Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Neurosci ; 19(2): 290-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26691833

ABSTRACT

During non-rapid eye movement (NREM) sleep, synchronous synaptic activity in the thalamocortical network generates predominantly low-frequency oscillations (<4 Hz) that are modulated by inhibitory inputs from the thalamic reticular nucleus (TRN). Whether TRN cells integrate sleep-wake signals from subcortical circuits remains unclear. We found that GABA neurons from the lateral hypothalamus (LHGABA) exert a strong inhibitory control over TRN GABA neurons (TRNGABA). We found that optogenetic activation of this circuit recapitulated state-dependent changes of TRN neuron activity in behaving mice and induced rapid arousal during NREM, but not REM, sleep. During deep anesthesia, activation of this circuit induced sustained cortical arousal. In contrast, optogenetic silencing of LHGABA-TRNGABA transmission increased the duration of NREM sleep and amplitude of delta (1-4 Hz) oscillations. Collectively, these results demonstrate that TRN cells integrate subcortical arousal inputs selectively during NREM sleep and may participate in sleep intensity.


Subject(s)
Arousal/physiology , Cerebral Cortex/physiology , Consciousness/physiology , Hypothalamus/physiology , Thalamus/physiology , Anesthesia , Animals , Behavior, Animal/physiology , Delta Rhythm , Female , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Nerve Net/physiology , Optogenetics , Sleep/physiology , Sleep, REM/physiology , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , gamma-Aminobutyric Acid/physiology
2.
PLoS One ; 10(6): e0130177, 2015.
Article in English | MEDLINE | ID: mdl-26076358

ABSTRACT

In addition to cognitive decline, individuals affected by Alzheimer's disease (AD) can experience important neuropsychiatric symptoms including sleep disturbances. We characterized the sleep-wake cycle in the TgCRND8 mouse model of AD, which overexpresses a mutant human form of amyloid precursor protein resulting in high levels of ß-amyloid and plaque formation by 3 months of age. Polysomnographic recordings in freely-moving mice were conducted to study sleep-wake cycle architecture at 3, 7 and 11 months of age and corresponding levels of ß-amyloid in brain regions regulating sleep-wake states were measured. At all ages, TgCRND8 mice showed increased wakefulness and reduced non-rapid eye movement (NREM) sleep during the resting and active phases. Increased wakefulness in TgCRND8 mice was accompanied by a shift in the waking power spectrum towards fast frequency oscillations in the beta (14-20 Hz) and low gamma range (20-50 Hz). Given the phenotype of hyperarousal observed in TgCRND8 mice, the role of noradrenergic transmission in the promotion of arousal, and previous work reporting an early disruption of the noradrenergic system in TgCRND8, we tested the effects of the alpha-1-adrenoreceptor antagonist, prazosin, on sleep-wake patterns in TgCRND8 and non-transgenic (NTg) mice. We found that a lower dose (2 mg/kg) of prazosin increased NREM sleep in NTg but not in TgCRND8 mice, whereas a higher dose (5 mg/kg) increased NREM sleep in both genotypes, suggesting altered sensitivity to noradrenergic blockade in TgCRND8 mice. Collectively our results demonstrate that amyloidosis in TgCRND8 mice is associated with sleep-wake cycle dysfunction, characterized by hyperarousal, validating this model as a tool towards understanding the relationship between ß-amyloid overproduction and disrupted sleep-wake patterns in AD.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/physiology , Amyloidosis/etiology , Disease Models, Animal , Sleep Wake Disorders/complications , Sleep/physiology , Wakefulness/physiology , Amyloidosis/pathology , Animals , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Polysomnography , Sleep Wake Disorders/pathology
3.
J Neurosci ; 35(14): 5435-41, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25855162

ABSTRACT

The lateral hypothalamus (LH) is a key regulator of multiple vital behaviors. The firing of brain-wide-projecting LH neurons releases neuropeptides promoting wakefulness (orexin/hypocretin; OH), or sleep (melanin-concentrating hormone; MCH). OH neurons, which coexpress glutamate and dynorphin, have been proposed to excite their neighbors, including MCH neurons, suggesting that LH may sometimes coengage its antagonistic outputs. However, it remains unclear if, when, and how OH actions promote temporal separation of the sleep and wake signals, a process that fails in narcolepsy caused by OH loss. To explore this directly, we paired optogenetic stimulation of OH cells (at rates that promoted awakening in vivo) with electrical monitoring of MCH cells in mouse brain slices. Membrane potential recordings showed that OH cell firing inhibited action potential firing in most MCH neurons, an effect that required GABAA but not dynorphin receptors. Membrane current analysis showed that OH cell firing increased the frequency of fast GABAergic currents in MCH cells, an effect blocked by antagonists of OH but not dynorphin or glutamate receptors, and mimicked by bath-applied OH peptide. In turn, neural network imaging with a calcium indicator genetically targeted to MCH neurons showed that excitation by bath-applied OH peptides occurs in a minority of MCH cells. Collectively, our data provide functional microcircuit evidence that intra-LH feedforward loops may facilitate appropriate switching between sleep and wake signals, potentially preventing sleep disorders.


Subject(s)
Hypothalamic Hormones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Melanins/metabolism , Nerve Net/physiology , Neural Inhibition/physiology , Neurons/physiology , Neuropeptides/metabolism , Optogenetics , Pituitary Hormones/metabolism , Signal Transduction/physiology , Animals , Calcium/metabolism , Channelrhodopsins , Excitatory Amino Acid Antagonists/pharmacology , Hypothalamic Area, Lateral/cytology , Hypothalamic Area, Lateral/metabolism , Hypothalamic Hormones/genetics , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lysine/analogs & derivatives , Lysine/metabolism , Melanins/genetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Neural Inhibition/drug effects , Neuropeptides/genetics , Neuropeptides/pharmacology , Orexins , Patch-Clamp Techniques , Pituitary Hormones/genetics , Transduction, Genetic , gamma-Aminobutyric Acid/metabolism , Red Fluorescent Protein
4.
Article in English | MEDLINE | ID: mdl-24324458

ABSTRACT

The cyclic peptide Melanin Concentrating Hormone (MCH) is known to control a large number of brain functions in mammals such as food intake and metabolism, stress response, anxiety, sleep/wake cycle, memory, and reward. Based on neuro-anatomical and electrophysiological studies these functions were attributed to neuronal circuits expressing MCHR1, the single MCH receptor in rodents. In complement to our recently published work (1) we provided here new data regarding the action of MCH on ependymocytes in the mouse brain. First, we establish that MCHR1 mRNA is expressed in the ependymal cells of the third ventricle epithelium. Second, we demonstrated a tonic control of MCH-expressing neurons on ependymal cilia beat frequency using in vitro optogenics. Finally, we performed in vivo measurements of CSF flow using fluorescent micro-beads in wild-type and MCHR1-knockout mice. Collectively, our results demonstrated that MCH-expressing neurons modulate ciliary beating of ependymal cells at the third ventricle and could contribute to maintain cerebro-spinal fluid homeostasis.

6.
Nat Neurosci ; 16(11): 1637-43, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24056699

ABSTRACT

Rapid-eye movement (REM) sleep correlates with neuronal activity in the brainstem, basal forebrain and lateral hypothalamus. Lateral hypothalamus melanin-concentrating hormone (MCH)-expressing neurons are active during sleep, but their effects on REM sleep remain unclear. Using optogenetic tools in newly generated Tg(Pmch-cre) mice, we found that acute activation of MCH neurons (ChETA, SSFO) at the onset of REM sleep extended the duration of REM, but not non-REM, sleep episodes. In contrast, their acute silencing (eNpHR3.0, archaerhodopsin) reduced the frequency and amplitude of hippocampal theta rhythm without affecting REM sleep duration. In vitro activation of MCH neuron terminals induced GABAA-mediated inhibitory postsynaptic currents in wake-promoting histaminergic neurons of the tuberomammillary nucleus (TMN), and in vivo activation of MCH neuron terminals in TMN or medial septum also prolonged REM sleep episodes. Collectively, these results suggest that activation of MCH neurons maintains REM sleep, possibly through inhibition of arousal circuits in the mammalian brain.


Subject(s)
Hypothalamus/physiology , Nerve Net/physiology , Neural Pathways/physiology , Optogenetics , Sleep, REM/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Animals, Newborn , Bicuculline/pharmacology , Channelrhodopsins , Excitatory Amino Acid Antagonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , Gene Expression Regulation , Hypothalamic Hormones/genetics , Hypothalamus/cytology , Hypothalamus/drug effects , Melanins/genetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/drug effects , Neural Pathways/drug effects , Neurons/drug effects , Neurons/physiology , Pituitary Hormones/genetics , Theta Rhythm/drug effects , Theta Rhythm/genetics , Transduction, Genetic , Valine/analogs & derivatives , Valine/pharmacology
7.
PLoS One ; 8(6): e66122, 2013.
Article in English | MEDLINE | ID: mdl-23785477

ABSTRACT

Optimal norepinephrine levels in the prefrontal cortex (PFC) increase delay-related firing and enhance working memory, whereas stress-related or pathologically high levels of norepinephrine are believed to inhibit working memory via α1 adrenoceptors. However, it has been shown that activation of Gq-coupled and phospholipase C-linked receptors can induce persistent firing, a cellular correlate of working memory, in cortical pyramidal neurons. Therefore, despite its importance in stress and cognition, the exact role of norepinephrine in modulating PFC activity remains elusive. Using electrophysiology and optogenetics, we report here that norepinephrine induces persistent firing in pyramidal neurons of the PFC independent of recurrent fast synaptic excitation. This persistent excitatory effect involves presynaptic α1 adrenoceptors facilitating glutamate release and subsequent activation of postsynaptic mGluR5 receptors, and is enhanced by postsynaptic α2 adrenoceptors inhibiting HCN channel activity. Activation of α2 adrenoceptors or inhibition of HCN channels also enhances cholinergic persistent responses in pyramidal neurons, providing a mechanism of crosstalk between noradrenergic and cholinergic inputs. The present study describes a novel cellular basis for the noradrenergic control of cortical information processing and supports a synergistic combination of intrinsic and network mechanisms for the expression of mnemonic properties in pyramidal neurons.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Norepinephrine/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Action Potentials/drug effects , Animals , Glutamic Acid/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Male , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/physiology , Presynaptic Terminals/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Receptors, Kainic Acid/metabolism
8.
PLoS One ; 7(12): e52525, 2012.
Article in English | MEDLINE | ID: mdl-23300698

ABSTRACT

The recently discovered Nesfatin-1 plays a role in appetite regulation as a satiety factor through hypothalamic leptin-independent mechanisms. Nesfatin-1 is co-expressed with Melanin-Concentrating Hormone (MCH) in neurons from the tuberal hypothalamic area (THA) which are recruited during sleep states, especially paradoxical sleep (PS). To help decipher the contribution of this contingent of THA neurons to sleep regulatory mechanisms, we thus investigated in rats whether the co-factor Nesfatin-1 is also endowed with sleep-modulating properties. Here, we found that the disruption of the brain Nesfatin-1 signaling achieved by icv administration of Nesfatin-1 antiserum or antisense against the nucleobindin2 (NUCB2) prohormone suppressed PS with little, if any alteration of slow wave sleep (SWS). Further, the infusion of Nesfatin-1 antiserum after a selective PS deprivation, designed for elevating PS needs, severely prevented the ensuing expected PS recovery. Strengthening these pharmacological data, we finally demonstrated by using c-Fos as an index of neuronal activation that the recruitment of Nesfatin-1-immunoreactive neurons within THA is positively correlated to PS but not to SWS amounts experienced by rats prior to sacrifice. In conclusion, this work supports a functional contribution of the Nesfatin-1 signaling, operated by THA neurons, to PS regulatory mechanisms. We propose that these neurons, likely releasing MCH as a synergistic factor, constitute an appropriate lever by which the hypothalamus may integrate endogenous signals to adapt the ultradian rhythm and maintenance of PS in a manner dictated by homeostatic needs. This could be done through the inhibition of downstream targets comprised primarily of the local hypothalamic wake-active orexin- and histamine-containing neurons.


Subject(s)
Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Homeostasis , Hypothalamus/cytology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Satiety Response , Sleep, REM/physiology , Animals , Gene Expression Regulation , Hypothalamus/pathology , Hypothalamus/physiology , Hypothalamus/physiopathology , Male , Neurons/cytology , Neurons/pathology , Nucleobindins , Polysomnography , Rats , Rats, Sprague-Dawley , Signal Transduction , Sleep Deprivation/metabolism , Sleep Deprivation/pathology , Sleep Deprivation/physiopathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...