Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nature ; 612(7940): 512-518, 2022 12.
Article in English | MEDLINE | ID: mdl-36477539

ABSTRACT

Progress has been made in the elucidation of sleep and wakefulness regulation at the neurocircuit level1,2. However, the intracellular signalling pathways that regulate sleep and the neuron groups in which these intracellular mechanisms work remain largely unknown. Here, using a forward genetics approach in mice, we identify histone deacetylase 4 (HDAC4) as a sleep-regulating molecule. Haploinsufficiency of Hdac4, a substrate of salt-inducible kinase 3 (SIK3)3, increased sleep. By contrast, mice that lacked SIK3 or its upstream kinase LKB1 in neurons or with a Hdac4S245A mutation that confers resistance to phosphorylation by SIK3 showed decreased sleep. These findings indicate that LKB1-SIK3-HDAC4 constitute a signalling cascade that regulates sleep and wakefulness. We also performed targeted manipulation of SIK3 and HDAC4 in specific neurons and brain regions. This showed that SIK3 signalling in excitatory neurons located in the cerebral cortex and the hypothalamus positively regulates EEG delta power during non-rapid eye movement sleep (NREMS) and NREMS amount, respectively. A subset of transcripts biased towards synaptic functions was commonly regulated in cortical glutamatergic neurons through the expression of a gain-of-function allele of Sik3 and through sleep deprivation. These findings suggest that NREMS quantity and depth are regulated by distinct groups of excitatory neurons through common intracellular signals. This study provides a basis for linking intracellular events and circuit-level mechanisms that control NREMS.


Subject(s)
Neurons , Sleep Duration , Sleep , Wakefulness , Animals , Mice , Electroencephalography , Neurons/metabolism , Neurons/physiology , Sleep/genetics , Sleep/physiology , Sleep Deprivation/genetics , Wakefulness/genetics , Wakefulness/physiology , Signal Transduction , Delta Rhythm , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Glutamic Acid/metabolism , Sleep, Slow-Wave/genetics , Sleep, Slow-Wave/physiology
2.
Proc Natl Acad Sci U S A ; 120(11): e2218209120, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36877841

ABSTRACT

Mammals exhibit circadian cycles of sleep and wakefulness under the control of the suprachiasmatic nucleus (SCN), such as the strong arousal phase-locked to the beginning of the dark phase in laboratory mice. Here, we demonstrate that salt-inducible kinase 3 (SIK3) deficiency in gamma-aminobutyric acid (GABA)-ergic neurons or neuromedin S (NMS)-producing neurons delayed the arousal peak phase and lengthened the behavioral circadian cycle under both 12-h light:12-h dark condition (LD) and constant dark condition (DD) without changing daily sleep amounts. In contrast, the induction of a gain-of-function mutant allele of Sik3 in GABAergic neurons exhibited advanced activity onset and a shorter circadian period. Loss of SIK3 in arginine vasopressin (AVP)-producing neurons lengthened the circadian cycle, but the arousal peak phase was similar to that in control mice. Heterozygous deficiency of histone deacetylase (HDAC) 4, a SIK3 substrate, shortened the circadian cycle, whereas mice with HDAC4 S245A, which is resistant to phosphorylation by SIK3, delayed the arousal peak phase. Phase-delayed core clock gene expressions were detected in the liver of mice lacking SIK3 in GABAergic neurons. These results suggest that the SIK3-HDAC4 pathway regulates the circadian period length and the timing of arousal through NMS-positive neurons in the SCN.


Subject(s)
Arousal , Histone Deacetylases , Protein Serine-Threonine Kinases , Wakefulness , Animals , Mice , Alleles , Arginine Vasopressin , Protein Serine-Threonine Kinases/genetics , Suprachiasmatic Nucleus , Histone Deacetylases/genetics
3.
Nature ; 558(7710): 435-439, 2018 06.
Article in English | MEDLINE | ID: mdl-29899451

ABSTRACT

Sleep and wake have global effects on brain physiology, from molecular changes1-4 and neuronal activities to synaptic plasticity3-7. Sleep-wake homeostasis is maintained by the generation of a sleep need that accumulates during waking and dissipates during sleep8-11. Here we investigate the molecular basis of sleep need using quantitative phosphoproteomic analysis of the sleep-deprived and Sleepy mouse models of increased sleep need. Sleep deprivation induces cumulative phosphorylation of the brain proteome, which dissipates during sleep. Sleepy mice, owing to a gain-of-function mutation in the Sik3 gene 12 , have a constitutively high sleep need despite increased sleep amount. The brain proteome of these mice exhibits hyperphosphorylation, similar to that seen in the brain of sleep-deprived mice. Comparison of the two models identifies 80 mostly synaptic sleep-need-index phosphoproteins (SNIPPs), in which phosphorylation states closely parallel changes of sleep need. SLEEPY, the mutant SIK3 protein, preferentially associates with and phosphorylates SNIPPs. Inhibition of SIK3 activity reduces phosphorylation of SNIPPs and slow wave activity during non-rapid-eye-movement sleep, the best known measurable index of sleep need, in both Sleepy mice and sleep-deprived wild-type mice. Our results suggest that phosphorylation of SNIPPs accumulates and dissipates in relation to sleep need, and therefore SNIPP phosphorylation is a molecular signature of sleep need. Whereas waking encodes memories by potentiating synapses, sleep consolidates memories and restores synaptic homeostasis by globally downscaling excitatory synapses4-6. Thus, the phosphorylation-dephosphorylation cycle of SNIPPs may represent a major regulatory mechanism that underlies both synaptic homeostasis and sleep-wake homeostasis.


Subject(s)
Brain/metabolism , Homeostasis , Phosphoproteins/analysis , Phosphoproteins/metabolism , Proteome/analysis , Proteomics , Sleep/physiology , Animals , Brain/physiology , Gain of Function Mutation , Male , Memory Consolidation/physiology , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteome/metabolism , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology , Synapses/physiology , Wakefulness/physiology
4.
J Neurosci ; 41(12): 2733-2746, 2021 03 24.
Article in English | MEDLINE | ID: mdl-33558433

ABSTRACT

Sleep is regulated in a homeostatic manner. Sleep deprivation increases sleep need, which is compensated mainly by increased EEG δ power during non-rapid eye movement sleep (NREMS) and, to a lesser extent, by increased sleep amount. Although genetic factors determine the constitutive level of sleep need and sleep amount in mice and humans, the molecular entity behind sleep need remains unknown. Recently, we found that a gain-of-function Sleepy (Slp) mutation in the salt-inducible kinase 3 (Sik3) gene, which produces the mutant SIK3(SLP) protein, leads to an increase in NREMS EEG δ power and sleep amount. Since Sik3Slp mice express SIK3(SLP) in various types of cells in the brain as well as multiple peripheral tissues from the embryonic stage, the cell type and developmental stage responsible for the sleep phenotype in Sik3Slp mice remain to be elucidated. Here, we generated two mouse lines, synapsin1CreERT2 and Sik3ex13flox mice, which enable inducible Cre-mediated, conditional expression of SIK3(SLP) in neurons on tamoxifen administration. Administration of tamoxifen to synapsin1CreERT2 mice during late infancy resulted in higher recombination efficiency than administration during adolescence. SIK3(SLP) expression after late infancy increased NREMS and NREMS δ power in male synapsin1CreERT2; Sik3ex13flox/+ mice. The expression of SIK3(SLP) after adolescence led to a higher NREMS δ power without a significant change in NREMS amounts. Thus, neuron-specific expression of SIK3(SLP) after late infancy is sufficient to increase sleep.SIGNIFICANCE STATEMENT The propensity to accumulate sleep need during wakefulness and to dissipate it during sleep underlies the homeostatic regulation of sleep. However, little is known about the developmental stage and cell types involved in determining the homeostatic regulation of sleep. Here, we show that Sik3Slp allele induction in mature neurons in late infancy is sufficient to increase non-rapid eye movement sleep amount and non-rapid eye movement sleep δ power. SIK3 signaling in neurons constitutes an intracellular mechanism to increase sleep.


Subject(s)
Alleles , Mutation/physiology , Neurons/physiology , Protein Serine-Threonine Kinases/biosynthesis , Sleep/physiology , Wakefulness/physiology , Age Factors , Animals , Animals, Newborn , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics
5.
Nature ; 539(7629): 378-383, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27806374

ABSTRACT

Sleep is conserved from invertebrates to vertebrates, and is tightly regulated in a homeostatic manner. The molecular and cellular mechanisms that determine the amount of rapid eye movement sleep (REMS) and non-REMS (NREMS) remain unknown. Here we identify two dominant mutations that affect sleep and wakefulness by using an electroencephalogram/electromyogram-based screen of randomly mutagenized mice. A splicing mutation in the Sik3 protein kinase gene causes a profound decrease in total wake time, owing to an increase in inherent sleep need. Sleep deprivation affects phosphorylation of regulatory sites on the kinase, suggesting a role for SIK3 in the homeostatic regulation of sleep amount. Sik3 orthologues also regulate sleep in fruitflies and roundworms. A missense, gain-of-function mutation in the sodium leak channel NALCN reduces the total amount and episode duration of REMS, apparently by increasing the excitability of REMS-inhibiting neurons. Our results substantiate the use of a forward-genetics approach for studying sleep behaviours in mice, and demonstrate the role of SIK3 and NALCN in regulating the amount of NREMS and REMS, respectively.


Subject(s)
Ion Channels/genetics , Mutagenesis , Mutation , Nerve Tissue Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Sleep/genetics , Sleep/physiology , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Conserved Sequence , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Electroencephalography , Electromyography , Homeostasis/genetics , Ion Channels/chemistry , Ion Channels/metabolism , Membrane Proteins , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , RNA Splicing/genetics , Random Allocation , Sleep Deprivation , Sleep, REM/genetics , Sleep, REM/physiology , Time Factors , Wakefulness/genetics , Wakefulness/physiology
6.
Proc Natl Acad Sci U S A ; 116(32): 16062-16067, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31337678

ABSTRACT

The regulatory network of genes and molecules in sleep/wakefulness remains to be elucidated. Here we describe the methodology and workflow of the dominant screening of randomly mutagenized mice and discuss theoretical basis of forward genetics research for sleep in mice. Our high-throughput screening employs electroencephalogram (EEG) and electromyogram (EMG) to stage vigilance states into a wake, rapid eye movement sleep (REMS) and non-REM sleep (NREMS). Based on their near-identical sleep/wake behavior, C57BL/6J (B6J) and C57BL/6N (B6N) are chosen as mutagenized and counter strains, respectively. The total time spent in the wake and NREMS, as well as the REMS episode duration, shows sufficient reproducibility with small coefficients of variance, indicating that these parameters are most suitable for quantitative phenotype-driven screening. Coarse linkage analysis of the quantitative trait, combined with whole-exome sequencing, can identify the gene mutation associated with sleep abnormality. Our simulations calculate the achievable LOD score as a function of the phenotype strength and the numbers of mice examined. A pedigree showing a mild decrease in total wake time resulting from a heterozygous point mutation in the Cacna1a gene is described as an example.


Subject(s)
Genetic Testing/methods , Sleep/genetics , Wakefulness/genetics , Animals , Calcium Channels, N-Type/genetics , Computer Simulation , Crosses, Genetic , Disorders of Excessive Somnolence/genetics , Ethylnitrosourea , Female , Genes, Dominant , Homozygote , Lod Score , Male , Mice, Inbred C57BL , Mutation/genetics , Pedigree , Phenotype , Reproducibility of Results
7.
J Biol Chem ; 289(46): 31950-31959, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25278019

ABSTRACT

The lack of the neuropeptide orexin, also known as hypocretin, results in narcolepsy, a chronic sleep disorder characterized by frequent sleep/cataplexy attacks and rapid eye movement sleep abnormalities. However, the downstream pathways of orexin signaling are not clearly understood. Here, we show that orexin activates the mTOR pathway, a central regulator of cell growth and metabolism, in the mouse brain and multiple recombinant cell lines that express the G protein-coupled receptors (GPCRs), orexin 1 receptor (OX1R) or orexin 2 receptor (OX2R). This orexin/GPCR-stimulated mTOR activation is sensitive to rapamycin, an inhibitor of mTOR complex 1 (mTORC1) but is independent of two well known mTORC1 activators, Erk and Akt. Rather, our studies indicate that orexin activates mTORC1 via extracellular calcium influx and the lysosome pathway involving v-ATPase and Rag GTPases. Moreover, a cytoplasmic calcium transient is sufficient to mimic orexin/GPCR signaling to mTORC1 activation in a v-ATPase-dependent manner. Together, our studies suggest that the mTORC1 pathway functions downstream of orexin/GPCR signaling, which plays a crucial role in many physiological and metabolic processes.


Subject(s)
Brain/metabolism , Multiprotein Complexes/metabolism , Neuropeptides/metabolism , Orexin Receptors/metabolism , TOR Serine-Threonine Kinases/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Calcium/metabolism , Cell Line , Cytoplasm/metabolism , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Enzymologic , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orexins , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction
8.
Neurosci Res ; 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-38029860

ABSTRACT

Although sleep is tightly regulated by multiple neuronal circuits in the brain, nonneuronal cells such as glial cells have been increasingly recognized as crucial sleep regulators. Recent studies have shown that microglia may act to maintain wakefulness. Here, we investigated the possible involvement of microglia in the regulation of sleep quantity and quality under baseline and stress conditions through electroencephalography (EEG)/electromyography (EMG) recordings, and by employing pharmacological methods to eliminate microglial cells in the adult mouse brain. We found that severe microglial depletion induced by the colony-stimulating factor 1 receptor (CSF1R) antagonist PLX5622 (PLX) reversibly decreased the total wake time and the wake episode duration and increased the EEG slow-wave power during wakefulness under baseline conditions. To examine the role of microglia in sleep/wake regulation under mental stress, we used the acute social defeat stress (ASDS) paradigm, an ethological model for psychosocial stress. Sleep analysis under ASDS revealed that microglial depletion exacerbated the stress-induced decrease in the total wake time and increase in anxiety-like behaviors in the open field test. These results demonstrate that microglia actively modulate sleep quantity and architecture under both baseline and stress conditions. Our findings suggest that microglia may potentially provide resilience against acute psychosocial stress by regulating restorative sleep.

9.
J Neurophysiol ; 106(4): 1782-92, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21734105

ABSTRACT

Inhibitory neurons are an essential element of the locomotor network in the mammalian spinal cord. However, little is known about the firing pattern and synaptic modulation during locomotion in the majority of them. In this study, we performed whole cell recording in visually identified ventrolaterally located GABAergic neurons (VL-GNs) in the rostral (L2 segment) and caudal (L5 segment) lumbar cord using isolated spinal cord preparations taken from glutamate decarboxylase 67-green fluorescent protein (GAD67-GFP) knock-in mouse neonates. These neurons did not respond to electrical stimulation of the ventral root, indicating that they were not Renshaw cells. Ninety-five percent of VL-GNs in the L2 segment and fifty percent of those in the L5 segment showed significant rhythmic firing during locomotor-like rhythmic activity induced by bath application of 5-HT and NMDA. Seventy percent of these neurons fired mainly during the extensor phase, and twenty-five percent fired mainly during the flexor phase. Voltage-clamp recordings revealed that most of these neurons received rhythmic inhibition during the nonfiring phase and excitatory synaptic inputs during the firing phase. Morphological examination of recorded neurons filled with neurobiotin showed that their soma was located lateral to the motoneuron pool and that they extended their processes into the local ipsilateral ventromedial region and dorsal regions. The present study indicates that these GABAergic interneurons located in the ventrolateral region adjacent to the motoneuron pool are rhythmically active during locomotion and involved in the inhibitory modulation of local locomotor network in the lumbar spinal cord.


Subject(s)
Anterior Horn Cells/physiology , GABAergic Neurons/physiology , Interneurons/physiology , Locomotion/physiology , Spinal Cord/cytology , gamma-Aminobutyric Acid/physiology , Action Potentials/physiology , Animals , Animals, Newborn , Electric Stimulation , Excitatory Postsynaptic Potentials/physiology , Gene Knock-In Techniques , Genes, Reporter , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , In Vitro Techniques , Inhibitory Postsynaptic Potentials/physiology , Mice , N-Methylaspartate/pharmacology , Patch-Clamp Techniques , Serotonin/pharmacology , Spinal Nerve Roots/physiology , Synaptic Transmission/drug effects
10.
J Neurophysiol ; 103(6): 3437-47, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20410357

ABSTRACT

In the mammalian spinal cord, Renshaw cells (RCs) are excited by axon collaterals of motoneurons (MNs), and in turn, provide recurrent inhibition of MNs. They are considered an important element in controlling the motor output. However, how RCs are modulated by spinal circuits during motor behaviors remains unclear. In this study, the physiological nature of inhibitory synaptic inputs to RCs in the lumbar segment during spontaneous motoneuronal activity was examined in the isolated spinal cord taken from glutamate decarboxylase 67-green fluorescent protein (GAD67-GFP) knock-in mouse neonates. Whole cell recordings of RCs in current-clamp mode showed that they receive phasic inhibition that could modulate the RC firing evoked by excitation of MNs. In voltage-clamp recording, we observed a barrage of spontaneous inhibitory postsynaptic currents (sIPSCs) mediated by glycine and/or GABA. These sIPSCs persisted in the presence of mecamylamine, a nicotinic receptor antagonist, indicating that excitation of other RCs by MN axon collaterals may not be essential for these inhibitory actions. Simultaneous recording of RC and the ventral root in the same segment showed that the RCs received inhibitory inputs when spontaneous MN firing occurred. Paired recordings of a RC and a MN showed that during the bursting activity in the ventral root, the magnitude of the RC sIPSCs and the magnitude of the excitatory inputs that MNs receive are highly correlated. These results indicate that RCs are modulated by inhibition that matches the MN excitation in timing and amplitude during motor behaviors.


Subject(s)
Interneurons/physiology , Motor Neurons/physiology , Neural Inhibition/physiology , Spinal Cord/cytology , Synaptic Transmission/physiology , Action Potentials/physiology , Animals , Animals, Newborn , Electric Stimulation/methods , Glutamate Decarboxylase/deficiency , Green Fluorescent Proteins/genetics , In Vitro Techniques , Lumbosacral Region , Mecamylamine/pharmacology , Mice , Mice, Transgenic , Neural Pathways/physiology , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques/methods , Synaptic Transmission/drug effects , Synaptic Transmission/genetics
11.
Sci Rep ; 10(1): 8676, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32457359

ABSTRACT

Although sleep is one of the most conserved behaviors, the intracellular mechanism regulating sleep/wakefulness remains unknown. We recently identified a protein kinase, SIK3, as a sleep-regulating molecule. Mice that lack a well-conserved protein kinase A (PKA) phosphorylation site, S551, showed longer non-rapid eye movement (NREM) sleep and increased NREMS delta density. S551 of SIK3 is conserved in other members of the SIK family, such as SIK1 (S577) and SIK2 (S587). Here, we examined whether the PKA phosphorylation sites of SIK1 and SIK2 are involved in sleep regulation by generating Sik1S577A and Sik2S587A mice. The homozygous Sik1S577A mice showed a shorter wake time, longer NREMS time, and higher NREMS delta density than the wild-type mice. The heterozygous and homozygous Sik2S587A mice showed increased NREMS delta density. Both the Sik1S577A and Sik2S587A mice exhibited proper homeostatic regulation of sleep need after sleep deprivation. Despite abundant expression of Sik1 in the suprachiasmatic nucleus, the Sik1S577A mice showed normal circadian behavior. Although Sik2 is highly expressed in brown adipose tissue, the male and female Sik2S587A mice that were fed either a chow or high-fat diet showed similar weight gain as the wild-type littermates. These results suggest that PKA-SIK signaling is involved in the regulation of sleep need.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Sleep, Slow-Wave/genetics , Wakefulness/genetics , Adipose Tissue, Brown/metabolism , Amino Acid Substitution/genetics , Animals , Body Weight/genetics , Brain Waves/genetics , Cell Line , Circadian Rhythm/genetics , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Sleep, Slow-Wave/physiology , Wakefulness/physiology
12.
Genetics ; 216(3): 753-764, 2020 11.
Article in English | MEDLINE | ID: mdl-32878901

ABSTRACT

The molecular mechanism regulating sleep largely remains to be elucidated. In humans, families that carry mutations in TFAP2B, which encodes the transcription factor AP-2ß, self-reported sleep abnormalities such as short-sleep and parasomnia. Notably, AP-2 transcription factors play essential roles in sleep regulation in the nematode Caenorhabditis elegans and the fruit fly Drosophila melanogaster Thus, AP-2 transcription factors might have a conserved role in sleep regulation across the animal phyla. However, direct evidence supporting the involvement of TFAP2B in mammalian sleep was lacking. In this study, by using the CRISPR/Cas9 technology, we generated two Tfap2b mutant mouse strains, Tfap2bK144 and Tfap2bK145 , each harboring a single-nucleotide mutation within the introns of Tfap2b mimicking the mutations in two human kindreds that self-reported sleep abnormalities. The effects of these mutations were compared with those of a Tfap2b knockout allele (Tfap2b-). The protein expression level of TFAP2B in the embryonic brain was reduced to about half in Tfap2b+/- mice and was further reduced in Tfap2b-/- mice. By contrast, the protein expression level was normal in Tfap2bK145/+ mice but was reduced in Tfap2bK145/K145 mice to a similar extent as Tfap2b-/- mice. Tfap2bK144/+ and Tfap2bK144/K144 showed normal protein expression levels. Tfap2b+/- female mice showed increased wakefulness time and decreased nonrapid eye movement sleep (NREMS) time. By contrast, Tfap2bK145/+ female mice showed an apparently normal amount of sleep but instead exhibited fragmented NREMS, whereas Tfap2bK144/+ male mice showed reduced NREMS time specifically in the dark phase. Finally, in the adult brain, Tfap2b-LacZ expression was detected in the superior colliculus, locus coeruleus, cerebellum, and the nucleus of solitary tract. These findings provide direct evidence that TFAP2B influences NREMS amounts in mice and also show that different mutations in Tfap2b can lead to diverse effects on sleep architecture.


Subject(s)
Sleep Stages , Transcription Factor AP-2/genetics , Animals , Brain/embryology , Brain/metabolism , Female , Introns , Male , Mice , Point Mutation , Transcription Factor AP-2/metabolism
13.
iScience ; 20: 1-13, 2019 Oct 25.
Article in English | MEDLINE | ID: mdl-31546102

ABSTRACT

Orexins are hypothalamic neuropeptides that regulate feeding, energy expenditure, and sleep. Although orexin-deficient mice are susceptible to obesity, little is known about the roles of the orexin receptors in long-term energy metabolism. Here, we performed the metabolic characterization of orexin receptor-deficient mice. Ox1r-deficient mice were resistant to diet-induced obesity, and their food intake was similar between chow and high-fat food. Ox2r-deficient mice exhibited less energy expenditure than wild-type mice when fed a high-fat diet. Neither Ox1r-deficient nor Ox2r-deficient mice showed body weight gain similar to orexin-deficient mice. Although the presence of a running wheel suppressed diet-induced obesity in wild-type mice, the effect was weaker in orexin neuron-ablated mice. Finally, we did not detect abnormalities in brown adipose tissues of orexin-deficient mice. Thus, each orexin receptor signaling has a unique role in energy metabolism, and orexin neurons are involved in the interactive effect of diet and exercise on body weight gain.

14.
Front Neurosci ; 12: 535, 2018.
Article in English | MEDLINE | ID: mdl-30131671

ABSTRACT

Sleep/wake behavior is regulated by distinct groups of neurons, such as dopaminergic, noradrenergic, and orexinergic neurons. Although monoaminergic neurons are usually considered to be wake-promoting, the role of serotonergic neurons in sleep/wake behavior remains inconclusive because of the effect of serotonin (5-HT)-deficiency on brain development and the compensation for inborn 5-HT deficiency by other sleep/wake-regulating neurons. Here, we performed selective ablation of central 5-HT neurons in the newly developed Rosa-diphtheria toxin receptor (DTR)-tdTomato mouse line that was crossed with Pet1Cre/+ mice to examine the role of 5-HT neurons in the sleep/wake behavior of adult mice. Intracerebroventricular administration of diphtheria toxin completely ablated tdTomato-positive cells in Pet1Cre/+; Rosa-DTR-tdTomato mice. Electroencephalogram/electromyogram-based sleep/wake analysis demonstrated that central 5-HT neuron ablation in adult mice decreased the time spent in rapid eye movement (REM) sleep, which was associated with fewer transitions from non-REM (NREM) sleep to REM sleep than in control mice. Central 5-HT neuron-ablated mice showed attenuated wake response to a novel environment and increased theta power during wakefulness compared to control mice. The current findings indicated that adult 5-HT neurons work to support wakefulness and regulate REM sleep time through a biased transition from NREM sleep to REM sleep.

15.
Cell Rep ; 24(1): 79-94, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29972793

ABSTRACT

The mammalian brain undergoes sexual differentiation by gonadal hormones during the perinatal critical period. However, the machinery at earlier stages has not been well studied. We found that Ptf1a is expressed in certain neuroepithelial cells and immature neurons around the third ventricle that give rise to various neurons in several hypothalamic nuclei. We show that conditional Ptf1a-deficient mice (Ptf1a cKO) exhibit abnormalities in sex-biased behaviors and reproductive organs in both sexes. Gonadal hormone administration to gonadectomized animals revealed that the abnormal behavior is caused by disorganized sexual development of the knockout brain. Accordingly, expression of sex-biased genes was severely altered in the cKO hypothalamus. In particular, Kiss1, important for sexual differentiation of the brain, was drastically reduced in the cKO hypothalamus, which may contribute to the observed phenotypes in the Ptf1a cKO. These findings suggest that forebrain Ptf1a is one of the earliest regulators for sexual differentiation of the brain.


Subject(s)
Prosencephalon/embryology , Sex Differentiation , Transcription Factors/metabolism , Animals , Cell Lineage , Embryo, Mammalian/metabolism , Female , Gene Expression Regulation, Developmental , Gonads/abnormalities , Hypothalamus/embryology , Hypothalamus/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Sex Differentiation/genetics , Sexual Behavior, Animal , Transcription Factors/deficiency
16.
Sci Rep ; 6: 32453, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27585985

ABSTRACT

The discovery of leptin substantiated the usefulness of a forward genetic approach in elucidating the molecular network regulating energy metabolism. However, no successful dominant screening for obesity has been reported, which may be due to the influence of quantitative trait loci between the screening and counter strains and the low fertility of obese mice. Here, we performed a dominant screening for obesity using C57BL/6 substrains, C57BL/6J and C57BL/6N, with the routine use of in vitro fertilization. The screening of more than 5000 mutagenized mice established two obese pedigrees in which single nucleotide substitutions in Mc4r and Sim1 genes were identified through whole-exome sequencing. The mutation in the Mc4r gene produces a premature stop codon, and the mutant SIM1 protein lacks transcriptional activity, showing that the haploinsufficiency of SIM1 and MC4R results in obesity. We further examined the hypothalamic neuropeptide expressions in the mutant pedigrees and mice with diet-induced obesity, which showed that each obesity mouse model has distinct neuropeptide expression profiles. This forward genetic screening scheme is useful and applicable to any research field in which mouse models work.


Subject(s)
Genes, Dominant , Genetic Predisposition to Disease , Genetic Testing , Mutation/genetics , Obesity/genetics , Amino Acid Sequence , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Chromosome Mapping , Diet , Disease Models, Animal , Female , Gene Expression Regulation , Hypothalamus/metabolism , Luciferases/metabolism , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Neuropeptides/genetics , Neuropeptides/metabolism , Obesity/metabolism , Obesity/pathology , Pedigree , Phenotype , Polymorphism, Single Nucleotide/genetics , Receptor, Melanocortin, Type 4/genetics , Repressor Proteins/chemistry , Repressor Proteins/genetics , Exome Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL