoding another G-protein subunit 24. Together, these results confirm the importance of G-protein signaling pathways in worm sleep.

Screens in which the effects of gene overexpression on sleep were studied were performed in Drosophila and zebrafish. By phenotyping about 12,000 fly lines for sleep duration Nemuri, encoding an uncharacterized antimicrobial peptide, was identified, which when overexpressed in adults induced sleep, thereby linking sleep and the immune system 10. In a zebrafish-overexpression screen for secreted proteins, the neuropeptide Neuromedin U ( Nmu) was identified to regulate sleep-wake behavior 11. Interestingly, anatomical and functional analyses found that Nmu-induced arousal is mediated by corticotropin-releasing hormone (CRH) signaling in the brainstem specifically, independent of the role of CRH in the hypothalamic–pituitary–adrenal axis. Also, overexpression of the neuropeptide Neuropeptide Y (NPY) in this screen was found to affect sleep and the authors could establish that this effect was mediated through inhibition of noradrenergic signaling known to promote wakefulness 25.

The ongoing efforts of the International Knockout Mouse Consortium (IKMC) to “knock out” each protein-coding gene and of the International Mouse Phenotyping Consortium (IMPC) to determine the phenotypic consequences 26 should be mentioned here because this (in principle) “reverse genetic” approach must be considered “forward” given its generally unbiased and genome-wide character, as already pointed out above. Sleep is part of the IMPC phenotyping pipeline and thus far 512 null-mutant strains have been evaluated; 72 of these displayed aberrant values for some of the 13 sleep-related variables quantified ( www.mousephenotype.org). These results suggest that a high percentage of gene products influence sleep in some way, including breathing rates during sleep ( https://doi.org/10.1101/517680). Several of the genes identified, such as Ppp1r9b, Pitx3, Ap4e1, and Myh1, affected time spent asleep to a similar degree as found in mutagenesis screens, suggesting that both mutagenesis and screens of knockout mice have found only a very small fraction of the genes that can influence sleep when mutated or eliminated. This again supports the notion that no core set of genes may underlie sleep regulation, at least to the same degree found for clock genes that underlie circadian rhythm generation. However, as discussed below, the transcriptional/translational oscillation of clock genes, especially outside the suprachiasmatic nuclei (SCN), appears to play a key role in sleep homeostasis as well (and a number of other functions).

The IKMC uses a combination of gene trapping and gene targeting in C57BL/6N mouse embryonic stem cells to mutate protein-coding genes. With CRISPR-Cas9, a more efficient technology has become available to target genes and might be used in genome-wide screens 27. Already, through the use of a triple-target CRISPR approach to generate bi-allelic knockouts in a single generation, targeting entire gene families has become feasible and applied to sleep (for example, acetylcholine receptors, N-methyl-D-aspartate receptors, and Ca 2+-dependent K + channels 2830) with some striking results; mice lacking the muscarinic acetylcholine receptors, Chrm1 and Chrm3, were also found to lack REM sleep 28.

Forward genetic mapping of natural genetic variants involved in sleep

The above-mentioned forward genetic screens report on the effects of single genes on a mostly uniform genetic background. However, many sleep traits are known to be genetically complex and each contributing gene usually explains but a small fraction of the overall phenotypic variance. Examples from the circadian field powerfully illustrate that genetic background matters. The first circadian clock gene identified in a mammalian mutagenesis was Clock 2. The ENU-induced Clock Δ19 mutation discovered in that study caused circadian period lengthening and periodicity loss in mice of the C57BL/6J background but would have gone unnoticed if the melatonin-producing C3H/HeJ strain had been used in the screen instead 31. Moreover, linkage analysis of circadian traits quantified in a C57BL/6J × BALB/cJ - F2 population identified a number of quantitative trait loci (QTLs) that did not map to known clock genes 32, suggesting not only that many genes important in circadian rhythm generation await their discovery but also that some of the genes identified through mutagenesis might be invariant in natural populations and not contribute to phenotypic variability.

Although the phenotypic variance explained by QTLs underlying complex traits is generally small, which makes identifying the causative genes difficult, the above findings convinced sleep researchers of the necessity to study natural populations or population panels constructed through selective breeding, such as GRPs, to map the natural genetic variants and their interactions underlying differences in sleep traits. With this aim, genome-wide association (GWA) studies have been performed in flies, mice, and humans. Although the genetic contribution over environmental factors is generally high (h 2 >0.5 for many sleep-wake traits and h 2 >0.8 for a number of EEG-related traits 5, 33, 34), it arises from many possible genetic variants (so far, 650 million variations have been identified in human populations 35) and their interactions. This very large genotypic diversity and the small effect size of each necessitates sample sizes of several thousands to reach the P value threshold required to identify a variant significantly associated with a given trait (usually set to P <5 × 10 −8), even when the interactions among them are ignored (epistasis 36). This is a problem for almost all normal and pathological traits that have been studied, including sleep. To address this problem, GRPs have been constructed in model species (see below).

The first “sleep” GWA study in humans was a case control study for the sleep-related disorder restless legs syndrome (RLS) and identified a number of genetic risk variants each conferring only minor increases in disease risk 37, 38. To date, six such RLS risk-conferring genomic loci have been found. Among those, functional contributions have been demonstrated in follow-up studies, including the use of mouse models, for the transcription factors Meis1, important for nervous system development and affecting dopamine signaling, and Btbd9, which sets the activity of medium spiny neurons in the striatum 3942.

GWA studies focusing on the quantitative aspects of normal sleep in the population rely on self-reported estimates of sleep and sleep problems through questionnaires or wrist actimetry devices or both. (Because of the large sample size required, classic EEG-based sleep phenotyping is time- and cost-prohibitive.) Variables such as sleep duration and timing, chronotype, sleep-onset latency, daytime sleepiness, or sleep problems (or a combination of these) have been assessed in an increasing number of studies by mining databases such as the UK Biobank 43 and 23andME combined or not with other large cohorts with aggregate sample sizes reaching more than 100,000 samples 4454. Many significant loci have been reported, some confirming results of other reports but all with quite small effect sizes (for example, explaining 3.5-minute differences in sleep duration) and without a coherent emerging picture explaining the large difference in sleep in the general population. The largest GWA analyses to date used more than 1.3 million individual samples to study insomnia complaints and found correlations able to explain 2.6% of the variance in insomnia 55. Many significant loci (202) were found pointing to about 1,000 genes that were identified through gene set enrichment analyses and available expression QTL (eQTL) and chromatin mapping data. Interestingly, overlap with RLS and RLS risk-conferring loci, including Meis1 and Btbd9, and striatal medium spiny neurons were reported, pointing to shared etiology or to pleiotropic gene effects not uncommon in sleep and neurological disorders 56. Although computational approaches and the use of datasets on intermediate phenotypes (intermediate between genome and phenotype such as transcriptome and chromatin data) strengthen the likelihood that the identified candidate genes are causally involved, the challenge remains to experimentally demonstrate their biological relevance for the sleep phenotypes they associate with.

GWA studies have been performed in animals as well. For example, in a large cohort of CFW outbred mice, in which more than 5.7 million variants segregate, 19 aspects of sleep were quantified by using a piezoelectric device 57 as part of a larger phenotyping pipeline 58. QTLs for sleep quality containing single genes identified Ppargc1a and Unc13c, both of which are involved in synaptic plasticity and transmission 59, 60.

Mapping of natural genetic variants underlying sleep traits in genetic reference populations

To better harness the genetic complexity contained in natural populations, mouse and fly GRPs have been created with a reduced but better-controlled and better-characterized genetic diversity. Fewer samples are needed to significantly associate sleep phenotypes to genotypes and to have the power to identify causal variants. In the fly, the Drosophila-GRP (DGRP), a set of about 200 recombinant inbred lines, was generated for the analyses of quantitative traits at a population level 61. A GWA study for sleep and activity phenotypes was performed with this GRP which yielded many single-nucleotide polymorphisms (SNPs) significantly associated with each trait, several of which mapped to genes over-represented in the epidermal growth factor receptor (Egfr) signaling pathway 62. Another GRP for Drosophila concerns a panel of 39 fully sequenced inbred lines selected for long and short sleep phenotypes: the Sleep Inbred Panel 63. In this panel, differences in sleep duration were associated with 126 polymorphisms pointing to 80 candidate genes related to the Egfr, Wnt, Hippo, and Mapk signaling pathways 64.

In the mouse, several GRPs have been derived from two or more isogenic strains such as the BXD 65, Collaborative Cross (CC) 66, Diversity Outbred (DO) 67, and Hybrid Mouse Diversity 68 panels. Some of these GRPs have been phenotyped for sleep. In the DO panel, moderate to high heritabilities for a number of sleep and circadian traits were observed by using a high-throughput phenotyping strategy 69. During construction of the CC panel, 50 sleep-related parameters and their response to sleep deprivation were assessed in about 600 “pre”-inbred mice (about F2:G5 or 75% inbred) carrying alleles from eight parental strains 70. For the resumption of activity after sleep deprivation, a significant and narrow QTL was identified encompassing two protein-coding genes ( Ntm and Snx19) involved in neural cell adhesion and insulin-containing vesicles maintenance, respectively. The BXD panel is perhaps the most widely used and best-characterized GRP. Earlier sleep analyses in the older BXD/ TyJ panel of recombinant inbred strains yielded Homer1, encoding a post-synaptic density scaffold protein involved in synaptic plasticity and calcium signaling, as a candidate gene for the increase in delta power, an EEG-derived proxy for sleep homeostatic drive, after sleep deprivation 71, 72. Additionally, retinoic acid receptor beta ( Rarb) was found to determine the contribution of slow waves to the NREM sleep EEG 73.

The QTLs identified in GRP panels can be quite large, encompassing many genes. Adding transcriptome data and other intermediate phenotypes such as proteome and metabolome data not only can help to select candidate genes located within QTLs but also can give insight into the flow of information from DNA to phenotype at the level of a population and how environmental challenges such as sleep deprivation alter this information flow. This multi-level, integrative approach has been termed systems genetics 74. Systems genetics approaches for sleep have been pioneered in the fly and mouse 75, 76. Sleep phenotyping and transcriptome analysis of four brain regions in a C57BL/6J × 129S1/SvImJ intercross identified gene expression networks underlying sleep regulation, such as an Arc-driven gene modules associated with EEG delta power 77. Systems genetics analyses have also been performed in the more recent BXD/ RwwJ GRP. In addition to the comprehensive sleep-wake phenome already mentioned above, cortical and liver transcriptome data and plasma metabolomic data were collected as intermediate phenotypes all assessed under undisturbed baseline conditions and after sleep deprivation 5. Sleep deprivation was found to extensively reshape the systems genetics landscape by altering 60 to 78% of the transcriptomes and the metabolome, and numerous genetic loci affected the magnitude and in some cases even the direction of change. Systems genetics analyses imply α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor trafficking and fatty acid turnover as substrates of the negative effects of insufficient sleep. Perhaps the most surprising finding concerned the implication of Acot11, an enzyme involved in fatty acid regulation in the liver, in determining the recovery of NREM sleep after sleep deprivation, demonstrating that peripheral systems can be crucial in explaining phenotypes that are generally considered to be controlled centrally. (“Sleep is of the brain, by the brain, and for the brain” 78.)

Furthermore, integrating gene expression data such as from the Genotype-Tissue Expression (GTEx) database 79 with GWA studies in humans facilitated the identification of the specific cell types implicated in insomnia (for example, medium spiny neurons 55), identification of eQTLs (for example, Cluap1 associated with the number of nocturnal sleep episodes 52), and feature construction for machine learning methods (for example, 4,374 eQTLs identified for morningness behavior like Nmur2, a Neuromedin receptor 80).

Novel insights into sleep using familial pedigrees

Studying familial pedigrees in which extreme phenotypes segregate can be highly informative in discovering rare mutations, likely to be missed in large-population GWA studies. This was powerfully illustrated with cases of the circadian sleep disorder familial advanced sleep phase syndrome (FASPS), which led to the discovery of causal mutations in the core clock genes Period 2 ( Per2) and Casein kinase I delta ( Csnk1d) 81, 82. Subsequently, a number of studies identified additional gene mutations affecting chronotype in humans, such as Timeless (Tim), Cryptochrome 2 (Cry2), and Period 3 ( Per3) (reviewed in 83). Many of the rare mutations identified had in common that they were of high penetrance and of large effect size which is a prerequisite for functional studies in model species such as fly and mouse, as illustrated in the next examples concerning sleep.

Besides the timing of sleep, which is generally considered a circadian phenotype (but see 84), a number of pedigrees in which a short sleep phenotype segregates have been reported. The first such study concerned the discovery of a mutation in Dec2 conferring short sleep in humans and mice 85. Besides its role in circadian timekeeping 86, Dec2 was found to act as a transcriptional repressor of Hypocretin ( Hcrt) encoding the two neuropeptides HCRT1 and 2, which are important in sleep-wake regulation 87. In another family with natural short sleepers, the causative mutation was found in the β1-adrenergic receptor ( Adr1), leading to decreased protein stability 88. Results in mice carrying the same mutation also showed a short sleep phenotype, albeit less pronounced compared with humans, and established an important role of β1-adrenergic receptors in sleep-wake regulation 88. A final example concerns a missense mutation in the G protein–coupled Neuropeptide S receptor 1 ( Npsr1), also found to be associated with a natural short sleep phenotype in humans 89. Mice expressing the homologous mutations slept less and interestingly were resistant to the memory deficits usually associated with sleep loss, thus suggesting that the NPS/NPSR1 pathway determines sleep duration and links sleep homeostasis and memory consolidation 89.

Discussion

In the above sections, we briefly outlined recent progress in the field of sleep genetics according to the different approaches used because a coherent picture on the core genes or gene pathways regulating specific aspects of sleep has yet to emerge. Sleep’s phenotypic complexity and the presumed redundancy of molecular pathways regulating specific aspects of sleep might be at the base of this. To some extent, such redundancy is also apparent at the neuro-anatomical level; a singular sleep center does not seem to exist that, when lesioned, eliminates sleep analogous to the effect of lesioning the SCN, which results in the elimination of circadian organization for most overt behaviors. To carry this analogy further, eliminating core clock genes (or combinations thereof) will render animals arrhythmic 90, while no genes for which null mutants preclude sleep are known. This might indicate that lacking sleep is embryonic lethal as long-term sleep deprivations are for adult rats and flies 91, 92 (but also see 93). In this respect, the finding of an all-but-complete abolishment of the EEG manifestation of REM sleep in mice lacking Chrm1 and 3 is both surprising and encouraging 28.

The field of sleep genetics might profit by focusing on core aspects of sleep, such as its homeostatic regulation 94, one of its defining features. Only a few genetic screens assessed the effects of sleep loss, even fewer have quantified the dynamics of the processes that accumulate during wakefulness and dissipate during sleep, and none systematically evaluated the negative impact of sleep loss on, for example, cognitive outcome variables. Quantifying the sleep homeostatic process is not trivial. When sleep deprivation is included as part of the phenotyping pipeline, usually a single sleep-deprivation duration is chosen, precluding the empirical quantification of the relationship between time spent awake and the resulting compensatory change in the sleep variable concerned. Instead, sleep variables quantified after sleep deprivation are contrasted to their levels during undisturbed baseline conditions, thereby implicitly assuming that these baseline levels represent what the subject “needs”. Whether baseline levels are indeed the levels that are homeostatically defended seems unlikely, especially under standard laboratory conditions (with little stimulation and challenge), animals might sleep more than required. Moreover, the widely used sleep homeostatic proxy 95—EEG delta power—is phenotypically and genetically complex and not all changes observed in this variable can be interpreted as proof of an altered underlying sleep homeostat 5, 94.

The concept of sleep homeostasis as a process in which a need for sleep accumulates during wakefulness and decreases during sleep is compatible with a feedback oscillation similar to that underlying circadian rhythms with the important distinction that the resulting oscillation is sleep-wake–driven in the case of the sleep homeostat whereas it is self-sustained in the case of the circadian clock. Although the role of clock genes in circadian rhythm generation is firmly established, their function goes well beyond what their name suggests. One example of this is the notion that they also fulfill a function in sleep homeostasis 96. Our recent study demonstrated that the cortical expression of the core clock genes Npas2 and Clock are sleep-wake–driven and not circadian 97. Moreover, the same study showed that a short 6-hour sleep deprivation caused a long-term (>48 hours) dampening of the circadian amplitude in the cortical expression of most other clock genes. Such results suggest that in peripheral and especially in extra-SCN brain tissues the clock gene circuitry may be used to track time spent awake and asleep rather than circadian time, which is compatible with the notion that clock genes integrate environmental and systemic cues such as temperature, corticosterone, light, and metabolic state, a number of which change according to sleep-wake state 96, 98. Therefore, the clock genes might already provide us a core set of sleep (homeostatic) genes that are preserved mostly from Drosophila to humans 96.

The approaches we presented can be broadly divided into two complementary approaches: one aimed at discovering naturally occurring allelic variants affecting sleep and the other at discovering genes by inducing mutations (randomly) on a single genetic background. The former seeks to explain the large phenotypic variability present in natural populations. Identifying the underlying genetic variants that have been maintained during natural selection, though challenging, might give insight into molecular pathways upon which evolutionary forces shape variation within and between species as well as insight into the variants determining disease risk. By disrupting single genes, the latter approach seeks to identify causal, large-effect genes to uncover specific molecular or physiological pathways. These genes may or may not vary in natural populations or be very rare. Thus, both approaches are needed to move the field forward.

We already presented an example from the circadian field in which an induced mutation of large effect was phenotypically rescued when expressed on a different genetic background 31. Similarly, the effect of major disease risk variants such as for the Brca1 and 2 genes in hereditary breast cancer is strongly affected by polygenic background ( https://doi.org/10.1101/19013086). Identifying the modifier genes that can offset the monogenic risk effects is obviously of importance in increasing the accuracy of risk assessment and deciding on therapeutic strategies. To systemically explore the effects of genetic background and to discover modifier genes affecting disease risk, established transgenic mouse models of Alzheimer disease have been crossed to a GRP, such as the BXD recombinant inbred panel, to create a resource for experimental precision medicine 99. Such combined approaches could also be taken for major-effect genes identified in mutagenesis or knockout screens for sleep to study the epistatic interactions and charting the molecular pathways involved.

Though not the focus of this review, studying intermediate phenotypes (that is, *omics) can aid in identifying genes implicated in sleep and its regulation, especially when combined with genomic information 5, 76, 77. For example, sleep-wake–driven changes in transcriptome, metabolome, or phosphoproteome studies led to molecular pathways for sleep homeostasis and key genes therein such as Homer1, Acot11, and Sik3 5, 17, 72, 100. Given the enormous number of “genome-wide” data sets that have been accumulated, methods to mine and integrate them become increasingly important. Supervised and unsupervised machine learning methods (for example, 101103) are being actively developed to better integrate multi-omics datasets and infer causality, and their implementations in the sleep field are ongoing. Finally, the information contained in the DNA alone is insufficient to explain phenotypic variability, and the importance of the contribution of activity or accessibility of non-coding regulatory elements (for example, enhancers, silencers, and promotors) is increasingly acknowledged in the sleep field 104, 105 as these were shown to be sensitive to sleep loss with surprisingly fast dynamics 97, 106.

Abbreviations

BXD, panel of recombinant inbred mouse lines derived from a C57BL/6J (B) x DBA/2J (D) intercross; CC, Collaborative Cross; CFW, outbred population of mice derived from Swiss Webster (W) at Carworth Farms (CF); CRISPR-Cas9, clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9; DGRP, Drosophila genetic reference population; EEG, electroencephalogram; EMS, the mutagen ethyl methanesulfonate; ENU, N-ethyl-N-nitrosourea; eQTL, expression quantitative trait locus; GRP, genetic reference population; GTEx, Genotype-Tissue Expression; GWA, genome-wide association; IKMC, International Knockout Mouse Consortium; IMPC, International Mouse Phenotyping Consortium; NREM, non–rapid eye movement; QTL, quantitative trait locus; REM, rapid eye movement; RLS, restless legs syndrome; SCN, suprachiasmatic nuclei; SNP, single-nucleotide polymorphism; UAS, upstream activation sequence

Editorial Note on the Review Process

F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty and are edited as a service to readers. In order to make these reviews as comprehensive and accessible as possible, the referees provide input before publication and only the final, revised version is published. The referees who approved the final version are listed with their names and affiliations but without their reports on earlier versions (any comments will already have been addressed in the published version).

The referees who approved this article are:

  • Allan I. Pack, Center for Sleep and Circadian Neurobiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA

  • Ying-Hui Fu, Department of Neurology, University of California San Francisco, San Francisco, CA, USA

  • Alex C Keene, Jupiter Life Science Initiative, , , 33458, USA, Florida Atlantic University, Jupiter, FL, USA

Funding Statement

Funding supporting MJ and PF was obtained from the Swiss National Science Foundation (grants CRSII3_13620, 31003A_146694, and 31003A_173182) and the state of Vaud, Switzerland. Funding supporting BFO was obtained from the National Institutes of Health (grants R43NS83218, R44NS083218, and R41HL142489).

The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

[version 1; peer review: 3 approved]

References

  • 1. Konopka RJ, Benzer S: Clock mutants of Drosophila melanogaster. Proc Natl Acad Sci U S A. 1971;68(9):2112–6. 10.1073/pnas.68.9.2112 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 2. Vitaterna MH, King DP, Chang AM, et al. : Mutagenesis and mapping of a mouse gene, Clock, essential for circadian behavior. Science. 1994;264(5159):719–25. 10.1126/science.8171325 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Allada R, Emery P, Takahashi JS, et al. : Stopping time: The genetics of fly and mouse circadian clocks. Annu Rev Neurosci. 2001;24:1091–119. 10.1146/annurev.neuro.24.1.1091 [DOI] [PubMed] [Google Scholar]
  • 4. Leung LC, Wang GX, Madelaine R, et al. : Neural signatures of sleep in zebrafish. Nature. 2019;571(7764):198–204. 10.1038/s41586-019-1336-7 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 5. Diessler S, Jan M, Emmenegger Y, et al. : A systems genetics resource and analysis of sleep regulation in the mouse. PLoS Biol. 2018;16(8):e2005750. 10.1371/journal.pbio.2005750 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Keene AC, Duboue ER: The origins and evolution of sleep. J Exp Biol. 2018;221(Pt 11):pii: jeb159533. 10.1242/jeb.159533 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 7. Tyree SM, de Lecea L: Optogenetic Investigation of Arousal Circuits. Int J Mol Sci. 2017;18(8): pii: E1773. 10.3390/ijms18081773 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Deisseroth K: Optogenetics: 10 years of microbial opsins in neuroscience. Nat Neurosci. 2015;18(9):1213–25. 10.1038/nn.4091 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Bauer DC, McMorran BJ, Foote SJ, et al. : Genome-wide analysis of chemically induced mutations in mouse in phenotype-driven screens. BMC Genomics. 2015;16:866. 10.1186/s12864-015-2073-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Toda H, Williams JA, Gulledge M, et al. : A sleep-inducing gene, nemuri, links sleep and immune function in Drosophila. Science. 2019;363(6426):509–15. 10.1126/science.aat1650 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 11. Chiu CN, Rihel J, Lee DA, et al. : A Zebrafish Genetic Screen Identifies Neuromedin U as a Regulator of Sleep/Wake States. Neuron. 2016;89(4):842–56. 10.1016/j.neuron.2016.01.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Cirelli C, Bushey D, Hill S, et al. : Reduced sleep in Drosophila Shaker mutants. Nature. 2005;434(7037):1087–92. 10.1038/nature03486 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 13. Koh K, Joiner WJ, Wu MN, et al. : Identification of SLEEPLESS, a sleep-promoting factor. Science. 2008;321(5887):372–6. 10.1126/science.1155942 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Wu MN, Joiner WJ, Dean T, et al. : SLEEPLESS, a Ly-6/neurotoxin family member, regulates the levels, localization and activity of Shaker. Nat Neurosci. 2010;13(1):69–75. 10.1038/nn.2454 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 15. Funato H, Miyoshi C, Fujiyama T, et al. : Forward-genetics analysis of sleep in randomly mutagenized mice. Nature. 2016;539(7629):378–83. 10.1038/nature20142 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 16. Honda T, Fujiyama T, Miyoshi C, et al. : A single phosphorylation site of SIK3 regulates daily sleep amounts and sleep need in mice. Proc Natl Acad Sci U S A. 2018;115(41):10458–63. 10.1073/pnas.1810823115 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 17. Wang Z, Ma J, Miyoshi C, et al. : Quantitative phosphoproteomic analysis of the molecular substrates of sleep need. Nature. 2018;558(7710):435–9. 10.1038/s41586-018-0218-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Brüning F, Noya SB, Bange T, et al. : Sleep-wake cycles drive daily dynamics of synaptic phosphorylation. Science. 2019;366(6462):pii: eaav3617. 10.1126/science.aav3617 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 19. Miyoshi C, Kim SJ, Ezaki T, et al. : Methodology and theoretical basis of forward genetic screening for sleep/wakefulness in mice. Proc Natl Acad Sci U S A. 2019;116(32):16062–7. 10.1073/pnas.1906774116 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 20. Trojanowski NF, Raizen DM: Call it Worm Sleep. Trends Neurosci. 2016;39(2):54–62. 10.1016/j.tins.2015.12.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Nelson MD, Lee KH, Churgin MA, et al. : FMRFamide-like FLP-13 neuropeptides promote quiescence following heat stress in Caenorhabditis elegans. Curr Biol. 2014;24(20):2406–10. 10.1016/j.cub.2014.08.037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Yuan J, Zhou J, Raizen DM, et al. : High-throughput, motility-based sorter for microswimmers such as C. elegans. Lab Chip. 2015;15(13):2790–8. 10.1039/c5lc00305a [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Iannacone MJ, Beets I, Lopes LE, et al. : The RFamide receptor DMSR-1 regulates stress-induced sleep in C. elegans. Elife. 2017;6: pii: e19837. 10.7554/eLife.19837 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 24. Huang H, Zhu CT, Skuja LL, et al. : Genome-Wide Screen for Genes Involved in Caenorhabditis elegans Developmentally Timed Sleep. G3 (Bethesda). 2017;7(9):2907–17. 10.1534/g3.117.300071 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 25. Singh C, Rihel J, Prober DA: Neuropeptide Y Regulates Sleep by Modulating Noradrenergic Signaling. Curr Biol. 2017;27(24):3796–3811.e5. 10.1016/j.cub.2017.11.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Skarnes WC, Rosen B, West AP, et al. : A conditional knockout resource for the genome-wide study of mouse gene function. Nature. 2011;474(7351):337–42. 10.1038/nature10163 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 27. Yu JSL, Yusa K: Genome-wide CRISPR-Cas9 screening in mammalian cells. Methods. 2019;164–165:29–35. 10.1016/j.ymeth.2019.04.015 [DOI] [PubMed] [Google Scholar]
  • 28. Niwa Y, Kanda GN, Yamada RG, et al. : Muscarinic Acetylcholine Receptors Chrm1 and Chrm3 Are Essential for REM Sleep. Cell Rep. 2018;24(9):2231–2247.e7. 10.1016/j.celrep.2018.07.082 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 29. Sunagawa GA, Sumiyama K, Ukai-Tadenuma M, et al. : Mammalian Reverse Genetics without Crossing Reveals Nr3a as a Short-Sleeper Gene. Cell Rep. 2016;14(3):662–77. 10.1016/j.celrep.2015.12.052 [DOI] [PubMed] [Google Scholar]
  • 30. Tatsuki F, Sunagawa GA, Shi S, et al. : Involvement of Ca(2+)-Dependent Hyperpolarization in Sleep Duration in Mammals. Neuron. 2016;90(1):70–85. 10.1016/j.neuron.2016.02.032 [DOI] [PubMed] [Google Scholar]
  • 31. Shimomura K, Lowrey PL, Vitaterna MH, et al. : Genetic suppression of the circadian Clock mutation by the melatonin biosynthesis pathway. Proc Natl Acad Sci U S A. 2010;107(18):8399–403. 10.1073/pnas.1004368107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Shimomura K, Low-Zeddies SS, King DP, et al. : Genome-wide epistatic interaction analysis reveals complex genetic determinants of circadian behavior in mice. Genome Res. 2001;11(6):959–80. 10.1101/gr.171601 [DOI] [PubMed] [Google Scholar]
  • 33. Rusterholz T, Hamann C, Markovic A, et al. : Nature and Nurture: Brain Region-Specific Inheritance of Sleep Neurophysiology in Adolescence. J Neurosci. 2018;38(43):9275–9285. 10.1523/JNEUROSCI.0945-18.2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Andretic R, Franken P, Tafti M: Genetics of Sleep. Annu Rev Genet. 2008;42:361–88. 10.1146/annurev.genet.42.110807.091541 [DOI] [PubMed] [Google Scholar]
  • 35. Sherry ST, Ward MH, Kholodov M, et al. : dbSNP: The NCBI database of genetic variation. Nucleic Acids Res. 2001;29(1):308–11. 10.1093/nar/29.1.308 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Ritchie MD, van Steen K: The search for gene-gene interactions in genome-wide association studies: Challenges in abundance of methods, practical considerations, and biological interpretation. Ann Transl Med. 2018;6(8):157. 10.21037/atm.2018.04.05 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Miyagawa T, Kawashima M, Nishida N, et al. : Variant between CPT1B and CHKB associated with susceptibility to narcolepsy. Nat Genet. 2008;40(11):1324–8. 10.1038/ng.231 [DOI] [PubMed] [Google Scholar]
  • 38. Winkelmann J, Schormair B, Lichtner P, et al. : Genome-wide association study of restless legs syndrome identifies common variants in three genomic regions. Nat Genet. 2007;39(8):1000–6. 10.1038/ng2099 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 39. Salminen AV, Lam DD, Winkelmann J: Role of MEIS1 in restless legs syndrome: From GWAS to functional studies in mice. Adv Pharmacol. 2019;84:175–84. 10.1016/bs.apha.2019.03.003 [DOI] [PubMed] [Google Scholar]
  • 40. Allen RP, Donelson NC, Jones BC, et al. : Animal models of RLS phenotypes. Sleep Med. 2017;31:23–8. 10.1016/j.sleep.2016.08.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Lyu S, Xing H, DeAndrade MP, et al. : The Role of BTBD9 in Striatum and Restless Legs Syndrome. eNeuro. 2019;6(5): pii: ENEURO.0277-19.2019. 10.1523/ENEURO.0277-19.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 42. Catoire H, Sarayloo F, Mourabit Amari K, et al. : A direct interaction between two Restless Legs Syndrome predisposing genes: MEIS1 and SKOR1. Sci Rep. 2018;8(1):12173. 10.1038/s41598-018-30665-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Bycroft C, Freeman C, Petkova D, et al. : The UK Biobank resource with deep phenotyping and genomic data. Nature. 2018;562(7726):203–9. 10.1038/s41586-018-0579-z [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 44. Jones SE, Tyrrell J, Wood AR, et al. : Genome-Wide Association Analyses in 128,266 Individuals Identifies New Morningness and Sleep Duration Loci. PLoS Genet. 2016;12(8):e1006125. 10.1371/journal.pgen.1006125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Lane JM, Liang J, Vlasac I, et al. : Genome-wide association analyses of sleep disturbance traits identify new loci and highlight shared genetics with neuropsychiatric and metabolic traits. Nat Genet. 2017;49(2):274–81. 10.1038/ng.3749 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Kalmbach DA, Schneider LD, Cheung J, et al. : Genetic Basis of Chronotype in Humans: Insights From Three Landmark GWAS. Sleep. 2017;40(2):429. 10.1093/sleep/zsw048 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Doherty A, Smith-Byrne K, Ferreira T, et al. : GWAS identifies 14 loci for device-measured physical activity and sleep duration. Nat Commun. 2018;9(1):5257. 10.1038/s41467-018-07743-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Dashti HS, Jones SE, Wood AR, et al. : Genome-wide association study identifies genetic loci for self-reported habitual sleep duration supported by accelerometer-derived estimates. Nat Commun. 2019;10(1):1100. 10.1038/s41467-019-08917-4 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 49. Rhodes JA, Lane JM, Vlasac IM, et al. : Association of DAT1 genetic variants with habitual sleep duration in the UK Biobank. Sleep. 2019;42(1). 10.1093/sleep/zsy193 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 50. Jones SE, Lane JM, Wood AR, et al. : Genome-wide association analyses of chronotype in 697,828 individuals provides insights into circadian rhythms. Nat Commun. 2019;10(1):343. 10.1038/s41467-018-08259-7 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 51. Wang H, Lane JM, Jones SE, et al. : Genome-wide association analysis of self-reported daytime sleepiness identifies 42 loci that suggest biological subtypes. Nat Commun. 2019;10(1):3503. 10.1038/s41467-019-11456-7 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 52. Jones SE, van Hees VT, Mazzotti DR, et al. : Genetic studies of accelerometer-based sleep measures yield new insights into human sleep behaviour. Nat Commun. 2019;10(1):1585. 10.1038/s41467-019-09576-1 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 53. Lane JM, Vlasac I, Anderson SG, et al. : Genome-wide association analysis identifies novel loci for chronotype in 100,420 individuals from the UK Biobank. Nat Commun. 2016;7:10889. 10.1038/ncomms10889 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Noordam R, Bos MM, Wang H, et al. : Multi-ancestry sleep-by-SNP interaction analysis in 126,926 individuals reveals lipid loci stratified by sleep duration. Nat Commun. 2019;10(1):5121. 10.1038/s41467-019-12958-0 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 55. Jansen PR, Watanabe K, Stringer S, et al. : Genome-wide analysis of insomnia in 1,331,010 individuals identifies new risk loci and functional pathways. Nat Genet. 2019;51(3):394–403. 10.1038/s41588-018-0333-3 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 56. Veatch OJ, Keenan BT, Gehrman PR, et al. : Pleiotropic genetic effects influencing sleep and neurological disorders. Lancet Neurol. 2017;16(2):158–70. 10.1016/S1474-4422(16)30339-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Mang GM, Nicod J, Emmenegger Y, et al. : Evaluation of a piezoelectric system as an alternative to electroencephalogram/ electromyogram recordings in mouse sleep studies. Sleep. 2014;37(8):1383–92. 10.5665/sleep.3936 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Nicod J, Davies RW, Cai N, et al. : Genome-wide association of multiple complex traits in outbred mice by ultra-low-coverage sequencing. Nat Genet. 2016;48(8):912–8. 10.1038/ng.3595 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Chen Z, Cooper B, Kalla S, et al. : The Munc13 proteins differentially regulate readily releasable pool dynamics and calcium-dependent recovery at a central synapse. J Neurosci. 2013;33(119):8336–51. 10.1523/JNEUROSCI.5128-12.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Dougherty SE, Bartley AF, Lucas EK, et al. : Mice lacking the transcriptional coactivator PGC-1α exhibit alterations in inhibitory synaptic transmission in the motor cortex. Neuroscience. 2014;271:137–48. 10.1016/j.neuroscience.2014.04.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Mackay TFC, Richards S, Stone EA, et al. : The Drosophila melanogaster Genetic Reference Panel. Nature. 2012;482(7384):173–8. 10.1038/nature10811 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 62. Harbison ST, McCoy LJ, Mackay TFC: Genome-wide association study of sleep in Drosophila melanogaster. BMC Genomics. 2013;14:281. 10.1186/1471-2164-14-281 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Serrano Negron YL, Hansen NF, Harbison ST: The Sleep Inbred Panel, a Collection of Inbred Drosophila melanogaster with Extreme Long and Short Sleep Duration. G3 (Bethesda). 2018;8(9):2865–73. 10.1534/g3.118.200503 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Harbison ST, Serrano Negron YL, Hansen NF, et al. : Selection for long and short sleep duration in Drosophila melanogaster reveals the complex genetic network underlying natural variation in sleep. PLoS Genet. 2017;13(12):e1007098. 10.1371/journal.pgen.1007098 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 65. Peirce JL, Lu L, Gu J, et al. : A new set of BXD recombinant inbred lines from advanced intercross populations in mice. BMC Genet. 2004;5:7. 10.1186/1471-2156-5-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Srivastava A, Morgan AP, Najarian ML, et al. : Genomes of the Mouse Collaborative Cross. Genetics. 2017;206(2):537–56. 10.1534/genetics.116.198838 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Churchill GA, Gatti DM, Munger SC, et al. : The Diversity Outbred mouse population. Mamm Genome. 2012;23(9–10):713–8. 10.1007/s00335-012-9414-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Lusis AJ, Seldin MM, Allayee H, et al. : The Hybrid Mouse Diversity Panel: A resource for systems genetics analyses of metabolic and cardiovascular traits. J Lipid Res:. 2016;57(6):925–42. 10.1194/jlr.R066944 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Keenan BT, Galante RJ, Lian J, et al. : High-throughput sleep phenotyping produces robust and heritable traits in Diversity Outbred mice and their founder strains. Sleep. 2020. pii: zsz278. 10.1093/sleep/zsz278 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 70. Philip VM, Sokoloff G, Ackert-Bicknell CL, et al. : Genetic analysis in the Collaborative Cross breeding population. Genome Res. 2011;21(8):1223–38. 10.1101/gr.113886.110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Mackiewicz M, Paigen B, Naidoo N, et al. : Analysis of the QTL for sleep homeostasis in mice: Homer1a is a likely candidate. Physiol Genomics. 2008;33(1):91–9. 10.1152/physiolgenomics.00189.2007 [DOI] [PubMed] [Google Scholar]
  • 72. Maret S, Dorsaz S, Gurcel L, et al. : Homer1a is a core brain molecular correlate of sleep loss. Proc Natl Acad Sci U S A. 2007;104(50):20090–5. 10.1073/pnas.0710131104 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 73. Maret S, Franken P, Dauvilliers Y, et al. : Retinoic acid signaling affects cortical synchrony during sleep. Science. 2005;310(5745):111–3. 10.1126/science.1117623 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 74. Civelek M, Lusis AJ: Systems genetics approaches to understand complex traits. Nat Rev Genet. 2014;15(1):34–48. 10.1038/nrg3575 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Harbison ST, Carbone MA, Ayroles JF, et al. : Co-regulated transcriptional networks contribute to natural genetic variation in Drosophila sleep. Nat Genet. 2009;41(3):371–5. 10.1038/ng.330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Jiang P, Scarpa JR, Fitzpatrick K, et al. : A systems approach identifies networks and genes linking sleep and stress: implications for neuropsychiatric disorders. Cell Rep. 2015;11(5):835–48. 10.1016/j.celrep.2015.04.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Scarpa JR, Jiang P, Gao VD, et al. : Cross-species systems analysis identifies gene networks differentially altered by sleep loss and depression. Sci Adv. 2018;4(7):eaat1294. 10.1126/sciadv.aat1294 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 78. Hobson JA: Sleep is of the brain, by the brain and for the brain. Nature. 2005;437(7063):1254–6. 10.1038/nature04283 [DOI] [PubMed] [Google Scholar]
  • 79. Carithers LJ, Ardlie K, Barcus M, et al. : A Novel Approach to High-Quality Postmortem Tissue Procurement: The GTEx Project. Biopreserv Biobank. 2015;13(5):311–9. 10.1089/bio.2015.0032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Li J, Huang T: Predicting and analyzing early wake-up associated gene expressions by integrating GWAS and eQTL studies. Biochim Biophys Acta Mol Basis Dis. 2018;1864(6 Pt B):2241–6. 10.1016/j.bbadis.2017.10.036 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 81. Toh KL, Jones CR, He Y, et al. : An h Per2 phosphorylation site mutation in familial advanced sleep phase syndrome. Science. 2001;291(5506):1040–3. 10.1126/science.1057499 [DOI] [PubMed] [Google Scholar]
  • 82. Xu Y, Padiath QS, Shapiro RE, et al. : Functional consequences of a CKIdelta mutation causing familial advanced sleep phase syndrome. Nature. 2005;434(7033):640–4. 10.1038/nature03453 [DOI] [PubMed] [Google Scholar]
  • 83. Ashbrook LH, Krystal AD, Fu YH, et al. : Genetics of the human circadian clock and sleep homeostat. Neuropsychopharmacology. 2020;45(1):45–54. 10.1038/s41386-019-0476-7 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 84. Mongrain V, Carrier J, Dumont M: Circadian and homeostatic sleep regulation in morningness-eveningness. J Sleep Res. 2006;15(2):162–6. 10.1111/j.1365-2869.2006.00532.x [DOI] [PubMed] [Google Scholar]
  • 85. He Y, Jones CR, Fujiki N, et al. : The transcriptional repressor DEC2 regulates sleep length in mammals. Science. 2009;325(5942):866–70. 10.1126/science.1174443 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 86. Honma S, Kawamoto T, Takagi Y, et al. : Dec1 and Dec2 are regulators of the mammalian molecular clock. Nature. 2002;419(6909):841–4. 10.1038/nature01123 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 87. Hirano A, Hsu PK, Zhang L, et al. : DEC2 modulates orexin expression and regulates sleep. Proc Natl Acad Sci U S A. 2018;115(13):3434–9. 10.1073/pnas.1801693115 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 88. Shi G, Xing L, Wu D, et al. : A Rare Mutation of β 1-Adrenergic Receptor Affects Sleep/Wake Behaviors. Neuron. 2019;103(6):1044–1055.e7. 10.1016/j.neuron.2019.07.026 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 89. Xing L, Shi G, Mostovoy Y, et al. : Mutant neuropeptide S receptor reduces sleep duration with preserved memory consolidation. Sci Transl Med. 2019;11(514): pii: eaax2014. 10.1126/scitranslmed.aax2014 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 90. Cox KH, Takahashi JS: Circadian clock genes and the transcriptional architecture of the clock mechanism. J Mol Endocrinol. 2019;63(4):R93–R102. 10.1530/JME-19-0153 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 91. Shaw PJ, Tononi G, Greenspan RJ, et al. : Stress response genes protect against lethal effects of sleep deprivation in Drosophila. Nature. 2002;417(6886):287–91. 10.1038/417287a [DOI] [PubMed] [Google Scholar]
  • 92. Rechtschaffen A, Bergmann BM, Everson CA, et al. : Sleep deprivation in the rat: X. Integration and discussion of the findings. 1989. Sleep. 2002;25(1):68–87. 10.1093/sleep/25.1.68 [DOI] [PubMed] [Google Scholar]
  • 93. Geissmann Q, Beckwith EJ, Gilestro GF: Most sleep does not serve a vital function: Evidence from Drosophila melanogaster. Sci Adv. 2019;5(2):eaau9253. 10.1126/sciadv.aau9253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Mang GM, Franken P: Genetic dissection of sleep homeostasis. Curr Top Behav Neurosci. 2015;25:25–63. 10.1007/7854_2013_270 [DOI] [PubMed] [Google Scholar]
  • 95. Daan S, Beersma DG, Borbély AA: Timing of human sleep: Recovery process gated by a circadian pacemaker. Am J Physiol. 1984;246(2 Pt 2):R161-83. 10.1152/ajpregu.1984.246.2.R161 [DOI] [PubMed] [Google Scholar]
  • 96. Franken P: A role for clock genes in sleep homeostasis. Curr Opin Neurobiol. 2013;23(5):864–72. 10.1016/j.conb.2013.05.002 [DOI] [PubMed] [Google Scholar]
  • 97. Hor CN, Yeung J, Jan M, et al. : 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. 2019;116(51):25773–83. 10.1073/pnas.1910590116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Schibler U, Gotic I, Saini C, et al. : Clock-Talk: Interactions between Central and Peripheral Circadian Oscillators in Mammals. Cold Spring Harb Symp Quant Biol. 2015;80:223–32. 10.1101/sqb.2015.80.027490 [DOI] [PubMed] [Google Scholar]
  • 99. Neuner SM, Heuer SE, Huentelman MJ, et al. : Harnessing Genetic Complexity to Enhance Translatability of Alzheimer’s Disease Mouse Models: A Path toward Precision Medicine. Neuron. 2019;101(3):399-411.e5. 10.1016/j.neuron.2018.11.040 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 100. Martin SC, Monroe SK, Diering GH: Homer1a and mGluR1/5 Signaling in Homeostatic Sleep Drive and Output. Yale J Biol Med. 2019;92(1):93–101. [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 101. Argelaguet R, Clark SJ, Mohammed H, et al. : Multi-omics profiling of mouse gastrulation at single-cell resolution. Nature. 2019;576(7787):487–91. 10.1038/s41586-019-1825-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Argelaguet R, Velten B, Arnol D, et al. : Multi-Omics Factor Analysis-a framework for unsupervised integration of multi-omics data sets. Mol Syst Biol. 2018;14(6):e8124. 10.15252/msb.20178124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Singh A, Shannon CP, Gautier B, et al. : DIABLO: an integrative approach for identifying key molecular drivers from multi-omics assays. Bioinformatics. 2019;35(17):3055–62. 10.1093/bioinformatics/bty1054 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 104. Gaine ME, Chatterjee S, Abel T: Sleep Deprivation and the Epigenome. Front Neural Circuits. 2018;12:14. 10.3389/fncir.2018.00014 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 105. O'Callaghan EK, Green EW, Franken P, et al. : Omics Approaches in Sleep-Wake Regulation. Handb Exp Pharmacol. 2019;253:59–81. 10.1007/164_2018_125 [DOI] [PubMed] [Google Scholar]
  • 106. Massart R, Freyburger M, Suderman M, et al. : The genome-wide landscape of DNA methylation and hydroxymethylation in response to sleep deprivation impacts on synaptic plasticity genes. Transl Psychiatry. 2014;4:e347. 10.1038/tp.2013.120 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from F1000Research are provided here courtesy of F1000 Research Ltd

RESOURCES