Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Neurosci ; 41(25): 5440-5452, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34006590

ABSTRACT

Neural activity is diverse, and varies depending on brain regions and sleep/wakefulness states. However, whether astrocyte activity differs between sleep/wakefulness states, and whether there are differences in astrocyte activity among brain regions remain poorly understood. Therefore, in this study, we recorded astrocyte intracellular calcium (Ca2+) concentrations of mice during sleep/wakefulness states in the cortex, hippocampus, hypothalamus, cerebellum, and pons using fiber photometry. For this purpose, male transgenic mice expressing the genetically encoded ratiometric Ca2+ sensor YCnano50 specifically in their astrocytes were used. We demonstrated that Ca2+ levels in astrocytes substantially decrease during rapid eye movement (REM) sleep, and increase after the onset of wakefulness. In contrast, differences in Ca2+ levels during non-REM (NREM) sleep were observed among the different brain regions, and no significant decrease was observed in the hypothalamus and pons. Further analyses focusing on the transition between sleep/wakefulness states and correlation analysis with the duration of REM sleep showed that Ca2+ dynamics differs among brain regions, suggesting the existence of several clusters, i.e., the first comprising the cortex and hippocampus, the second comprising the hypothalamus and pons, and the third comprising the cerebellum. Our study thus demonstrated that astrocyte Ca2+ levels change substantially according to sleep/wakefulness states. These changes were consistent in general unlike neural activity. However, we also clarified that Ca2+ dynamics varies depending on the brain region, implying that astrocytes may play various physiological roles in sleep.SIGNIFICANCE STATEMENT Sleep is an instinctive behavior of many organisms. In the previous five decades, the mechanism of the neural circuits controlling sleep/wakefulness states and the neural activities associated with sleep/wakefulness states in various brain regions have been elucidated. However, whether astrocytes, which are a type of glial cell, change their activity during different sleep/wakefulness states was poorly understood. Here, we demonstrated that dynamic changes in astrocyte Ca2+ concentrations occur in the cortex, hippocampus, hypothalamus, cerebellum, and pons of mice during natural sleep. Further analyses demonstrated that Ca2+ dynamics slightly differ among different brain regions, implying that the physiological roles of astrocytes in sleep/wakefulness might vary depending on the brain region.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Calcium/metabolism , Sleep/physiology , Wakefulness/physiology , Animals , Male , Mice , Mice, Transgenic
2.
Adv Exp Med Biol ; 1293: 391-406, 2021.
Article in English | MEDLINE | ID: mdl-33398828

ABSTRACT

Although sleep is an absolutely essential physiological phenomenon for maintaining normal health in animals, little is known about its function to date. In this section, I introduce the application of optogenetics to freely behaving animals for the purpose of characterizing neural circuits involved in the regulation of sleep/wakefulness. Applying optogenetics to the specific neurons involved in sleep/wakefulness regulation enabled the precise control of the sleep/wakefulness states between wakefulness, non-rapid eye movement (NREM) sleep, and REM sleep states. For example, selective activation of orexin neurons using channelrhodopsin-2 and melanopsin induced a transition from sleep to wakefulness. In contrast, suppression of these neurons using halorhodopsin and archaerhodopsin induced a transition from wakefulness to NREM sleep and increased the time spent in NREM sleep. Selective activation of melanin-concentrating hormone (MCH) neurons induced a transition from NREM sleep to REM sleep and prolonged the time spent in REM sleep, which was accompanied by a decrease in NREM sleep time. Optogenetics was first introduced to orexin neurons in 2007 and has since rapidly spread throughout the field of neuroscience. In the last 13 years or so, neural nuclei and the cell types that control sleep/wakefulness have been identified. The use of optogenetic studies has greatly contributed to the elucidation of the neural circuits involved in the regulation of sleep/wakefulness.


Subject(s)
Optogenetics , Wakefulness , Animals , Neurons , Sleep , Sleep, REM
3.
J Neurosci ; 39(47): 9435-9452, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31628177

ABSTRACT

Narcolepsy, characterized by excessive daytime sleepiness, is associated with dysfunction of the hypothalamic hypocretin/orexin (Hcrt) system, either due to extensive loss of Hcrt cells (Type 1, NT1) or hypothesized Hcrt signaling impairment (Type 2, NT2). Accordingly, efforts to recapitulate narcolepsy-like symptoms in mice have involved ablating these cells or interrupting Hcrt signaling. Here, we describe orexin/Arch mice, in which a modified archaerhodopsin-3 gene was inserted downstream of the prepro-orexin promoter, resulting in expression of the yellow light-sensitive Arch-3 proton pump specifically within Hcrt neurons. Histological examination along with ex vivo and in vivo electrophysiological recordings of male and female orexin/Arch mice demonstrated silencing of Hcrt neurons when these cells were photoilluminated. However, high expression of the Arch transgene affected cellular and physiological parameters independent of photoillumination. The excitability of Hcrt neurons was reduced, and both circadian and metabolic parameters were perturbed in a subset of orexin/Arch mice that exhibited high levels of Arch expression. Orexin/Arch mice also had increased REM sleep under baseline conditions but did not exhibit cataplexy, a sudden loss of muscle tone during wakefulness characteristic of NT1. These aberrations resembled some aspects of mouse models with Hcrt neuron ablation, yet the number of Hcrt neurons in orexin/Arch mice was not reduced. Thus, orexin/Arch mice may be useful to investigate Hcrt system dysfunction when these neurons are intact, as is thought to occur in narcolepsy without cataplexy (NT2). These results also demonstrate the utility of extended phenotypic screening of transgenic models when specific neural circuits have been manipulated.SIGNIFICANCE STATEMENT Optogenetics has become an invaluable tool for functional dissection of neural circuitry. While opsin expression is often achieved by viral injection, stably integrated transgenes offer some practical advantages. Here, we demonstrate successful transgenic expression of an inhibitory opsin in hypocretin/orexin neurons, which are thought to promote or maintain wakefulness. Both brief and prolonged illumination resulted in inhibition of these neurons and induced sleep. However, even in the absence of illumination, these cells exhibited altered electrical characteristics, particularly when transgene expression was high. These aberrant properties affected metabolism and sleep, resulting in a phenotype reminiscent of the narcolepsy Type 2, a sleep disorder for which no good animal model currently exists.


Subject(s)
Archaeal Proteins/biosynthesis , Brain/metabolism , Narcolepsy/metabolism , Neurons/metabolism , Orexins/metabolism , Animals , Archaeal Proteins/genetics , Brain/cytology , Brain Chemistry/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Narcolepsy/genetics , Neurons/chemistry , Optogenetics/methods , Orexins/genetics , Organ Culture Techniques
4.
J Neurosci ; 34(19): 6495-509, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24806676

ABSTRACT

The sleep disorder narcolepsy results from loss of hypothalamic orexin/hypocretin neurons. Although narcolepsy onset is usually postpubertal, current mouse models involve loss of either orexin peptides or orexin neurons from birth. To create a model of orexin/hypocretin deficiency with closer fidelity to human narcolepsy, diphtheria toxin A (DTA) was expressed in orexin neurons under control of the Tet-off system. Upon doxycycline removal from the diet of postpubertal orexin-tTA;TetO DTA mice, orexin neurodegeneration was rapid, with 80% cell loss within 7 d, and resulted in disrupted sleep architecture. Cataplexy, the pathognomic symptom of narcolepsy, occurred by 14 d when ∼5% of the orexin neurons remained. Cataplexy frequency increased for at least 11 weeks after doxycycline. Temporary doxycycline removal followed by reintroduction after several days enabled partial lesion of orexin neurons. DTA-induced orexin neurodegeneration caused a body weight increase without a change in food consumption, mimicking metabolic aspects of human narcolepsy. Because the orexin/hypocretin system has been implicated in the control of metabolism and addiction as well as sleep/wake regulation, orexin-tTA; TetO DTA mice are a novel model in which to study these functions, for pharmacological studies of cataplexy, and to study network reorganization as orexin input is lost.


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Narcolepsy/drug therapy , Neurons/drug effects , Neuropeptides/antagonists & inhibitors , Animals , Body Weight/drug effects , Body Weight/physiology , Cataplexy/physiopathology , Diphtheria Toxin/genetics , Disease Models, Animal , Doxycycline/pharmacology , Drinking/drug effects , Drinking/physiology , Eating/drug effects , Eating/physiology , Electroencephalography , Electromyography , Female , Food , Male , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Narcolepsy/physiopathology , Orexins , Sleep/physiology , Wakefulness/physiology
5.
J Neurosci ; 34(20): 6896-909, 2014 May 14.
Article in English | MEDLINE | ID: mdl-24828644

ABSTRACT

Melanin-concentrating hormone (MCH) is a neuropeptide produced in neurons sparsely distributed in the lateral hypothalamic area. Recent studies have reported that MCH neurons are active during rapid eye movement (REM) sleep, but their physiological role in the regulation of sleep/wakefulness is not fully understood. To determine the physiological role of MCH neurons, newly developed transgenic mouse strains that enable manipulation of the activity and fate of MCH neurons in vivo were generated using the recently developed knockin-mediated enhanced gene expression by improved tetracycline-controlled gene induction system. The activity of these cells was controlled by optogenetics by expressing channelrhodopsin2 (E123T/T159C) or archaerhodopsin-T in MCH neurons. Acute optogenetic activation of MCH neurons at 10 Hz induced transitions from non-REM (NREM) to REM sleep and increased REM sleep time in conjunction with decreased NREM sleep. Activation of MCH neurons while mice were in NREM sleep induced REM sleep, but activation during wakefulness was ineffective. Acute optogenetic silencing of MCH neurons using archaerhodopsin-T had no effect on any vigilance states. Temporally controlled ablation of MCH neurons by cell-specific expression of diphtheria toxin A increased wakefulness and decreased NREM sleep duration without affecting REM sleep. Together, these results indicate that acute activation of MCH neurons is sufficient, but not necessary, to trigger the transition from NREM to REM sleep and that MCH neurons also play a role in the initiation and maintenance of NREM sleep.


Subject(s)
Hypothalamic Hormones/physiology , Melanins/physiology , Neurons/metabolism , Pituitary Hormones/physiology , Sleep/physiology , Wakefulness/physiology , Animals , Mice , Mice, Transgenic , Optogenetics
6.
Int J Neuropsychopharmacol ; 17(11): 1777-83, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24834486

ABSTRACT

Whether increased serotonin (5-HT) release in the forebrain attenuates or enhances anxiety has been controversial for over 25 yr. Although there is considerable indirect evidence, there is no direct evidence that indicates a relationship between acute 5-HT release and anxiety. In particular, there is no known method that can reversibly, selectively, and temporally control serotonergic activity. To address this issue, we generated transgenic animals to manipulate the firing rates of central 5-HT neurons by optogenetic methods. Activation of serotonergic neurons in the median raphe nucleus was correlated to enhanced anxiety-like behaviour in mice, whereas activation of serotonergic neurons in the dorsal raphe nucleus had no effect on anxiety-like behaviour. These results indicate that an acute increase in 5-HT release from the median raphe nucleus enhances anxiety.


Subject(s)
Anxiety/etiology , Anxiety/metabolism , Midbrain Raphe Nuclei/cytology , Optogenetics , Serotonergic Neurons/physiology , Serotonin/metabolism , Action Potentials/physiology , Analysis of Variance , Animals , Anxiety/pathology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Channelrhodopsins , In Vitro Techniques , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microdialysis , Mutation/genetics , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
7.
Neurosci Res ; 189: 54-59, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36572252

ABSTRACT

Dreams are mental experiences, including perceptions, thoughts, and emotions, that occur during sleep. In dreams, hallucinatory perceptions, particularly visual and motoric, are often accompanied by negative emotions. When people dream, they perceive them as real even though they are bizarre and distorted in time and space. People often cannot recall their dreams, even though people dream every night. Dreaming is a strange physiological phenomenon. Research has demonstrated that dreaming is closely associated with rapid eye movement (REM) sleep. It is known that dreaming also occurs during non-REM (NREM) sleep, but the content appears to be different. Dreams during REM sleep tend to be longer, more vivid, more story-like, and more bizarre than those during NREM sleep. In this review, the neural circuits underlying dreaming and the physiological functions associated with it are summarized. Two major theories have been proposed regarding the neural circuits involved in dreaming. One is that dreams are generated by the activation of neural activity in the brainstem and its signal transmission to the cortex. The other is that dreams are caused by forebrain activation by dopamine. Whereas the physiological function of dreams remains unclear, several hypotheses have been proposed that are associated with memory and emotions.


Subject(s)
Dreams , Sleep , Humans , Dreams/physiology , Dreams/psychology , Sleep, REM/physiology , Emotions , Mental Recall/physiology
8.
Sleep ; 46(9)2023 09 08.
Article in English | MEDLINE | ID: mdl-37478470

ABSTRACT

Ponto-geniculo-occipital or pontine (P) waves have long been recognized as an electrophysiological signature of rapid eye movement (REM) sleep. However, P-waves can be observed not just during REM sleep, but also during non-REM (NREM) sleep. Recent studies have uncovered that P-waves are functionally coupled with hippocampal sharp wave ripples (SWRs) during NREM sleep. However, it remains unclear to what extent P-waves during NREM sleep share their characteristics with P-waves during REM sleep and how the functional coupling to P-waves modulates SWRs. Here, we address these issues by performing multiple types of electrophysiological recordings and fiber photometry in both sexes of mice. P-waves during NREM sleep share their waveform shapes and local neural ensemble dynamics at a short (~100 milliseconds) timescale with their REM sleep counterparts. However, the dynamics of mesopontine cholinergic neurons are distinct at a longer (~10 seconds) timescale: although P-waves are accompanied by cholinergic transients, the cholinergic tone gradually reduces before P-wave genesis during NREM sleep. While P-waves are coupled to hippocampal theta rhythms during REM sleep, P-waves during NREM sleep are accompanied by a rapid reduction in hippocampal ripple power. SWRs coupled with P-waves are short-lived and hippocampal neural firing is also reduced after P-waves. These results demonstrate that P-waves are part of coordinated sleep-related activity by functionally coupling with hippocampal ensembles in a state-dependent manner.


Subject(s)
Eye Movements , Occipital Lobe , Male , Female , Animals , Mice , Occipital Lobe/physiology , Geniculate Bodies/physiology , Sleep/physiology , Hippocampus/physiology , Pons/physiology
9.
J Neurosci ; 31(29): 10529-39, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21775598

ABSTRACT

Orexin/hypocretin neurons have a crucial role in the regulation of sleep and wakefulness. To help determine how these neurons promote wakefulness, we generated transgenic mice in which orexin neurons expressed halorhodopsin (orexin/Halo mice), an orange light-activated neuronal silencer. Slice patch-clamp recordings of orexin neurons that expressed halorhodopsin demonstrated that orange light photic illumination immediately hyperpolarized membrane potential and inhibited orexin neuron discharge in proportion to illumination intensity. Acute silencing of orexin neurons in vivo during the day (the inactive period) induced synchronization of the electroencephalogram and a reduction in amplitude of the electromyogram that is characteristic of slow-wave sleep (SWS). In contrast, orexin neuron photoinhibition was ineffective during the night (active period). Acute photoinhibition of orexin neurons during the day in orexin/Halo mice also reduced discharge of neurons in an orexin terminal field, the dorsal raphe (DR) nucleus. However, serotonergic DR neurons exhibited normal discharge rates in mice lacking orexin neurons. Thus, although usually highly dependent on orexin neuronal activity, serotonergic DR neuronal activity can be regulated appropriately in the chronic absence of orexin input. Together, these results demonstrate that acute inhibition of orexin neurons results in time-of-day-dependent induction of SWS and in reduced firing rate of neurons in an efferent projection site thought to be involved in arousal state regulation. The results presented here advance our understanding of the role of orexin neurons in the regulation of sleep/wakefulness and may be relevant to the mechanisms that underlie symptom progression in narcolepsy.


Subject(s)
Brain Waves/physiology , Brain/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Sleep/physiology , Action Potentials/drug effects , Action Potentials/genetics , Analysis of Variance , Animals , Brain Waves/drug effects , Brain Waves/genetics , Color , Electroencephalography/methods , Electromyography , Female , Green Fluorescent Proteins/genetics , Halorhodopsins/genetics , Halorhodopsins/metabolism , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/deficiency , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Transgenic , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons/drug effects , Neuropeptides/deficiency , Orexins , Sleep/drug effects , Sodium Channel Blockers/pharmacology , Spectrum Analysis , Tetrodotoxin/pharmacology , Wakefulness/drug effects , Wakefulness/physiology
10.
Nat Commun ; 13(1): 4039, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35864121

ABSTRACT

The dorsal raphe nucleus (DRN) is known to control aggressive behavior in mice. Here, we found that glutamatergic projections from the lateral habenula (LHb) to the DRN were activated in male mice that experienced pre-exposure to a rival male mouse ("social instigation") resulting in heightened intermale aggression. Both chemogenetic and optogenetic suppression of the LHb-DRN projection blocked heightened aggression after social instigation in male mice. In contrast, inhibition of this pathway did not affect basal levels of aggressive behavior, suggesting that the activity of the LHb-DRN projection is not necessary for the expression of species-typical aggressive behavior, but required for the increase of aggressive behavior resulting from social instigation. Anatomical analysis showed that LHb neurons synapse on non-serotonergic DRN neurons that project to the ventral tegmental area (VTA), and optogenetic activation of the DRN-VTA projection increased aggressive behaviors. Our results demonstrate that the LHb glutamatergic inputs to the DRN promote aggressive arousal induced by social instigation, which contributes to aggressive behavior by activating VTA-projecting non-serotonergic DRN neurons as one of its potential targets.


Subject(s)
Dorsal Raphe Nucleus , Habenula , Aggression/physiology , Animals , Arousal , Dorsal Raphe Nucleus/physiology , Habenula/physiology , Male , Mice , Neural Pathways/physiology , Neurons/metabolism
11.
J Neurosci ; 30(38): 12642-52, 2010 Sep 22.
Article in English | MEDLINE | ID: mdl-20861370

ABSTRACT

Orexin neurons (hypocretin neurons) have a critical role in the regulation of sleep/wakefulness, especially in the maintenance of arousal. Here, we revealed that orexin neurons are directly and indirectly activated by orexin via the orexin 2 receptor (OX2R). Orexin B (1 µM) induced depolarization in orexin neurons, which was still observed in the presence of TTX (1 µM), AP-5 (50 µM), and CNQX (20 µM). In addition, orexin B induced inward currents in the presence of TTX, suggesting a direct activation of orexin neurons. Although orexin B application induced depolarization in orexin neurons of OX1R knock-out mice at comparable levels to wild-type mice, the observation that orexin B failed to depolarize orexin neurons in the OX2R knock-out mice suggested that OX2R was a primary receptor for this response. Moreover, immunoelectron microscopic analyses revealed direct contacts among orexin neurons, which exhibited structural similarities to the glutamatergic synapses. Together, these results suggest that orexin neurons form a positive-feedback circuit through indirect and direct pathways, which results in the preservation of the orexin neuron network at a high activity level and/or for a longer period. Therefore, the activation of orexin neurons through OX2R might have an important role in the maintenance of arousal.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Neurons/drug effects , Neurons/metabolism , Neuropeptides/metabolism , Neuropeptides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Analysis of Variance , Animals , Electrophysiology , Glutamic Acid/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Mice , Mice, Knockout , Orexin Receptors , Orexins , Synapses/metabolism
12.
Biology (Basel) ; 10(11)2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34827122

ABSTRACT

The majority of neurodegenerative diseases are pathologically associated with protein misfolding and aggregation. Alzheimer's disease (AD) is a type of dementia that slowly affects memory and cognitive function, and is characterized by the aggregation of the ß-amyloid protein and tau neurofibrillary tangles in the brain. Parkinson's disease (PD) is a movement disorder typically resulting in rigidity and tremor, which is pathologically linked to the aggregation of α-synuclein, particularly in dopaminergic neurons in the midbrain. Sleep disorders commonly occur in AD and PD patients, and it can precede the onset of these diseases. For example, cognitively normal older individuals who have highly fragmented sleep had a 1.5-fold increased risk of subsequently developing AD. This suggests that sleep abnormalities may be a potential biomarker of these diseases. In this review, we describe the alterations of sleep in AD and PD, and discuss their potential in the early diagnosis of these diseases. We further discuss whether sleep disturbance could be a target for the treatment of these diseases.

13.
Elife ; 92020 01 14.
Article in English | MEDLINE | ID: mdl-31934862

ABSTRACT

The brainstem plays a crucial role in sleep-wake regulation. However, the ensemble dynamics underlying sleep regulation remain poorly understood. Here, we show slow, state-predictive brainstem ensemble dynamics and state-dependent interactions between the brainstem and the hippocampus in mice. On a timescale of seconds to minutes, brainstem populations can predict pupil dilation and vigilance states and exhibit longer prediction power than hippocampal CA1 neurons. On a timescale of sub-seconds, pontine waves (P-waves) are accompanied by synchronous firing of brainstem neurons during both rapid eye movement (REM) and non-REM (NREM) sleep. Crucially, P-waves functionally interact with CA1 activity in a state-dependent manner: during NREM sleep, hippocampal sharp wave-ripples (SWRs) precede P-waves. On the other hand, P-waves during REM sleep are phase-locked with ongoing theta oscillations and are followed by burst firing of CA1 neurons. This state-dependent global coordination between the brainstem and hippocampus implicates distinct functional roles of sleep.


Subject(s)
Brain Stem/physiology , Hippocampus/physiology , Neurons/physiology , Sleep/physiology , Wakefulness/physiology , Animals , Electroencephalography , Mice , Mice, Inbred C57BL , Pons/physiology , Sleep Stages , Sleep, REM
14.
Commun Biol ; 3(1): 491, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32895482

ABSTRACT

Whilst the brain is assumed to exert homeostatic functions to keep the cellular energy status constant under physiological conditions, this has not been experimentally proven. Here, we conducted in vivo optical recordings of intracellular concentration of adenosine 5'-triphosphate (ATP), the major cellular energy metabolite, using a genetically encoded sensor in the mouse brain. We demonstrate that intracellular ATP levels in cortical excitatory neurons fluctuate in a cortex-wide manner depending on the sleep-wake states, correlating with arousal. Interestingly, ATP levels profoundly decreased during rapid eye movement sleep, suggesting a negative energy balance in neurons despite a simultaneous increase in cerebral hemodynamics for energy supply. The reduction in intracellular ATP was also observed in response to local electrical stimulation for neuronal activation, whereas the hemodynamics were simultaneously enhanced. These observations indicate that cerebral energy metabolism may not always meet neuronal energy demands, consequently resulting in physiological fluctuations of intracellular ATP levels in neurons.


Subject(s)
Adenosine Triphosphate/metabolism , Cerebral Cortex/cytology , Intracellular Space/metabolism , Neurons/physiology , Sleep/physiology , Wakefulness/physiology , Animals , Cerebrovascular Circulation/physiology , Cortical Synchronization , Cytosol/metabolism , Electric Stimulation , Mice, Inbred C57BL , Optical Imaging
15.
J Neurosci ; 28(1): 228-38, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18171940

ABSTRACT

Water homeostasis is a critical challenge to survival for land mammals. Mice display increased locomotor activity when dehydrated, a behavior that improves the likelihood of locating new sources of water and simultaneously places additional demands on compromised hydration levels. The neurophysiology underlying this well known behavior has not been previously elucidated. We report that the anti-diuretic hormone arginine-vasopressin (AVP) is involved in this response. AVP and oxytocin directly induced depolarization and an inward current in orexin/hypocretin neurons. AVP-induced activation of orexin neurons was inhibited by a V1a receptor (V1aR)-selective antagonist and was not observed in V1aR knock-out mice, suggesting an involvement of V1aR. Subsequently activation of phospholipase Cbeta triggers an increase in intracellular calcium by both calcium influx through nonselective cation channels and calcium release from calcium stores in orexin neurons. Intracerebroventricular injection of AVP or water deprivation increased locomotor activity in wild-type mice, but not in transgenic mice lacking orexin neurons. V1aR knock-out mice were less active than wild-type mice. These results suggest that the activation of orexin neurons by AVP or oxytocin has an important role in the regulation of spontaneous locomotor activity in mice. This system appears to play a key role in water deprivation-induced hyperlocomotor activity, a response to dehydration that increases the chance of locating water in nature.


Subject(s)
Antidiuretic Agents/pharmacology , Locomotion/drug effects , Neurons/physiology , Receptors, Vasopressin/physiology , Vasopressins/pharmacology , Analysis of Variance , Animals , Behavior, Animal/drug effects , Brain/cytology , Calcium/metabolism , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/metabolism , Homeostasis/drug effects , Homeostasis/physiology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Mice, Transgenic , Neurons/drug effects , Neuropeptides/genetics , Orexins , Oxytocin/pharmacology , Patch-Clamp Techniques/methods , Receptors, Vasopressin/deficiency , Tetrodotoxin/pharmacology , Water Deprivation/physiology
17.
Front Neural Circuits ; 11: 64, 2017.
Article in English | MEDLINE | ID: mdl-28959191

ABSTRACT

The basal forebrain (BF) has long been implicated in attention, learning and memory, and recent studies have established a causal relationship between artificial BF activation and arousal. However, neural ensemble dynamics in the BF still remains unclear. Here, recording neural population activity in the BF and comparing it with simultaneously recorded cortical population under both anesthetized and unanesthetized conditions, we investigate the difference in the structure of spontaneous population activity between the BF and the auditory cortex (AC) in mice. The AC neuronal population show a skewed spike rate distribution, a higher proportion of short (≤80 ms) inter-spike intervals (ISIs) and a rich repertoire of rhythmic firing across frequencies. Although the distribution of spontaneous firing rate in the BF is also skewed, a proportion of short ISIs can be explained by a Poisson model at short time scales (≤20 ms) and spike count correlations are lower compared to AC cells, with optogenetically identified cholinergic cell pairs showing exceptionally higher correlations. Furthermore, a smaller fraction of BF neurons shows spike-field entrainment across frequencies: a subset of BF neurons fire rhythmically at slow (≤6 Hz) frequencies, with varied phase preferences to ongoing field potentials, in contrast to a consistent phase preference of AC populations. Firing of these slow rhythmic BF cells is correlated to a greater degree than other rhythmic BF cell pairs. Overall, the fundamental difference in the structure of population activity between the AC and BF is their temporal coordination, in particular their operational timescales. These results suggest that BF neurons slowly modulate downstream populations whereas cortical circuits transmit signals on multiple timescales. Thus, the characterization of the neural ensemble dynamics in the BF provides further insight into the neural mechanisms, by which brain states are regulated.


Subject(s)
Action Potentials/physiology , Auditory Cortex/physiology , Basal Forebrain/physiology , Nerve Net/physiology , Neurons/physiology , Animals , Auditory Cortex/cytology , Basal Forebrain/cytology , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Electroencephalography , Female , Male , Mice , Mice, Transgenic , Optogenetics , Parvalbumins/genetics , Parvalbumins/metabolism
18.
Sci Rep ; 6: 28381, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27334849

ABSTRACT

Controlling neural circuits is a powerful approach to uncover a causal link between neural activity and behaviour. Optogenetics has been widely adopted by the neuroscience community as it offers cell-type-specific perturbation with millisecond precision. However, these studies require light delivery in complex patterns with cellular-scale resolution, while covering a large volume of tissue at depth in vivo. Here we describe a novel high-density silicon-based microscale light-emitting diode (µLED) array, consisting of up to ninety-six 25 µm-diameter µLEDs emitting at a wavelength of 450 nm with a peak irradiance of 400 mW/mm(2). A width of 100 µm, tapering to a 1 µm point, and a 40 µm thickness help minimise tissue damage during insertion. Thermal properties permit a set of optogenetic operating regimes, with ~0.5 °C average temperature increase. We demonstrate depth-dependent activation of mouse neocortical neurons in vivo, offering an inexpensive novel tool for the precise manipulation of neural activity.


Subject(s)
Neurons/physiology , Optogenetics/instrumentation , Animals , Mice , Photic Stimulation
19.
Neuropharmacology ; 85: 451-60, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24951857

ABSTRACT

Orexin neurons in the hypothalamus regulate energy homeostasis by coordinating various physiological responses. Past studies have shown the role of the orexin peptide itself; however, orexin neurons contain not only orexin but also other neurotransmitters such as glutamate and dynorphin. In this study, we examined the physiological role of orexin neurons in feeding behavior and metabolism by pharmacogenetic activation and chronic ablation. We generated novel orexin-Cre mice and utilized Cre-dependent adeno-associated virus vectors to express Gq-coupled modified GPCR, hM3Dq or diphtheria toxin fragment A in orexin neurons. By intraperitoneal injection of clozapine-N oxide in orexin-Cre mice expressing hM3Dq in orexin neurons, we could selectively manipulate the activity of orexin neurons. Pharmacogenetic stimulation of orexin neurons simultaneously increased locomotive activity, food intake, water intake and the respiratory exchange ratio (RER). Elevation of blood glucose levels and RER persisted even after locomotion and feeding behaviors returned to basal levels. Accordantly, 83% ablation of orexin neurons resulted in decreased food and water intake, while 70% ablation had almost no effect on these parameters. Our results indicate that orexin neurons play an integral role in regulation of both feeding behavior and metabolism. This regulation is so robust that greater than 80% of orexin neurons were ablated before significant changes in feeding behavior emerged.


Subject(s)
Drinking/physiology , Eating/physiology , Feeding Behavior/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Motor Activity/physiology , Neurons/physiology , Neuropeptides/metabolism , Animals , Blood Glucose , Dependovirus/genetics , Drinking Water/administration & dosage , Genetic Vectors , Male , Mice , Orexins
20.
Neurosci Res ; 75(1): 23-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22868039

ABSTRACT

Melanopsin (OPN4) is a photosensitive G-protein-coupled photopigment and its ectopic expression enables control of neural activity induced by blue light. Here we report that we successfully expressed OPN4 in hypothalamic orexin/hypocretin neurons of double-transgenic mice (orexin-tTA; Bitet-O human OPN4 [hOPN4]/mCherry mice). In the double-transgenic mice, hypothalamic orexin neurons selectively expressed hOPN4 as well as mCherry as a reporter. We conducted slice patch-clamp recordings on hOPN4/mCherry-expressing orexin neurons, which showed long-lasting activation initiated by blue light even after the light was switched off. Optical fiber-guided blue light stimulation in the hypothalamus successfully initiated the electroencephalography pattern that reflects long-lasting wakefulness in the mice in vivo. Taken together, the results indicate that ectopic expression of hOPN4 in orexin neurons enables long-lasting activation of orexin neurons by blue light to control sleep/wakefulness of the mice.


Subject(s)
Neurons/metabolism , Optogenetics/methods , Rod Opsins/genetics , Wakefulness/physiology , Animals , Brain/physiology , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Transgenic , Neuropeptides/metabolism , Orexins , Organ Culture Techniques , Patch-Clamp Techniques
SELECTION OF CITATIONS
SEARCH DETAIL