Zebrafish sleep: from geneZZZ to neuronZZZ

https://doi.org/10.1016/j.conb.2017.02.009Get rights and content

Highlights

  • Zebrafish have advanced genetic and functional imaging toolkits to study sleep.

  • Zebrafish have conserved sleep genes and neurons.

  • Recent zebrafish studies have identified novel regulators of sleep and arousal.

  • Behavioral drug screens can identify links between disease and sleep disruption.

All animals have a fundamental and unavoidable requirement for rest, yet we still do not fully understand the processes that initiate, maintain, and regulate sleep. The larval zebrafish is an optically translucent, genetically tractable model organism that exhibits sleep states regulated by conserved sleep circuits, thereby offering a unique system for investigating the genetic and neural control of sleep. Recent studies using high throughput monitoring of larval sleep/wake behaviour have unearthed novel modulators involved in regulating arousal and have provided new mechanistic insights into the role of established sleep/wake modulators. In addition, the application of computational tools to large behavioural datasets has allowed for the identification of neuroactive compounds that alleviate sleep symptoms associated with genetic neurological disorders.

Introduction

Work over the past 15 years has demonstrated that sleep is evolutionarily conserved across the animal kingdom, indicating that sleep serves an essential, possibly universal, function [1]. Moreover, the negative impact that sleep disruption has on immune, metabolic, cardiac and cognitive health demonstrates sleep’s critical role in optimising daily behaviour and general physiological well-being. Many molecular and neuronal control systems are in place to regulate sleep’s timing and duration; however, we are still discovering these systems and their rules. Two major discoveries have spearheaded advances in the field. First, the discovery that the human sleep disease, narcolepsy, is a dysfunction in hypocretin/orexin signalling demonstrated that small populations of peptidergic neurons can have profound consequences on human and animal sleep [2]. Second, the recognition that genetically tractable non-mammalian species exhibit sleep states has expanded the models available to sleep researchers and have facilitated screens for sleep mutants [3].

The study of sleep regulation in zebrafish larvae has taken up a unique model system niche, as they offer a sophisticated genetic toolkit coupled with the ability to monitor and manipulate the activity of conserved sleep/wake neurons in vivo (see Box 1). In this review, we discuss recent insights into sleep in this diurnal vertebrate and highlight novel methods that use the special properties of the zebrafish model.

Section snippets

Monitoring sleep and arousal in larval zebrafish

To enable relatively high-throughput monitoring of sleep/wake states in zebrafish larvae, the animals are tracked by videography in a 96-well plate format for several days on either a 24-hour light–dark cycle or on constant illumination (Figure 1a). Larval and adult zebrafish are diurnal and at night exhibit an increased number and duration of inactive bouts (Figure 1b), which have a typical duration around three minutes but can last for hours [4]. Quiescent bouts lasting at least one minute

Neurocircuitry of sleep in zebrafish

Classical and modern lesion studies in mammals highlighted numerous brain areas, including the basal forebrain, posterior hypothalamus, reticular formation, and hindbrain as critical for the regulation of arousal and sleep [16, 17, 18]. More recently, systematic searches for sleep/wake regulatory neurons using chemogenetic and optogenetic modulation of putative sleep/wake-inducing neurons are more precisely defining many key subpopulations as well as unravelling the complex dynamics at play

Sleep, disease, and predictive pharmacology

Many neuropsychiatric and neurodevelopmental disorders, including depression, schizophrenia, Alzheimer’s disease, and autism, are associated with sleep disturbances [44, 45], but it remains unclear in most cases to what extent sleep disruption contributes to disease severity and progression. Several studies have now identified sleep and arousal endophenotypes in zebrafish genetic models of disease. For example, a zebrafish model of the most common cause of mental retardation, Fragile-X

Concluding remarks

Studies into the genetic and neural components involved in regulating evolutionarily conserved behaviours such as sleep have profited from advances in zebrafish genetic and imaging tools. Validation of conserved network modules involved in regulating sleep and wake in zebrafish has paved the way for genetic and neuropeptide screens that have identified novel modulators of vertebrate sleep that are likely to be relevant in mammals. Moving forward, studies that combine in vivo neuronal imaging

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Conflict of interest

None.

Acknowledgements

We thank all the members of the Rihel lab for helpful comments on the manuscript. This work is supported by an ERC Starting Grant, a UCL Excellence Fellowship, and a Grand Challenges Grant awarded to JR.

References (53)

  • Z. Ben-Moshe Livne et al.

    Genetically blocking the zebrafish pineal clock affects circadian behavior

    PLoS Genet.

    (2016)
  • A. Chen et al.

    QRFP and its receptors regulate locomotor activity and sleep in zebrafish

    J. Neurosci.

    (2016)
  • H. Baribault et al.

    The G-protein-coupled receptor GPR103 regulates bone formation

    Mol. Cell. Biol.

    (2006)
  • J. Rihel et al.

    Sites of action of sleep and wake drugs: insights from model organisms

    Curr. Opin. Neurobiol.

    (2013)
  • L. Lin et al.

    The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor 2 gene

    Cell

    (1999)
  • J. Rihel et al.

    Monitoring sleep and arousal in zebrafish

    Methods Cell Biol.

    (2010)
  • C. Singh et al.

    Norepinephrine is required to promote wakefulness and for hypocretin-induced arousal in zebrafish

    Elife

    (2015)
  • M. Shein-Idelson et al.

    Slow waves, sharp waves, ripples, and REM in sleeping dragons

    Science

    (2016)
  • B. van Alphen et al.

    A dynamic deep sleep stage in Drosophila

    J. Neurosci.

    (2013)
  • K.E. Severi et al.

    Neural control and modulation of swimming speed in the larval zebrafish

    Neuron

    (2014)
  • I.G. Woods et al.

    Neuropeptidergic signaling partitions arousal behaviors in zebrafish

    J. Neurosci.

    (2014)
  • T.W. Dunn et al.

    Brain-wide mapping of neural activity controlling zebrafish exploratory locomotion

    Elife

    (2016)
  • M.B. Ahrens et al.

    Brain-wide neuronal dynamics during motor adaptation in zebrafish

    Nature

    (2012)
  • E.A. Naumann et al.

    From whole-brain data to functional circuit models: the zebrafish optomotor response

    Cell

    (2016)
  • C. von Economo

    Sleep as a problem of localization

    J. Nerv. Ment. Dis.

    (1930)
  • G. Moruzzi et al.

    Brain stem reticular formation and activation of the EEG

    Electroencephalogr. Clin. Neurophysiol.

    (1949)
  • Cited by (27)

    • Sleep circuits and physiology in non-mammalian systems

      2020, Current Opinion in Physiology
      Citation Excerpt :

      At least three sleep-homeostasis regulating neuronal populations have been recently discovered in the zebrafish: Galanin-expressing neurons [26••], neurons that produce the RF-amide Neuropeptide VF (NPVF) [47], and the serotonergic neurons of the raphe [48••]. The full extent to which these populations interact with zebrafish wake-promoting circuits of Hypocretin (Hcrt) [49], norepinephrine neurons of the locus coeruleus [50], the arousing neuropeptide Neuromedin U (Nmu) [51], and Insulin-like Growth Factor [52], or with other sleep-promoting signals, such as the circadian sleep-output signal melatonin [53], the locus coeruleus-inhibiting Neuropeptide Y [54], or even each other is not yet clear and has been reviewed elsewhere [55]. Additionally, the discovery of sleep regulatory circuits in mammals continues in tandem with elucidation of their conservation in zebrafish.

    • Modeling autism spectrum disorders in zebrafish

      2020, Behavioral and Neural Genetics of Zebrafish
    • NREM Sleep Regulation From Neuronal Assembly to Ion

      2019, Handbook of Behavioral Neuroscience
      Citation Excerpt :

      In both cases, EEG recordings show that one hemisphere is desynchronized, while the other displays a synchronized state indicative of SWS (Lyamin, Pryaslova, Lance, & Siegel, 2005; Mascetti, 2016; Rattenborg et al., 2016). Finally, as developed in several chapters of this volume, flies, fish, and worms are also very powerful models to study the genetics of sleep (Artiushin & Sehgal, 2017; Barlow & Rihel, 2017; Trojanowski & Raizen, 2016). A conceptual model for the timing of sleep onset was proposed in the 1980s (Borbely, 1982; Daan, Beersma, & Borbely, 1984) and has largely been confirmed as the most prevalent model today (Borbely, Daan, Wirz-Justice, & Deboer, 2016).

    • Pitpnc1a Regulates Zebrafish Sleep and Wake Behavior through Modulation of Insulin-like Growth Factor Signaling

      2018, Cell Reports
      Citation Excerpt :

      Expression analysis of dorsal forebrain (egr3, tbr1a, and eomesa) and hypothalamic (npvf) markers found no changes in pitpnc1a−/− larvae, suggesting that Pitpnc1a is not required for the specification or differentiation of these areas that strongly express pitpnc1a (Figures S3D and S3E). Because Pitpnc1a is exclusively detected in the zebrafish brain, including areas that have been implicated in larval sleep and arousal (Barlow and Rihel, 2017), we asked whether behavior might be affected in pitpnc1a−/− larvae by video-tracking them for several days (Rihel et al., 2010). Mutants showed a consistent increase in waking activity across a 14:10 hr light:dark cycle relative to pitpnc1a+/+ and pitpnc1a+/− (Figures 2C–2E).

    View all citing articles on Scopus
    View full text