Elsevier

Brain Research

Volume 973, Issue 2, 30 May 2003, Pages 214-222
Brain Research

Research report
Spontaneous sleep in mice with targeted disruptions of neuronal or inducible nitric oxide synthase genes

https://doi.org/10.1016/S0006-8993(03)02484-3Get rights and content

Abstract

Nitric oxide (NO) affects almost every physiological process, including the regulation of sleep. There is strong evidence that NO plays an important role in rapid eye movement sleep (REMS) regulation. To further investigate the role of NO in sleep, we characterized spontaneous sleep in mice with targeted disruptions (knockout; KO) in the neuronal nitric oxide synthase (nNOS) or inducible (i)NOS genes. REMS in nNOS KO mice was substantially lower than that of their control mice. In contrast, the iNOS KO mice had significantly more REMS than their controls. Inducible NOS KO mice also had less non-REMS (NREMS) during the dark period. Results suggest that nNOS and iNOS play opposite roles in REMS regulation.

Introduction

Nitric oxide (NO) is involved in sleep regulation. NO is derived from arginine via the action of nitric oxide synthase (NOS). There are three types of NOSs: neuronal (nNOS, or NOS-1), inducible (iNOS, or NOS-2), and endothelial (eNOS, or NOS-3). nNOS and eNOS are constitutively expressed and nNOS is normally the major source of brain NO [17]. eNOS is mainly found in vascular endothelial cells and plays a role in blood pressure regulation [47]. In contrast, iNOS is expressed in brain during development but is not normally present in adulthood, although it is induced in glial cells in response to inflammatory mediators [34].

nNOS is present within sleep-related brain structures, such as the pedunculopontine tegmental nuclei (PPT), the laterodorsal tegmental nucleus (LDT) and the dorsal Raphe nucleus (DRN) (e.g., Ref. [30]). There is substantial evidence that NO in these nuclei plays a critical role in rapid eye movement sleep (REMS) regulation. For example, nNOS colocalizes with cholinergic neurons in the LDT/PPT; these neurons project to the medial pontine reticular formation (mPRF) and are crucial in REMS generation [46]. Microinjection of SNAP, a NO donor, into the PPT of cats increases REMS substantially [8]. On the other hand, microinjection of l-NAME, an inhibitor of NOS, into the PPT reduces REMS [8]. In rats, microinjection of l-arginine into the PPT increases the duration of non-REMS and the number of REMS episodes, while microinjection of another NOS inhibitor (l-NAPNA) into this area reduces both NREMS and REMS [14]. Further, local inhibition of NOS within the mPRF of cats reduces acetylcholine (ACh) release and decreases the amount of REMS [32], [33]. Microinjection of 7-NI, a selective nNOS inhibitor, into the DRN of rats decreases REMS [4]. In contrast, microinjection of l-NAME into the anterior diencephalic region increases REMS during the dark period, followed by a negative rebound during the light period [44].

In contrast to these microinjection studies, which indicate a role for NO in REMS regulation, the results from studies using systemic or intracerebroventricular (i.c.v.) routes of NO inhibitor administration are less clear. Systemic or i.c.v. injection of NOS inhibitors or NO donors also affect sleep, although the direction of the effect may depend on the time of the day. For instance, l-NAME given intravenously at light onset suppresses spontaneous NREMS and REMS [23], [37]. In addition, intraperitoneal (i.p.) injection of l-NAME suppresses sleep rebound after sleep deprivation [43]. In contrast, i.p. injection of similar doses of l-NAME at dark onset increases both NREMS and REMS [4]. In addition, the 7-NI given i.p. suppresses sleep when given either at dark onset or light onset [4], [11]. Administration of l-NAME via i.c.v. injection also suppresses both NREMS and REMS in rabbits and rats [21], [23]. Conversely, the i.c.v. administration of the NO donors molsidomine (SIN-1) or S-nitroso-N-acetylpenicillamine (SNAP) at dark onset promotes NREMS after a latency of 9 h in rats [20].

The basal forebrain/preoptic area is another important brain structure involved in the regulation of sleep/wakefulness cycles; it is hypothesized that an interaction between cholinergic and GABAergic systems is involved [50]. Ascending cholinergic projections from the basal forebrain play an essential role in cortical activation. Basal forebrain neurons also send descending projections, probably GABAergic, to key REMS regulating nuclei, such as the LDT/PPT, locus coeruleus, and dorsal raphe nucleus [48]. NO can affect both Ach and GABA release in the basal forebrain [26], [52]. Finally, pretreatment of rats with a GABA receptor antagonist reverses the sleep decrease caused by l-NAME [38].

In rats, soluble NOS activity in the brain has a diurnal variation, with higher levels during the dark period (active period) and lower levels during the light period [2]. Consistent with this finding, Cespuglio et al. [7] found that the highest cortical levels of NO occur during the awake state. Similarly, Williams et al. [54] found that extracellular NO concentrations in the thalamus are high during wakefulness and REMS and significantly lower during NREMS. Superficially, these data are inconsistent with the notion that NO is involved in the induction of sleep. However, these studies do not distinguish the anatomical source of NO or the NOS isoform measured, and it has been shown that NO in one site has different effects than NO in other sites within the brain [52].

Thus, currently it is clear that manipulation of NOS affects sleep. However, specific effects depend upon how drugs are given, the time of day they are given and what specific substance is given. Further, the use of NOS inhibitors is confounded by their relative lack of specificity for individual NOS isoforms. In this study, we use mice with targeted disruptions in the nNOS or iNOS genes, commonly referred to as nNOS or iNOS knockouts (KO) to characterize their spontaneous sleep patterns. We report that mice lacking nNOS have less REMS while those lacking iNOS have more REMS.

Section snippets

Animals

Male 8–12-week-old mice (20–28 g) acquired from Jackson Laboratory (Bar Harbor, ME, USA) were used. The specific strains purchased were nNOS KO mice (B6;129S4-Nos1tm1Plh), nNOS control mice (B6129SF2/J); iNOS KO mice (B6.129P2-Nos2tm1Lau), and iNOS control mice (C57BL/6J). (The control strains used were those designated as optimal controls by Jackson Laboratory geneticists). The embryonic stem (ES) cell line used to generate the nNOS KOs was J1, derived from the 129/SvJae substrain. The ES cell

Results

All animals appeared behaviorally normal. Although there are reports of aggressive behavior of nNOS KO mice [40], this has not been confirmed in the Jackson Laboratory colony (Jackson website, http://jaxmice.jax.org/) nor was such behavior observed by us. There were also no abnormalities in baseline EEG recording in any of the strains.

Discussion

The nNOS mutant mice were generated by deletion of part of the exon that contains the initiation codon ATG, and nNOS KOs have a 95% reduction of NOS activity in the brain [17]. The residual activity is partly from eNOS, both in neurons and blood vessels, and partly from variations of nNOS RNA splicing forms that do not contain the disrupted exon [17]. Although nNOS KOs have enlarged stomachs and reduced infarct size following cerebral ischemia, they are viable and fertile. No neuroanatomic or

Acknowledgements

This work was supported in part by the National Institute of Health grants HD 36520 and NS 25378. We thank Richard A. Brown and Sanjib Mukherjee for their assistance.

References (54)

  • P.L. Huang et al.

    Genetic analysis of NOS isoforms using nNOS and eNOS knockout animals

    Prog. Brain Res.

    (1998)
  • J.P. Huston et al.

    Extracellular adenosine levels in neostriatum and hippocampus during rest and activity periods of rats

    Neuroscience

    (1996)
  • L. Kapás et al.

    Nitric oxide donors SIN-1 and SNAP promote nonrapid-eye-movement sleep in rats

    Brain Res. Bull.

    (1996)
  • L. Kapás et al.

    Inhibition of nitric oxide synthesis inhibits rat sleep

    Brain Res.

    (1994)
  • L. Kapas et al.

    The effects of immunolesions of nerve growth factor-receptive neurons by 192 IgG-saporin on sleep

    Brain Res.

    (1996)
  • M.M. Kraus et al.

    Involvement of nitric oxide, cyclic GMP and phosphodiesterase 5 in excitatory amino acid and GABA release in the nucleus accumbens evoked by activation of the hippocampal fimbria

    Neuroscience

    (2002)
  • R. Lathe

    Mice, gene targeting and behaviour: more than just genetic background

    Trends Neurosci.

    (1996)
  • L. Léger et al.

    Localization of nitric oxide-synthesizing neurons sending projections to the dorsal raphe nucleus of the rat

    Neurosci. Lett.

    (1998)
  • J. Licinio et al.

    Brain iNOS: current understanding and clinical implications

    Mol. Med. Today

    (1999)
  • J.M. Monti et al.

    Role of nitric oxide in sleep regulation: effects of l-NAME, an inhibitor of nitric oxide synthase, on sleep in rats

    Behav. Brain Res.

    (1999)
  • J.M. Monti et al.

    Increase of waking and reduction of NREM and REM sleep after nitric oxide synthase inhibition: prevention with GABA(A) or adenosine A(1) receptor agonists

    Behav. Brain Res.

    (2001)
  • K. Semba

    Multiple output pathways of the basal forebrain: organization, chemical heterogeneity, and roles in vigilance

    Behav. Brain Res.

    (2000)
  • H. Wang et al.

    Presence of neuronal nitric oxide synthase in the suprachiasmatic nuclei of mouse and rat

    Neuroscience

    (1996)
  • A.A. Borbely

    From slow waves to sleep homeostasis: new perspectives

    Arch. Ital. Biol.

    (2001)
  • M. Caillol et al.

    Endothelial and neuronal nitric oxide synthases are present in the suprachiasmatic nuclei of Syrian hamsters and rats

    Eur. J. Neurosci.

    (2000)
  • R. Cespuglio et al.

    5-Hydroxyindoles compounds and nitric oxide voltammetric detection in the rat brain: changes occurring throughout the sleep–wake cycle

    J. Neural Transm.

    (1998)
  • S. Datta et al.

    Endogenous and exogenous nitric oxide in the pedunculopontine tegmentum induces sleep

    Synapse

    (1997)
  • Cited by (71)

    • Ontogenesis of the molecular response to sleep loss

      2023, Neurobiology of Sleep and Circadian Rhythms
    • The implications of hypothalamic abnormalities for schizophrenia

      2021, Handbook of Clinical Neurology
      Citation Excerpt :

      Importantly, treatment with NO donors during the spontaneous sleep–wake cycle led to increased sleep quantity and quality (Kalinchuk et al., 2006). Moreover, there is evidence that nNOS mutant mice suffer from a drastically reduced REM sleep (Chen et al., 2003). No other sleep-regulating compounds have been studied in the hypothalamus of individuals with schizophrenia or in animal models.

    • Genes and Ion Channels in the Circadian and Homeostatic Regulation of Sleep

      2019, Handbook of Behavioral Neuroscience
      Citation Excerpt :

      Lesion studies and optogenetics have identified several loci whose activity corresponds to sleep pressure after sleep deprivation or affect the transition between sleep states (e.g., from nonrapid eye movement (NREM) sleep to rapid eye movement (REM) sleep) (Buzsaki et al., 1988; Chen et al., 2018; Chung et al., 2017; Fuller, Sherman, Pedersen, Saper, & Lu, 2011; Hassani, Lee, Henny, & Jones, 2009; Hobson, McCarley, & Wyzinski, 1975; John, Wu, Boehmer, & Siegel, 2004; Rasmussen, Morilak, & Jacobs, 1986; Saper, Chou, & Scammell, 2001; Takahashi, Lin, & Sakai, 2006; Vanni-Mercier, Gigout, Debilly, & Lin, 2003). Recent forward and reverse genetics have also identified several sleep genes whose mutant mice or flies have increased or decreased sleep amount per day, which suggests that sleep amount may be regulated by an intracellular mechanism (Alexandre et al., 2006; Anderson et al., 2005; Bohnet, Traynor, Majde, Kacsoh, & Krueger, 2004; Boutrel, Franc, Hen, Hamon, & Adrien, 1999; Boutrel, Monaca, Hen, Hamon, & Adrien, 2002; Chen, Majde, & Krueger, 2003; Cirelli et al., 2005; Comai, Ochoa-Sanchez, & Gobbi, 2013; Deboer, Fontana, & Tobler, 2002; Douglas et al., 2007; Espinosa, Marks, Heintz, & Joho, 2004; Fentress et al., 2013; Fitzpatrick et al., 2012; Fonck et al., 2005; Frank, Stryker, & Tecott, 2002; Freyburger, Poirier, Carrier, & Mongrain, 2017; Funato et al., 2016; Gondard et al., 2013; Goutagny et al., 2005; Graves et al., 2003; Hajdu, Obal, Fang, Krueger, & Rollo, 2002; Hunsley & Palmiter, 2003, 2004; Jhaveri, Ramkumar, Trammell, & Toth, 2006; Kim et al., 2015; Koh et al., 2008; Kovalzon et al., 2017; Laposky et al., 2006; Lee, Kim, & Shin, 2004; Lonart, Tang, Simsek-Duran, Machida, & Sanford, 2008; Madrid-Lopez et al., 2017; Massie, Boland, Kapas, & Szentirmai, 2018; Morrow & Opp, 2005; Obal, Alt, Taishi, Gardi, & Krueger, 2003; Obal et al., 2001, 2005; Ouyang, Hellman, Abel, & Thomas, 2004; Parmentier et al., 2002; Pimentel et al., 2016; Popa et al., 2005; Shiromani et al., 2000; Silvani et al., 2014; Tatsuki et al., 2016; Thomas, Schwartz, Saxe, & Kilduff, 2017; Wisor et al., 2001; Wisor et al., 2003; Young, Geurts, Hodges, & Cummings, 2017; Zhang, Obal, Fang, Collins, & Krueger, 1996). In this chapter, we discuss the cellular and molecular mechanisms of circadian and homeostatic regulation of sleep.

    • Memory consolidation and inducible nitric oxide synthase expression during different sleep stages in Parkinson disease

      2014, Sleep Medicine
      Citation Excerpt :

      However, the persistent activation of iNOS may lead to the overproduction of NO. In studies on nNOS or iNOS genes that have used transgenic mice, the prominence of NO in REM sleep regulation increased. In iNOS KO animals, REM sleep was increased compared with that of the control mice [9]. In our study, REM sleep significantly increased in the PD patients, suggesting that the downregulation of the iNOS expression might be associated with the increase in REM sleep in PD patients.

    • Nitric oxide in the regulation of the sleep-wake states

      2012, Sleep Medicine Reviews
      Citation Excerpt :

      nNOS inhibitors, including the most selective compound identified to date, 3-bromo-7-nitroindazole (7-NI), administered either i.p.81–83 or locally into sleep-permissive or sleep-executive structures, i.e., nRD,82 LDT,84 and PPT,85 significantly decrease SWS, SWA activity during SWS and REM sleep. Transgenic mice with a nNOS-targeted disruption show a REM sleep deficit compared to control wild-type animals.86 In adult rats, a contribution of the NO/NOSs production to the sleep rebound that follows SD was first suggested after pharmacological treatment with l-NAME (Nω-nitro-l-arginine methyl ester), a non-specific inhibitor of the NOSs.

    View all citing articles on Scopus
    View full text