Pharmacogenetic modulation of orexin neurons alters sleep/wakefulness states in mice

Koh Sasaki, Mika Suzuki, Michihiro Mieda, Natsuko Tsujino, Bryan Roth, Takeshi Sakurai, Koh Sasaki, Mika Suzuki, Michihiro Mieda, Natsuko Tsujino, Bryan Roth, Takeshi Sakurai

Abstract

Hypothalamic neurons expressing neuropeptide orexins are critically involved in the control of sleep and wakefulness. Although the activity of orexin neurons is thought to be influenced by various neuronal input as well as humoral factors, the direct consequences of changes in the activity of these neurons in an intact animal are largely unknown. We therefore examined the effects of orexin neuron-specific pharmacogenetic modulation in vivo by a new method called the Designer Receptors Exclusively Activated by Designer Drugs approach (DREADD). Using this system, we successfully activated and suppressed orexin neurons as measured by Fos staining. EEG and EMG recordings suggested that excitation of orexin neurons significantly increased the amount of time spent in wakefulness and decreased both non-rapid eye movement (NREM) and rapid eye movement (REM) sleep times. Inhibition of orexin neurons decreased wakefulness time and increased NREM sleep time. These findings clearly show that changes in the activity of orexin neurons can alter the behavioral state of animals and also validate this novel approach for manipulating neuronal activity in awake, freely-moving animals.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Specific expression of hM3D q…
Figure 1. Specific expression of hM3Dq and hM4Di in orexin neurons in mice.
A, Schematic representation of double-floxed Cre-dependent AAV vector expressing hM3Dq and hM4Di under control of EF-1α promoter (rAAV-DIO-HAhM3Dq and rAAV-DIO-HAhM4Di). DIO, double-floxed inverted open reading frame; ITR, inverted terminal repeat; WPRE, woodchuck hepatitis virus post-transcriptional regulatory element. B, Coronal brain sections at the level of LHA (bregma-1.8 mm), prepared from orexin-cre mice expressing hM3Dq or hM4Di following injection of Cre-dependent rAAV-DIO-HAhM3Dq or rAAV-DIO-HAhM4Di. Sections were stained with anti-HA antibody. Scale bars, 40 µm. C, Upper panels, Coronal section of LHA prepared from orexin-cre mice expressing hM3Dq and double stained with anti-orexin antibody (green) and anti-HA antibody (red), showing that most of orexin-ir neurons express HAhM3Dq. Lower panels, Coronal section of LHA prepared from orexin-cre mice expressing hM4Di and double stained with anti-orexin antibody (green) and anti-HA antibody (red), showing that most of orexin-ir neurons express HAhM4Di.
Figure 2. Activation or inhibition of orexin…
Figure 2. Activation or inhibition of orexin neurons by DREADD.
A–D, Representative images of Fos expression in orexin neurons, as shown by double staining of the LHA regions of wild type (A, C) and orexin-cre mice (B, D) injected with cre-activatable AAV carrying hM3Dq (A, B) or hM4Di (C, D) (rAAV-DIO-HAhM3Dq or rAAV-DIO-HAhM4Di) after injection of CNO. A, Only a small numbers of Fos-IR nuclei were observed in orexin-immunoreactive neurons from wild type mice administered CNO at 13:00 (ZT4) and killed at 15:00 (ZT6). Inset, high power view. B, Number of Fos- and orexin-double-positive neurons was higher in orexin-cre than in wild type (A) administered CNO at ZT4 and killed at ZT6. Inset, high power view. C, Many Fos- and orexin-double positive neurons were observed in wild type mice administered CNO at ZT12 and killed at ZT14. Inset, high power view. D, Fewer Fos- and orexin-double-positive neurons were observed in orexin-cre than in wild type mice administered CNO at ZT12 and killed at ZT14. Inset, high power view. E, Numbers of double-positive neurons at ZT6 in wild type and orexin-cre transgenic mice injected with hM3Dq virus and administered CNO (N = 5). F, Numbers of double-positive neurons at ZT14 in wild type and orexin-cre transgenic mice injected with hM4Di virus and administered CNO (N = 5).
Figure 3. Effect of stimulation of orexinergic…
Figure 3. Effect of stimulation of orexinergic tone by hM3Dq on vigilance states of mice during light and dark periods (left and right panels, respectively).
A. Hourly analysis of sleep/wake states in transgenic and wild-type mice, both injected with rAAV-DIO-HAhM3Dq, after administration of CNO at ZT4 or ZT12. Amounts of wakefulness (Wake, upper panels), NREM sleep (middle panels), and REM sleep (lower panels) are shown. B, Latency to NREM sleep and REM sleep after CNO administration (N = 6 for transgenic mice, N = 5 for wild-type mice).
Figure 4. Effect of inhibition of orexinergic…
Figure 4. Effect of inhibition of orexinergic tone by hM4Di on vigilance states of mice during light and dark periods (left and right panels, respectively).
A. Hourly analysis of sleep/wake states in transgenic and wild-type mice, both injected with rAAV-DIO-HAhM4Di, after administration of CNO at ZT4 or ZT12. Amounts of wakefulness (Wake, upper panels), NREM sleep (middle panels), and REM sleep (lower panels) are shown. B. Latency to NREM sleep and REM sleep after CNO administration (N = 5).

References

    1. Hagan JJ, Leslie RA, Patel S, Evans ML, Wattam TA, et al. Orexin A activates locus coeruleus cell firing and increases arousal in the rat. Proc Natl Acad Sci U S A. 1999;96:10911–10916.
    1. Sakurai T. The neural circuit of orexin (hypocretin): maintaining sleep and wakefulness. Nat Rev Neurosci. 2007;8:171–181.
    1. Lee MG, Hassani OK, Jones BE. Discharge of identified orexin/hypocretin neurons across the sleep-waking cycle. J Neurosci. 2005;25:6716–6720.
    1. Takahashi K, Lin JS, Sakai K. Neuronal activity of orexin and non-orexin waking-active neurons during wake-sleep states in the mouse. Neuroscience. 2008;153:860–870.
    1. Mileykovskiy BY, Kiyashchenko LI, Siegel JM. Behavioral correlates of activity in identified hypocretin/orexin neurons. Neuron. 2005;46:787–798.
    1. Carter ME, Adamantidis A, Ohtsu H, Deisseroth K, de Lecea L. Sleep homeostasis modulates hypocretin-mediated sleep-to-wake transitions. J Neurosci. 2009;29:10939–10949.
    1. Adamantidis AR, Zhang F, Aravanis AM, Deisseroth K, de Lecea L. Neural substrates of awakening probed with optogenetic control of hypocretin neurons. Nature. 2007;450:420–424.
    1. Armbruster BN, Li X, Pausch MH, Herlitze S, Roth BL. Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand. Proc Natl Acad Sci U S A. 2007;104:5163–5168.
    1. Alexander GM, Rogan SC, Abbas AI, Armbruster BN, Pei Y, et al. Remote control of neuronal activity in transgenic mice expressing evolved G protein-coupled receptors. Neuron. 2009;63:27–39.
    1. Ferguson SM, Eskenazi D, Ishikawa M, Wanat MJ, Phillips PE, et al. Transient neuronal inhibition reveals opposing roles of indirect and direct pathways in sensitization. Nat Neurosci. 2011;14:22–24.
    1. Tsujino N, Yamanaka A, Ichiki K, Muraki Y, Kilduff TS, et al. Cholecystokinin activates orexin/hypocretin neurons through the cholecystokinin A receptor. J Neurosci. 2005;25:7459–7469.
    1. Matsuki T, Nomiyama M, Takahira H, Hirashima N, Kunita S, et al. Selective loss of GABA(B) receptors in orexin-producing neurons results in disrupted sleep/wakefulness architecture. Proc Natl Acad Sci U S A. 2009;106:4459–4464.
    1. Tsai HC, Zhang F, Adamantidis A, Stuber GD, Bonci A, et al. Phasic firing in dopaminergic neurons is sufficient for behavioral conditioning. Science. 2009;324:1080–1084.
    1. Krashes MJ, Koda S, Ye C, Rogan SC, Adams AC, et al. J Clin Invest; 2011. Rapid, reversible activation of AgRP neurons drives feeding behavior in mice.
    1. Estabrooke IV, McCarthy MT, Ko E, Chou TC, Chemelli RM, et al. Fos expression in orexin neurons varies with behavioral state. J Neurosci. 2001;21:1656–1662.
    1. Yoshida Y, Fujiki N, Nakajima T, Ripley B, Matsumura H, et al. Fluctuation of extracellular hypocretin-1 (orexin A) levels in the rat in relation to the light-dark cycle and sleep-wake activities. Eur J Neurosci. 2001;14:1075–1081.
    1. Vanni-Mercier G, Sakai K, Jouvet M. Neurons specifiques de l'eveil dans l'hypothalamus posterieur du chat. CR Acad Sci. 1984;III 298:195–200.
    1. Lu J, Bjorkum AA, Xu M, Gaus SE, Shiromani PJ, et al. Selective activation of the extended ventrolateral preoptic nucleus during rapid eye movement sleep. J Neurosci. 2002;22:4568–4576.
    1. Sakurai T. Roles of orexins and orexin receptors in central regulation of feeding behavior and energy homeostasis. CNS Neurol Disord Drug Targets. 2006;5:313–325.
    1. Yoshida K, McCormack S, Espana RA, Crocker A, Scammell TE. Afferents to the orexin neurons of the rat brain. J Comp Neurol. 2006;494:845–861.
    1. Yamanaka A, Beuckmann CT, Willie JT, Hara J, Tsujino N, et al. Hypothalamic orexin neurons regulate arousal according to energy balance in mice. Neuron. 2003;38:701–713.
    1. Burdakov D, Gerasimenko O, Verkhratsky A. Physiological changes in glucose differentially modulate the excitability of hypothalamic melanin-concentrating hormone and orexin neurons in situ. J Neurosci. 2005;25:2429–2433.
    1. Auricchio A, Hildinger M, O'Connor E, Gao GP, Wilson JM. Isolation of highly infectious and pure adeno-associated virus type 2 vectors with a single-step gravity-flow column. Hum Gene Ther. 2001;12:71–76.
    1. Zhong L, Li B, Mah CS, Govindasamy L, Agbandje-McKenna M, et al. Next generation of adeno-associated virus 2 vectors: point mutations in tyrosines lead to high-efficiency transduction at lower doses. Proc Natl Acad Sci U S A. 2008;105:7827–7832.
    1. Nambu T, Sakurai T, Mizukami K, Hosoya Y, Yanagisawa M, et al. Distribution of orexin neurons in the adult rat brain. Brain Res. 1999;827:243–260.

Source: PubMed

3
Tilaa