Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Transl Med ; 21(1): 258, 2023 04 15.
Article in English | MEDLINE | ID: mdl-37061718

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is characterized by a progressive loss of memory that cannot be efficiently managed by currently available AD therapeutics. So far, most treatments for AD that have the potential to improve memory target neural circuits to protect their integrity. However, the vulnerable neural circuits and their dynamic remodeling during AD progression remain largely undefined. METHODS: Circuit-based approaches, including anterograde and retrograde tracing, slice electrophysiology, and fiber photometry, were used to investigate the dynamic structural and functional remodeling of a GABAergic circuit projected from the medial septum (MS) to the dentate gyrus (DG) in 3xTg-AD mice during AD progression. RESULTS: We identified a long-distance GABAergic circuit that couples highly connected MS and DG GABAergic neurons during spatial memory encoding. Furthermore, we found hyperactivity of DG interneurons during early AD, which persisted into late AD stages. Interestingly, MS GABAergic projections developed a series of adaptive strategies to combat DG interneuron hyperactivity. During early-stage AD, MS-DG GABAergic projections exhibit increased inhibitory synaptic strength onto DG interneurons to inhibit their activities. During late-stage AD, MS-DG GABAergic projections form higher anatomical connectivity with DG interneurons and exhibit aberrant outgrowth to increase the inhibition onto DG interneurons. CONCLUSION: We report the structural and functional remodeling of the MS-DG GABAergic circuit during disease progression in 3xTg-AD mice. Dynamic MS-DG GABAergic circuit remodeling represents a compensatory mechanism to combat DG interneuron hyperactivity induced by reduced GABA transmission.


Subject(s)
Alzheimer Disease , Mice , Animals , Mice, Transgenic , Hippocampus
2.
Mol Psychiatry ; 26(7): 2912-2928, 2021 07.
Article in English | MEDLINE | ID: mdl-33057171

ABSTRACT

The ventral pallidum (VP) regulates motivation, drug addiction, and several behaviors that rely on heightened arousal. However, the role and underlying neural circuits of the VP in the control of wakefulness remain poorly understood. In the present study, we sought to elucidate the specific role of VP GABAergic neurons in controlling sleep-wake behaviors in mice. Fiber photometry revealed that the population activity of VP GABAergic neurons was increased during physiological transitions from non-rapid eye movement (non-REM, NREM) sleep to either wakefulness or REM sleep. Moreover, chemogenetic and optogenetic manipulations were leveraged to investigate a potential causal role of VP GABAergic neurons in initiating and/or maintaining arousal. In vivo optogenetic stimulation of VP GABAergic neurons innervating the ventral tegmental area (VTA) strongly promoted arousal via disinhibition of VTA dopaminergic neurons. Functional in vitro mapping revealed that VP GABAergic neurons, in principle, inhibited VTA GABAergic neurons but also inhibited VTA dopaminergic neurons. In addition, optogenetic stimulation of terminals of VP GABAergic neurons revealed that they promoted arousal by innervating the lateral hypothalamus, but not the mediodorsal thalamus or lateral habenula. The increased wakefulness chemogenetically evoked by VP GABAergic neuronal activation was completely abolished by pretreatment with dopaminergic D1 and D2/D3 receptor antagonists. Furthermore, activation of VP GABAergic neurons increased exploration time in both the open-field and light-dark box tests but did not modulate depression-like behaviors or food intake. Finally, chemogenetic inhibition of VP GABAergic neurons decreased arousal. Taken together, our findings indicate that VP GABAergic neurons are essential for arousal related to motivation.


Subject(s)
Basal Forebrain , Wakefulness , Animals , GABAergic Neurons , Mice , Motivation , Ventral Tegmental Area
3.
PLoS Biol ; 16(4): e2002909, 2018 04.
Article in English | MEDLINE | ID: mdl-29652889

ABSTRACT

The rostromedial tegmental nucleus (RMTg), also called the GABAergic tail of the ventral tegmental area, projects to the midbrain dopaminergic system, dorsal raphe nucleus, locus coeruleus, and other regions. Whether the RMTg is involved in sleep-wake regulation is unknown. In the present study, pharmacogenetic activation of rat RMTg neurons promoted non-rapid eye movement (NREM) sleep with increased slow-wave activity (SWA). Conversely, rats after neurotoxic lesions of 8 or 16 days showed decreased NREM sleep with reduced SWA at lights on. The reduced SWA persisted at least 25 days after lesions. Similarly, pharmacological and pharmacogenetic inactivation of rat RMTg neurons decreased NREM sleep. Electrophysiological experiments combined with optogenetics showed a direct inhibitory connection between the terminals of RMTg neurons and midbrain dopaminergic neurons. The bidirectional effects of the RMTg on the sleep-wake cycle were mimicked by the modulation of ventral tegmental area (VTA)/substantia nigra compacta (SNc) dopaminergic neuronal activity using a pharmacogenetic approach. Furthermore, during the 2-hour recovery period following 6-hour sleep deprivation, the amount of NREM sleep in both the lesion and control rats was significantly increased compared with baseline levels; however, only the control rats showed a significant increase in SWA compared with baseline levels. Collectively, our findings reveal an essential role of the RMTg in the promotion of NREM sleep and homeostatic regulation.


Subject(s)
Eye Movements/physiology , Neural Pathways/physiology , Receptors, Muscarinic/genetics , Sleep/physiology , Ventral Tegmental Area/physiology , Animals , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Clozapine/analogs & derivatives , Clozapine/pharmacology , Dopamine/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Dorsal Raphe Nucleus/anatomy & histology , Dorsal Raphe Nucleus/drug effects , Dorsal Raphe Nucleus/physiology , Electrodes, Implanted , Electroencephalography , Genes, Reporter , Ibotenic Acid/toxicity , Locus Coeruleus/anatomy & histology , Locus Coeruleus/drug effects , Locus Coeruleus/physiology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mesencephalon/anatomy & histology , Mesencephalon/drug effects , Mesencephalon/physiology , Neural Pathways/anatomy & histology , Neural Pathways/drug effects , Optogenetics , Pars Compacta/anatomy & histology , Pars Compacta/drug effects , Pars Compacta/physiology , Rats , Rats, Sprague-Dawley , Receptors, Muscarinic/metabolism , Sleep Deprivation/physiopathology , Stereotaxic Techniques , Ventral Tegmental Area/anatomy & histology , Ventral Tegmental Area/drug effects , Wakefulness/physiology , gamma-Aminobutyric Acid/metabolism , Red Fluorescent Protein
4.
J Neurochem ; 137(2): 277-86, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26811198

ABSTRACT

Histamine, an important modulator of the arousal states of the central nervous system, has been reported to contribute an excitatory drive at the hypoglossal motor nucleus to the genioglossus (GG) muscle, which is involved in the pathogenesis of obstructive sleep apnea. However, the effect of histamine on hypoglossal motoneurons (HMNs) and the underlying signaling mechanisms have remained elusive. Here, whole-cell patch-clamp recordings were conducted using neonatal rat brain sections, which showed that histamine excited HMNs with an inward current under voltage-clamp and a depolarization membrane potential under current-clamp via histamine H1 receptors (H1Rs). The phospholipase C inhibitor U-73122 blocked H1Rs-mediated excitatory effects, but protein kinase A inhibitor and protein kinase C inhibitor did not, indicating that the signal transduction cascades underlying the excitatory action of histamine on HMNs were H1R/Gq/11 /phospholipase C/inositol-1,4,5-trisphosphate (IP3). The effects of histamine were also dependent on extracellular Na(+) and intracellular Ca(2+), which took place via activation of Na(+)-Ca(2+) exchangers. These results identify the signaling molecules associated with the regulatory effect of histamine on HMNs. The findings of this study may provide new insights into therapeutic approaches in obstructive sleep apnea. We proposed the post-synaptic mechanisms underlying the modulation effect of histamine on hypoglossal motoneuron. Histamine activates the H1Rs via PLC and IP3, increases Ca(2+) releases from intracellular stores, promotes Na(+) influx and Ca(2+) efflux via the NCXs, and then produces an inward current and depolarizes the neurons. Histamine modulates the excitability of HMNs with other neuromodulators, such as noradrenaline, serotonin and orexin. We think that these findings should provide an important new direction for drug development for the treatment of obstructive sleep apnea.


Subject(s)
Action Potentials/drug effects , Histamine Agents/pharmacology , Histamine/pharmacology , Motor Neurons/drug effects , Signal Transduction/drug effects , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , In Vitro Techniques , Medulla Oblongata/cytology , Patch-Clamp Techniques , Pyrrolidinones/pharmacology , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
5.
J Pharmacol Exp Ther ; 356(1): 64-73, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26491061

ABSTRACT

Paeoniflorin (PF, C23H28O11), one of the principal active ingredients of Paeonia Radix, exerts depressant effects on the central nervous system. We determined whether PF could modulate sleep behaviors and the mechanisms involved. Electroencephalogram and electromyogram recordings in mice showed that intraperitoneal PF administered at a dose of 25 or 50 mg/kg significantly shortened the sleep latency and increased the amount of non-rapid eye movement (NREM). Immunohistochemical study revealed that PF decreased c-fos expression in the histaminergic tuberomammillary nucleus (TMN). The sleep-promoting effects and changes in c-fos induced by PF were reversed by 8-cyclopentyl-1,3-dimethylxanthine (CPT), an adenosine A1 receptor antagonist, and PF-induced sleep was not observed in adenosine A1 receptor knockout mice. Whole-cell patch clamping in mouse brain slices showed that PF significantly decreased the firing frequency of histaminergic neurons in TMN, which could be completely blocked by CPT. These results indicate that PF increased NREM sleep by inhibiting the histaminergic system via A1 receptors.


Subject(s)
Central Nervous System Depressants/pharmacology , Glucosides/pharmacology , Monoterpenes/pharmacology , Receptor, Adenosine A1/drug effects , Sleep/drug effects , Animals , Electroencephalography/drug effects , Electromyography/drug effects , Gene Expression/drug effects , Genes, fos/drug effects , Glucosides/antagonists & inhibitors , Histamine/physiology , Injections, Intraperitoneal , Male , Mammillary Bodies/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Monoterpenes/antagonists & inhibitors , Neurons/drug effects , Paeonia/chemistry , Patch-Clamp Techniques , Receptor, Adenosine A1/genetics , Theophylline/analogs & derivatives , Theophylline/pharmacology
6.
Acta Pharmacol Sin ; 37(10): 1325-1336, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27498778

ABSTRACT

AIM: Ethanol, one of the most frequently used and abused substances in our society, has a profound impact on sedation. However, the neuronal mechanisms underlying its sedative effect remain unclear. In this study, we investigated the effects of ethanol on histaminergic neurons in the tuberomammillary nucleus (TMN), a brain region thought to be critical for wakefulness. METHODS: Coronal brain slices (250 µm thick) containing the TMN were prepared from GAD67-GFP knock-in mice. GAD67-GFP was used to identify histaminergic neurons in the TMN. The spontaneous firing and membrane potential of histaminergic neurons, and GABAergic transmission onto these neurons were recorded using whole-cell patch-clamp recordings. Drugs were applied through superfusion. RESULTS: Histaminergic and GAD67-expressing neurons in the TMN of GAD67-GFP mice were highly co-localized. TMN GFP-positive neurons exhibited a regular spontaneous discharge at a rate of 2-4 Hz without burst firing. Brief superfusion of ethanol (64, 190, and 560 mmol/L) dose-dependently and reversibly suppressed the spontaneous firing of the neurons in the TMN; when synaptic transmission was blocked by tetrodotoxin (1 µmol/L), ethanol caused hyperpolarization of the membrane potential. Furthermore, superfusion of ethanol markedly increased the frequency and amplitude of spontaneous and miniature inhibitory postsynaptic currents (sIPSCs and mIPSCs), which were abolished in the presence of the GABAA receptor antagonist bicuculline (20 µmol/L). Finally, ethanol-mediated enhancement of sIPSCs and mIPSCs was significantly attenuated when the slices were pretreated with the GABAB agonist baclofen (30 µmol/L). CONCLUSION: Ethanol inhibits the excitability of histaminergic neurons in mouse TMN slices, possibly via potentiating GABAergic transmission onto the neurons at both pre- and postsynaptic sites.


Subject(s)
Ethanol/pharmacology , Histamine/metabolism , Hypothalamic Area, Lateral/drug effects , Receptors, GABA-A/metabolism , Receptors, GABA-B/metabolism , Animals , Baclofen/pharmacology , GABA Agonists/pharmacology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , In Vitro Techniques , Male , Membrane Potentials/drug effects , Mice , Miniature Postsynaptic Potentials/drug effects , Patch-Clamp Techniques
7.
Acta Pharmacol Sin ; 36(11): 1308-17, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26388157

ABSTRACT

AIM: Gelsemine, an alkaloid from the Chinese herb Gelsemium elegans (Gardn & Champ) Benth., is effective in mitigating chronic pain in rats. In the present study we investigated whether the alkaloid improved sleep disturbance, the most common comorbid symptoms of chronic pain, in a mouse model of neuropathic pain. METHODS: Mice were subjected to partial sciatic nerve ligation (PSNL). After the mice were injected with gelsemine or pregabalin (the positive control) intraperitoneally, mechanical allodynia and thermal hyperalgesia were assessed, and electroencephalogram (EEG)/electromyogram (EMG) recording was performed. Motor performance of the mice was assessed using rota-rod test. c-Fos expression in the brain was analyzed with immunohistochemical staining. RESULTS: In PSNL mice, gelsemine (2 and 4 mg/kg) increased the mechanical threshold for 4 h and prolonged the thermal latencies for 3 h. Furthermore, gelsemine (4 mg/kg, administered at 6:30 AM) increased non-rapid eye movement (non-REM, NREM) sleep, decreased wakefulness, but did not affect REM sleep during the first 3 h in PSNL mice. Sleep architecture analysis showed that gelsemine decreased the mean duration of wakefulness and increased the total number of episodes of NREM sleep during the first 3 h after the dosing. Gelsemine (4 mg/kg) did not impair motor coordination in PSNL mice. Immunohistochemical study showed that PSNL increased c-Fos expression in the neurons of the anterior cingulate cortex, and gelsemine (4 mg/kg) decreased c-Fos expression by 58%. Gelsemine (4 mg/kg, administered at either 6:30 AM or 8:30 PM) did not produce hypnotic effect in normal mice. Pregabalin produced similar antinociceptive and hypnotic effects, but impaired motor coordination in PSNL mice. CONCLUSION: Gelsemine is an effective agent for treatment of both neuropathic pain and sleep disturbance in PSNL mice; anterior cingulate cortex might play a role in the hypnotic effects of gelsemine.


Subject(s)
Alkaloids/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Hyperalgesia/drug therapy , Hypnotics and Sedatives/therapeutic use , Neuralgia/drug therapy , Sleep Wake Disorders/drug therapy , Alkaloids/chemistry , Animals , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , Gelsemium/chemistry , Male , Mice , Mice, Inbred C57BL , Sciatic Nerve/surgery , Sleep/drug effects
8.
Cell Rep ; 43(5): 114140, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38656873

ABSTRACT

Women are more vulnerable to stress and have a higher likelihood of developing mood disorders. The serotonin (5HT) system has been highly implicated in stress response and mood regulation. However, sex-dependent mechanisms underlying serotonergic regulation of stress vulnerability remain poorly understood. Here, we report that adult hippocampal neural stem cells (NSCs) of the Ascl1 lineage (Ascl1-NSCs) in female mice express functional 5HT1A receptors (5HT1ARs), and selective deletion of 5HT1ARs in Ascl1-NSCs decreases the Ascl1-NSC pool only in females. Mechanistically, 5HT1AR deletion in Ascl1-NSCs of females leads to 5HT-induced depolarization mediated by upregulation of 5HT7Rs. Furthermore, repeated restraint stress (RRS) impairs Ascl1-NSC maintenance through a 5HT1AR-mediated mechanism. By contrast, Ascl1-NSCs in males express 5HT7R receptors (5HT7Rs) that are downregulated by RRS, thus maintaining the Ascl1-NSC pool. These findings suggest that sex-specific expression of distinct 5HTRs and their differential interactions with stress may underlie sex differences in stress vulnerability.


Subject(s)
Hippocampus , Neural Stem Cells , Receptors, Serotonin , Stress, Psychological , Animals , Neural Stem Cells/metabolism , Female , Hippocampus/metabolism , Male , Mice , Receptors, Serotonin/metabolism , Receptors, Serotonin/genetics , Stress, Psychological/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1A/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Sex Characteristics , Mice, Inbred C57BL , Serotonin/metabolism
9.
Neuron ; 112(8): 1328-1341.e4, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38354737

ABSTRACT

Chronic pain often leads to the development of sleep disturbances. However, the precise neural circuit mechanisms responsible for sleep disorders in chronic pain have remained largely unknown. Here, we present compelling evidence that hyperactivity of pyramidal neurons (PNs) in the anterior cingulate cortex (ACC) drives insomnia in a mouse model of nerve-injury-induced chronic pain. After nerve injury, ACC PNs displayed spontaneous hyperactivity selectively in periods of insomnia. We then show that ACC PNs were both necessary for developing chronic-pain-induced insomnia and sufficient to mimic sleep loss in naive mice. Importantly, combining optogenetics and electrophysiological recordings, we found that the ACC projection to the dorsal medial striatum (DMS) underlies chronic-pain-induced insomnia through enhanced activity and plasticity of ACC-DMS dopamine D1R neuron synapses. Our findings shed light on the pivotal role of ACC PNs in developing chronic-pain-induced sleep disorders.


Subject(s)
Chronic Pain , Sleep Initiation and Maintenance Disorders , Mice , Animals , Gyrus Cinguli/physiology , Pyramidal Cells
10.
Curr Opin Neurobiol ; 79: 102693, 2023 04.
Article in English | MEDLINE | ID: mdl-36822141

ABSTRACT

Adult hippocampal neurogenesis (AHN) plays a key role in modulating memory and emotion processing. A fundamental question remains on how to effectively modulate AHN to improve hippocampal function. Here, we review recent work on how distinct aspects of hippocampal neurogenesis, including the number, maturation state, and activity of adult-born neurons (ABNs), contribute to overall hippocampal function. We propose multi-level enhancement of hippocampal neurogenesis with the combination of increased number, elevated activity, and enhanced maturation of ABNs as a potential strategy to optimize overall hippocampal performance. In addition, integration of ABNs induces significant remodeling of the local hippocampal circuits, which may in turn modulates brain-wide network dynamics. We discuss recent progress on how integration of ABNs contributes to local hippocampal circuit and brain-wide network dynamics during behavior.


Subject(s)
Hippocampus , Neurogenesis , Neurogenesis/physiology , Hippocampus/physiology , Neurons/physiology , Emotions , Cognition
11.
Cell Stem Cell ; 30(4): 415-432.e6, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37028406

ABSTRACT

Patients with Alzheimer's disease (AD) exhibit progressive memory loss, depression, and anxiety, accompanied by impaired adult hippocampal neurogenesis (AHN). Whether AHN can be enhanced in impaired AD brain to restore cognitive and affective function remains elusive. Here, we report that patterned optogenetic stimulation of the hypothalamic supramammillary nucleus (SuM) enhances AHN in two distinct AD mouse models, 5×FAD and 3×Tg-AD. Strikingly, the chemogenetic activation of SuM-enhanced adult-born neurons (ABNs) rescues memory and emotion deficits in these AD mice. By contrast, SuM stimulation alone or activation of ABNs without SuM modification fails to restore behavioral deficits. Furthermore, quantitative phosphoproteomics analyses reveal activation of the canonical pathways related to synaptic plasticity and microglia phagocytosis of plaques following acute chemogenetic activation of SuM-enhanced (vs. control) ABNs. Our study establishes the activity-dependent contribution of SuM-enhanced ABNs in modulating AD-related deficits and informs signaling mechanisms mediated by the activation of SuM-enhanced ABNs.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/metabolism , Neurons/metabolism , Hippocampus , Brain , Cognition , Disease Models, Animal , Mice, Transgenic , Neurogenesis/physiology
12.
iScience ; 26(8): 107385, 2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37609631

ABSTRACT

Insomnia is often comorbid with depression, but the underlying neuronal circuit mechanism remains elusive. Recently, we reported that GABAergic ventral pallidum (VP) neurons control wakefulness associated with motivation. However, whether and how other subtypes of VP neurons regulate arousal and emotion are largely unknown. Here, we report glutamatergic VP (VPVglut2) neurons control wakefulness and depressive-like behaviors. Physiologically, the calcium activity of VPVglut2 neurons was increased during both NREM sleep-to-wake transitions and depressive/anxiety-like behaviors in mice. Functionally, activation of VPVglut2 neurons was sufficient to increase wakefulness and induce anxiety/depressive-like behaviors, whereas inhibition attenuated both. Dissection of the circuit revealed that separated projections of VPVglut2 neurons to the lateral hypothalamus and lateral habenula promote arousal and depressive-like behaviors, respectively. Our results demonstrate a subtype of VP neurons is responsible for wakefulness and emotion through separated projections, and may provide new lines for the intervention of insomnia and depression in patients.

13.
medRxiv ; 2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37961307

ABSTRACT

Current amyloid beta-targeting approaches for Alzheimer's disease (AD) therapeutics only slow cognitive decline for small numbers of patients. This limited efficacy exists because AD is a multifactorial disease whose pathological mechanism(s) and diagnostic biomarkers are largely unknown. Here we report a new mechanism of AD pathogenesis in which the histone methyltransferase G9a noncanonically regulates translation of a hippocampal proteome that defines the proteopathic nature of AD. Accordingly, we developed a novel brain-penetrant inhibitor of G9a, MS1262, across the blood-brain barrier to block this G9a-regulated, proteopathologic mechanism. Intermittent MS1262 treatment of multiple AD mouse models consistently restored both cognitive and noncognitive functions to healthy levels. Comparison of proteomic/phosphoproteomic analyses of MS1262-treated AD mice with human AD patient data identified multiple pathological brain pathways that elaborate amyloid beta and neurofibrillary tangles as well as blood coagulation, from which biomarkers of early stage of AD including SMOC1 were found to be affected by MS1262 treatment. Notably, these results indicated that MS1262 treatment may reduce or avoid the risk of blood clot burst for brain bleeding or a stroke. This mouse-to-human conservation of G9a-translated AD proteopathology suggests that the global, multifaceted effects of MS1262 in mice could extend to relieve all symptoms of AD patients with minimum side effect. In addition, our mechanistically derived biomarkers can be used for stage-specific AD diagnosis and companion diagnosis of individualized drug effects. One-Sentence Summary: A brain-penetrant inhibitor of G9a methylase blocks G9a translational mechanism to reverse Alzheimer's disease related proteome for effective therapy.

14.
Nat Neurosci ; 25(5): 630-645, 2022 05.
Article in English | MEDLINE | ID: mdl-35524139

ABSTRACT

Adult hippocampal neurogenesis plays a critical role in memory and emotion processing, and this process is dynamically regulated by neural circuit activity. However, it remains unknown whether manipulation of neural circuit activity can achieve sufficient neurogenic effects to modulate behavior. Here we report that chronic patterned optogenetic stimulation of supramammillary nucleus (SuM) neurons in the mouse hypothalamus robustly promotes neurogenesis at multiple stages, leading to increased production of neural stem cells and behaviorally relevant adult-born neurons (ABNs) with enhanced maturity. Functionally, selective manipulation of the activity of these SuM-promoted ABNs modulates memory retrieval and anxiety-like behaviors. Furthermore, we show that SuM neurons are highly responsive to environmental novelty (EN) and are required for EN-induced enhancement of neurogenesis. Moreover, SuM is required for ABN activity-dependent behavioral modulation under a novel environment. Our study identifies a key hypothalamic circuit that couples novelty signals to the production and maturation of ABNs, and highlights the activity-dependent contribution of circuit-modified ABNs in behavioral regulation.


Subject(s)
Hippocampus , Neurogenesis , Animals , Anxiety , Hippocampus/physiology , Hypothalamus , Memory/physiology , Mice , Mice, Inbred C57BL , Neurogenesis/physiology
15.
Pain ; 161(2): 288-299, 2020 02.
Article in English | MEDLINE | ID: mdl-31651580

ABSTRACT

Patients with chronic pain often report being sensitive to pain at night before falling asleep, a time when the synchronization of cortical activity is initiated. However, how cortical activity relates to pain sensitivity is still unclear. Because sleep is characterized by enhanced cortical delta power, we hypothesized that enhanced cortical delta power may be an indicator of intensified pain. To test this hypothesis, we used pain thresholds tests, EEG/electromyogram recordings, c-Fos staining, and chemogenetic and pharmacological techniques in mice. We found that sleep deprivation or pharmacologic enhancement of EEG delta power by reserpine and scopolamine dramatically decreased mechanical pain thresholds, but not thermal withdrawal latency, in a partial sciatic nerve ligation model of neuropathic pain mice. On the contrary, suppression of EEG delta power using a wake-promoting agent modafinil significantly attenuated mechanical allodynia. Moreover, when EEG delta power was enhanced, c-Fos expression decreased in most regions of the cortex, except the anterior cingulate cortex (ACC), where c-Fos was increased in the somatostatin- and parvalbumin-positive GABAergic neurons. Chemogenetic activation of GABAergic neurons in ACC enhanced EEG delta power and lowered mechanical pain thresholds simultaneously in naive mice. However, chemogenetic inhibition of ACC GABAergic neurons could not block mechanical allodynia. These results provided compelling evidence that elevated EEG delta power is accompanied with aggravated neuropathic pain, whereas decreased delta power attenuated it, suggesting that enhanced delta power can be a specific marker of rising chronic neuropathic pain and that wake-promoting compounds could be used as analgesics in the clinic.


Subject(s)
Cerebral Cortex/physiopathology , Delta Rhythm/physiology , Hyperalgesia/physiopathology , Neuralgia/physiopathology , Pain Threshold/physiology , Sleep/physiology , Adrenergic Uptake Inhibitors/pharmacology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cholinergic Antagonists/pharmacology , Cortical Synchronization/drug effects , Cortical Synchronization/physiology , Delta Rhythm/drug effects , Electroencephalography , Electromyography , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Gyrus Cinguli/drug effects , Gyrus Cinguli/metabolism , Hyperalgesia/metabolism , Mice , Modafinil/pharmacology , Neuralgia/metabolism , Pain Threshold/drug effects , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Reserpine/pharmacology , Sciatic Nerve/surgery , Scopolamine/pharmacology , Sleep/drug effects , Sleep Deprivation/chemically induced , Sleep Deprivation/physiopathology , Wakefulness-Promoting Agents/pharmacology
16.
Neuron ; 108(2): 349-366.e6, 2020 10 28.
Article in English | MEDLINE | ID: mdl-32877641

ABSTRACT

Neural stem cells (NSCs) in the dentate gyrus (DG) reside in a specialized local niche that supports their neurogenic proliferation to produce adult-born neurons throughout life. How local niche cells interact at the circuit level to ensure continuous neurogenesis from NSCs remains unknown. Here we report the role of endogenous neuropeptide cholecystokinin (CCK), released from dentate CCK interneurons, in regulating neurogenic niche cells and NSCs. Specifically, stimulating CCK release supports neurogenic proliferation of NSCs through a dominant astrocyte-mediated glutamatergic signaling cascade. In contrast, reducing dentate CCK induces reactive astrocytes, which correlates with decreased neurogenic proliferation of NSCs and upregulation of genes involved in immune processes. Our findings provide novel circuit-based information on how CCK acts on local astrocytes to regulate the key behavior of adult NSCs.


Subject(s)
Astrocytes/physiology , Cholecystokinin/physiology , Dentate Gyrus/physiology , Interneurons/physiology , Neural Stem Cells/physiology , Neurogenesis , Neuropeptides/physiology , Animals , Female , Male , Membrane Potentials , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction
17.
Nat Commun ; 9(1): 1576, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29679009

ABSTRACT

Nucleus accumbens (NAc) is involved in behaviors that depend on heightened wakefulness, but its impact on arousal remains unclear. Here, we demonstrate that NAc dopamine D1 receptor (D1R)-expressing neurons are essential for behavioral arousal. Using in vivo fiber photometry in mice, we find arousal-dependent increases in population activity of NAc D1R neurons. Optogenetic activation of NAc D1R neurons induces immediate transitions from non-rapid eye movement sleep to wakefulness, and chemogenetic stimulation prolongs arousal, with decreased food intake. Patch-clamp, tracing, immunohistochemistry, and electron microscopy reveal that NAc D1R neurons project to the midbrain and lateral hypothalamus, and might disinhibit midbrain dopamine neurons and lateral hypothalamus orexin neurons. Photoactivation of terminals in the midbrain and lateral hypothalamus is sufficient to induce wakefulness. Silencing of NAc D1R neurons suppresses arousal, with increased nest-building behaviors. Collectively, our data indicate that NAc D1R neuron circuits are essential for the induction and maintenance of wakefulness.


Subject(s)
Dopaminergic Neurons/metabolism , Hypothalamic Area, Lateral/physiology , Mesencephalon/physiology , Nucleus Accumbens/metabolism , Receptors, Dopamine D1/metabolism , Wakefulness/physiology , Animals , Circadian Rhythm/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Patch-Clamp Techniques , Photometry/methods , Receptors, Dopamine D1/biosynthesis , Sleep/physiology
18.
Nat Commun ; 8(1): 734, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28963505

ABSTRACT

Sleep control is ascribed to a two-process model, a widely accepted concept that posits homoeostatic drive and a circadian process as the major sleep-regulating factors. Cognitive and emotional factors also influence sleep-wake behaviour; however, the precise circuit mechanisms underlying their effects on sleep control are unknown. Previous studies suggest that adenosine has a role affecting behavioural arousal in the nucleus accumbens (NAc), a brain area critical for reinforcement and reward. Here, we show that chemogenetic or optogenetic activation of excitatory adenosine A2A receptor-expressing indirect pathway neurons in the core region of the NAc strongly induces slow-wave sleep. Chemogenetic inhibition of the NAc indirect pathway neurons prevents the sleep induction, but does not affect the homoeostatic sleep rebound. In addition, motivational stimuli inhibit the activity of ventral pallidum-projecting NAc indirect pathway neurons and suppress sleep. Our findings reveal a prominent contribution of this indirect pathway to sleep control associated with motivation.In addition to circadian and homoeostatic drives, motivational levels influence sleep-wake cycles. Here the authors demonstrate that adenosine receptor-expressing neurons in the nucleus accumbens core that project to the ventral pallidum are inhibited by motivational stimuli and are causally involved in the control of slow-wave sleep.


Subject(s)
Nucleus Accumbens/physiology , Sleep/physiology , Animals , Circadian Rhythm , Female , Male , Mice , Mice, Inbred C57BL , Motivation , Patch-Clamp Techniques , Proto-Oncogene Proteins c-fos/metabolism , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2A/physiology
19.
Neuropsychopharmacology ; 41(8): 2133-46, 2016 07.
Article in English | MEDLINE | ID: mdl-26797244

ABSTRACT

The basal forebrain (BF) cholinergic neurons have long been thought to be involved in behavioral wakefulness and cortical activation. However, owing to the heterogeneity of BF neurons and poor selectivity of traditional methods, the precise role of BF cholinergic neurons in regulating the sleep-wake cycle remains unclear. We investigated the effects of cell-selective manipulation of BF cholinergic neurons on the sleep-wake behavior and electroencephalogram (EEG) power spectrum using the pharmacogenetic technique, the 'designer receptors exclusively activated by designer drugs (DREADD)' approach, and ChAT-IRES-Cre mice. Our results showed that activation of BF cholinergic neurons expressing hM3Dq receptors significantly and lastingly decreased the EEG delta power spectrum, produced low-delta non-rapid eye movement sleep, and slightly increased wakefulness in both light and dark phases, whereas inhibition of BF cholinergic neurons expressing hM4Di receptors significantly increased EEG delta power spectrum and slightly decreased wakefulness. Next, the projections of BF cholinergic neurons were traced by humanized Renilla green fluorescent protein (hrGFP). Abundant and highly dense hrGFP-positive fibers were observed in the secondary motor cortex and cingulate cortex, and sparse hrGFP-positive fibers were observed in the ventrolateral preoptic nucleus, a known sleep-related structure. Finally, we found that activation of BF cholinergic neurons significantly increased c-Fos expression in the secondary motor cortex and cingulate cortex, but decreased c-Fos expression in the ventrolateral preoptic nucleus. Taken together, these findings reveal that the primary function of BF cholinergic neurons is to inhibit EEG delta activity through the activation of cerebral cortex, rather than to induce behavioral wakefulness.


Subject(s)
Basal Forebrain/physiology , Cholinergic Neurons/physiology , Delta Rhythm , Sleep , Wakefulness , Animals , Basal Forebrain/cytology , Cerebral Cortex/cytology , Cholinergic Neurons/cytology , Electroencephalography , Male , Mice , Mice, Transgenic , Neural Pathways/cytology , Neural Pathways/physiology , Sleep Stages
20.
Neuropharmacology ; 63(6): 1191-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22771461

ABSTRACT

Magnolol (6,6',7,12-tetramethoxy-2,2'-dimethyl-1-beta-berbaman, C(18)H(18)O(2)), an active ingredient of the bark of Magnolia officinalis, has been reported to exert potent anti-epileptic effects via the GABA(A) receptor. The receptor also mediates sleep in humans and animals. The aim of this study was to determine whether magnolol could modulate sleep behaviors by recording EEG and electromyogram in mice. The results showed that magnolol administered i.p. at a dose of 5 or 25 mg/kg could significantly shorten the sleep latency, increase the amount of non-rapid eye movement (non-REM, NREM) and rapid eye movement (REM) sleep for 3 h after administration with an increase in the number of NREM and REM sleep episodes. Magnolol at doses of 5 and 25 mg/kg increased the number of bouts of wakefulness but decreased their duration. On the other hand, magnolol increased the number of state transitions from wakefulness to NREM sleep and subsequently from NREM sleep to wakefulness. Immunohistochemical study showed that magnolol increased c-Fos expression in the neurons of ventrolateral preoptic area, a sleep center in the anterior hypothalamus, and decreased c-Fos expression in the arousal tuberomammillary nucleus, which was located in the caudolateral hypothalamus. The sleep-promoting effects and changes in c-Fos induced by magnolol were reversed by flumazenil, an antagonist at the benzodiazepine site of the GABA(A) receptor. These results indicate that magnolol increased NREM and REM sleep via the GABA(A) receptor.


Subject(s)
Biphenyl Compounds/pharmacology , GABA Modulators , Lignans/pharmacology , Magnolia/chemistry , Receptors, GABA-A/drug effects , Sleep/drug effects , Animals , Arousal/drug effects , Biphenyl Compounds/chemistry , Diazepam/pharmacology , Drug Interactions , Electroencephalography , Electromyography , Flumazenil/pharmacology , Gene Expression/drug effects , Genes, fos/drug effects , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/physiology , Immunohistochemistry , Lignans/chemistry , Male , Mice , Mice, Inbred C57BL , Plant Bark/chemistry , Polysomnography , Preoptic Area/drug effects , Preoptic Area/metabolism , Sleep, REM/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL