Elsevier

Neuropharmacology

Volume 106, July 2016, Pages 20-34
Neuropharmacology

Role of histamine H1-receptor on behavioral states and wake maintenance during deficiency of a brain activating system: A study using a knockout mouse model

https://doi.org/10.1016/j.neuropharm.2015.12.014Get rights and content

Highlights

  • Long-term brain histamine deletion impairs EEG & sleep-wake quality causing sleepiness.

  • Such roles are largely mediated by the H1-receptor.

  • The cholinergic system is upregulated to ensure a wake amount in histamine-deficient mice.

  • This constitutes a major compensation when the brain is faced with deficient situations.

  • Understand how the brain remains awake in deficient situation is of physiological/clinical relevance.

Abstract

Using knockout (KO) mice lacking the histamine (HA)-synthesizing enzyme (histidine decarboxylase, HDC), we have previously shown the importance of histaminergic neurons in maintaining wakefulness (W) under behavioral challenges. Since the central actions of HA are mediated by several receptor subtypes, it remains to be determined which one(s) could be responsible for such a role. We have therefore compared the cortical-EEG, sleep and W under baseline conditions or behavioral/pharmacological stimuli in littermate wild-type (WT) and H1-receptor KO (H1−/−) mice. We found that H1−/− mice shared several characteristics with HDC KO mice, i.e. 1) a decrease in W after lights-off despite its normal baseline daily amount; 2) a decreased EEG slow wave sleep (SWS)/W power ratio; 3) inability to maintain W in response to behavioral challenges demonstrated by a decreased sleep latency when facing various stimuli. These effects were mediated by central H1-receptors. Indeed, in WT mice, injection of triprolidine, a brain-penetrating H1-receptor antagonist increased SWS, whereas ciproxifan (H3-receptor antagonist/inverse agonist) elicited W; all these injections had no effect in H1−/− mice. Finally, H1−/− mice showed markedly greater changes in EEG power (notably in the 0.8–5 Hz band) and sleep-wake cycle than in WT mice after application of a cholinergic antagonist or an indirect agonist, i.e., scopolamine or physostigmine. Hence, the role of HA in wake-promotion is largely ensured by H1-receptors. An upregulated cholinergic system may account for a quasi-normal daily amount of W in HDC or H1-receptor KO mice and likely constitutes a major compensatory mechanism when the brain is facing deficiency of an activating system.

This article is part of the Special Issue entitled ‘Histamine Receptors’.

Introduction

Neurons containing histamine (HA) are located in the tuberomamillary nucleus and the adjacent posterior hypothalamus. They send widespread inputs to most cerebral areas. In freely-moving cats and behaving mice, these neurons fire tonically and exclusively during wakefulness (W) (Sakai et al., 1990, Vanni-Mercier et al., 2003, Takahashi et al., 2006), a pattern of activity that is the most wake-selective one among the brain activating systems identified to date. This wake-selective activity enhances cellular excitability and activity of a wide range of target cells via different HA receptor subtypes (reviewed in Schwartz et al., 1991, Lin, 2000, Brown et al., 2001, Haas et al., 2008, Vu et al., 2015, Panula et al., 2015), contributing to a general cortical activation during W. Hence, impairment of the brain histaminergic transmission using pharmacological interventions or knockout (KO) mouse models causes somnolence and wake deficits, whereas enhancement of transmission promotes cortical arousal and W (Lin et al., 1986, Monti et al., 1991, Lin, 2000, Lin et al., 2011a). HA neurons make multiple interactions with other brain arousal systems such as cholinergic aminergic and orexinergic neurons (reviewed in McCormick, 1992, Lin, 2000, Haas et al., 2008, Haas and Lin, 2012). The latter ones send particularly dense inputs to HA cells and excite them by direct depolarization through orexin-2 receptors (Eriksson et al., 2001). Mice selectively lacking the orexin-2 receptors show signs of chronic sleepiness, a phenotype rescued by restoration of orexin-2 receptors on neurons in the tuberomamillary areas (Mochizuki et al., 2011). Recently, the wake-promoting effects of HA were shown to be balanced by its co-neurotransmitter GABA (Yu et al., 2015).

Using KO mice lacking the HA-synthesizing enzyme, histidine decarboxylase (HDC), we have previously shown that long-term abolition of HA impairs cortical EEG, deteriorates both sleep and waking quality, causing sleepiness and behavioral deficits. Consequently, these mice are unable to remain awake in crucial situations, such as at lights-off or when faced with an environmental change (Parmentier et al., 2002, Anaclet et al., 2009, Lin et al., 2011b).

Since the central actions of HA are mediated by several receptor subtypes, it remains to be determined which one(s) could be responsible for its wake-promoting effects. The importance of the H1-receptor has long been suggested because brain–penetrating H1-receptor antagonists cause sedation and drowsiness in man and increase in slow wave sleep in animals (Douglas, 1985, Nicholson and Stone, 1986, Schwartz et al., 1991, Yanai et al., 1999, Lin, 2000, Panula et al., 2015). However, whether the long-term loss of H1-receptors causes similar genotypes of HDC KO mice in terms of EEG, behavioral states and how the brain remains awake during deficiency of a brain activating system remains unknown. We have therefore studied and compared the cortical EEG and sleep-wake cycle under baseline conditions and after behavioral challenges in littermate wild type and H1-receptor knockout mice. In order to assess brain plasticity involved in wake maintenance during deficiency of an activating system, we also compared in those mice pharmacological responses to ligands of the aminergic and cholinergic systems known for their role in controlling cortical activities and the sleep-wake cycle.

Section snippets

Detection of the histamine H1-receptor gene using PCR and in situ hybridization

At 3 weeks of age, tail biopsies were taken from all mice and analyzed by PCR for genotyping. The WT allele was amplified using primers located within the H1 gene and the KO allele was amplified with one primer within the neomycin resistance cassette. These primers were 5′-TGA AGT ATC TGG CTC TGA GTG G-3′ (5′-primer 5′-upstream of H1R gene) and 5′- CCA TCG ATG GCT CCC TCC CTG GGA G -3′ (H1R gene 5′-primer complementary to H1R gene), the expected product size being 1100 base pairs. The mutant

Genotyping by PCR and in situ hybridization

We performed PCR on genomic DNA from tail biopsies from all mice. As shown in Fig. 1A, a strong H1 receptor signal, corresponding to a 1100 base pair band, was detected in the H1+/+ mice (animals 1–14 in Fig. 1), and a strong Neor signal, corresponding to a 900 base pair band, was detected in the H1−/− mice (animals 15–28), proof that, in the KO animals, the H1-receptor gene had been disrupted and the Neor gene inserted.

The deletion of H1-receptor gene was further confirmed by in situ

Major sleep-wake phenotypes of H1-receptor−/− mice

We showed here that the sleep-wake cycle in H1−/− mice was affected both quantitatively and qualitatively. On the one hand, these mice exhibited a deficit of W just after lights-off in spite of an unaffected daily amount. On the other hand, their cortical EEG showed a reduced SWS/W power ratio and a significant deficit of θ rhythms (3–9 Hz) during W. These changes likely impacted the animal's behavior, as they presented signs of sleepiness, uncovered through a significant decrease in sleep

Acknowledgments

This work was supported by INSERM U480, U628, U1028-Waking team, Claude Bernard University and by the European Contract No. QLRT-2001-00826 (5th PCRDT). The doctoral fellowship of R. Parmentier was provided by the Ministère de l’éducation nationale, de la recherche et de la technologie (France). The fellowship of Y. Zhao was provided by the Institut Franco-Chinois (Lyon, France). We thank the late Dr. Jean-Louis Valatx for help at the start of the project and Dr. Luc Gentet for critical reading

References (60)

  • C.H. Vanderwolf

    The electrocorticogram in relation to physiology and behavior: a new analysis

    Electroencephalogr. Clin. Neurophysiol.

    (1992)
  • G. Vanni-Mercier et al.

    Waking selective neurons in the posterior hypothalamus and their response to histamine H3-receptor ligands: an electrophysiological study in freely moving cats

    Behav. Brain Res.

    (2003)
  • G.1 Vanni-Mercier et al.

    Carbachol microinjections in the mediodorsal pontine tegmentum are unable to induce paradoxical sleep after caudal pontine and prebulbar transections in the cat

    Neurosci. Lett.

    (1991)
  • J. Velazquez-Moctezuma et al.

    Cholinergic antagonists and REM sleep generation

    Brain Res.

    (1991)
  • V.V. Vyazovskiy et al.

    Electrophysiological correlates of sleep homeostasis in freely behaving rats

    Prog. Brain Res.

    (2011)
  • X. Yu et al.

    Wakefulness is governed by GABA and histamine cotransmission

    Neuron

    (2015 Jul 1)
  • C. Anaclet et al.

    Orexin/hypocretin and histamine: distinct roles in the control of wakefulness demonstrated using knock-out mouse models

    J. Neurosci.

    (2009)
  • A.A. Borbely et al.

    Sleep homeostasis and models of sleep regulation

  • B. Boutrel et al.

    Involvement of 5-HT1A receptors in homeostatic and stress-induced adaptive regulations of paradoxical sleep: studies in 5-HT1A knock-out mice

    J. Neurosci.

    (2002)
  • R.E. Brown et al.

    Control of sleep and wakefulness

    Physiol. Rev.

    (2012)
  • C.R. Calcutt et al.

    Zolantidine (SK&F 95282) is a potent selective brain-penetrating histamine H2-receptor antagonist

    Br. J. Pharmacol.

    (1988)
  • S. Crochet et al.

    Effects of microdialysis application of monoamines on the EEG and behavioural states in the cat mesopontine tegmentum

    Eur J. Neurosci.

    (1999)
  • E. Dere et al.

    Changes in motoric, exploratory and emotional behaviours and neuronal acetylcholine content and 5-HT turnover in histidine decarboxylase-KO mice

    Eur. J. Neurosci.

    (2004)
  • W.W. Douglas

    Histamine and 5-hydroxytryptamine (serotonin) and their antagonists

  • K.S. Eriksson et al.

    Orexin/hypocretin excites the histaminergic neurons of the tuberomammillary nucleus

    J. Neurosci.

    (2001)
  • P. Franken et al.

    Genetic determinants of sleep regulation in inbred mice

    Sleep

    (1999)
  • E. Gondard et al.

    Increased histaminergic neurotransmission and sleep-wake alterations: study in histamine H3-receptor knock-out mice

    Neuropsychopharmacol.

    (2013)
  • H.L. Haas et al.

    Waking with the hypothalamus

    Pflugers Arch. Eur. J. Physiol.

    (2012)
  • H.L. Haas et al.

    Histamine in the nervous system

    Physiol. Rev.

    (2008)
  • L. Imeri et al.

    Differential effects of M2 and M3 muscarinic antagonists on the sleep-wake cycle

    Neuroreport

    (1991)
  • Cited by (38)

    • Neuroinflammation, sleep, and autoimmune disease

      2023, Encyclopedia of Sleep and Circadian Rhythms: Volume 1-6, Second Edition
    • Optogenetics in the study of the central nervous system during sleep

      2023, Encyclopedia of Sleep and Circadian Rhythms: Volume 1-6, Second Edition
    • Region-specific regulation of central histaminergic H3 receptor expression in a mouse model of cow's milk allergy

      2020, Brain Research
      Citation Excerpt :

      In contrast, H2R expression was unchanged in any of the brain regions (Fig. 3D). In addition to its role in wakefulness and arousal (Parmentier et al., 2016), H1R, but not H2R, has been implicated in the anxiogenic function of central histamine (Yuzurihara et al., 2000). Thus, the increased amount of H1R in the brain may also have contributed to the behavioral changes observed in our mouse model.

    • In vivo effects of the association of the psychoactive phenotiazine thioridazine on antitumor activity and hind limb paralysis induced by the native polypeptide crotamine

      2020, Toxicon
      Citation Excerpt :

      It is important to note that, due to its high lipophilicity, THD can cross the blood brain barrier, which could then induce more release of histamine by blocking the H3 receptors (Brabant et al., 2013). A final balance of these THD effects could conduct to a higher histamine release which then could induce the slow-wave sleep state and a membrane depolarization of motoneurons (Parmentier et al., 2016; Coslovich et al., 2018). In fact, THD was reported to increase the time necessary to fall in rotarod experiments of animal models of amyotrophic lateral sclerosis (ALS), which could also be an indicative of a possible role of this molecule in the muscle contractility (Seredenina et al., 2016).

    View all citing articles on Scopus
    1

    Equal contribution.

    2

    Present Institution: Forgetting Team, CRNL, INSERM-U1028, CNRS-UMR5292, Cl. Bernard University, Lyon, France.

    3

    Present Institution: University of Turku and Turku University Hospital, Finland.

    View full text