Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Curr Biol ; 34(15): 3301-3314.e4, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38944034

RESUMEN

Rapid eye movement (REM) sleep has been hypothesized to promote emotional resilience, but any neuronal circuits mediating this have not been identified. We find that in mice, somatostatin (Som) neurons in the entopeduncular nucleus (EPSom)/internal globus pallidus are predominantly active during REM sleep. This unique REM activity is both necessary and sufficient for maintaining normal REM sleep. Inhibiting or exciting EPSom neurons reduced or increased REM sleep duration, respectively. Activation of the sole downstream target of EPSom neurons, Vglut2 cells in the lateral habenula (LHb), increased sleep via the ventral tegmental area (VTA). A simple chemogenetic scheme to periodically inhibit the LHb over 4 days selectively removed a significant amount of cumulative REM sleep. Chronic, but not acute, REM reduction correlated with mice becoming anxious and more sensitive to aversive stimuli. Therefore, we suggest that cumulative REM sleep, in part generated by the EP → LHb → VTA circuit identified here, could contribute to stabilizing reactions to habitual aversive stimuli.


Asunto(s)
Ansiedad , Sueño REM , Animales , Ratones , Sueño REM/fisiología , Ansiedad/fisiopatología , Masculino , Área Tegmental Ventral/fisiología , Ratones Endogámicos C57BL , Ganglios Basales/fisiología , Ganglios Basales/fisiopatología , Neuronas/fisiología , Núcleo Entopeduncular/fisiología , Somatostatina/metabolismo , Habénula/fisiología , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/genética
2.
Nat Neurosci ; 26(10): 1805-1819, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37735497

RESUMEN

The prefrontal cortex (PFC) enables mammals to respond to situations, including internal states, with appropriate actions. One such internal state could be 'tiredness'. Here, using activity tagging in the mouse PFC, we identified particularly excitable, fast-spiking, somatostatin-expressing, γ-aminobutyric acid (GABA) (PFCSst-GABA) cells that responded to sleep deprivation. These cells projected to the lateral preoptic (LPO) hypothalamus and the lateral hypothalamus (LH). Stimulating PFCSst-GABA terminals in the LPO hypothalamus caused sleep-preparatory behavior (nesting, elevated theta power and elevated temperature), and stimulating PFCSst-GABA terminals in the LH mimicked recovery sleep (non-rapid eye-movement sleep with higher delta power and lower body temperature). PFCSst-GABA terminals had enhanced activity during nesting and sleep, inducing inhibitory postsynaptic currents on diverse cells in the LPO hypothalamus and the LH. The PFC also might feature in deciding sleep location in the absence of excessive fatigue. These findings suggest that the PFC instructs the hypothalamus to ensure that optimal sleep takes place in a suitable place.


Asunto(s)
Área Hipotalámica Lateral , Neuronas , Ratones , Animales , Área Hipotalámica Lateral/metabolismo , Neuronas/fisiología , Somatostatina/metabolismo , Sueño/fisiología , Hipotálamo/fisiología , Ácido gamma-Aminobutírico , Corteza Prefrontal/fisiología , Mamíferos/metabolismo
3.
J Neurosci ; 42(27): 5389-5409, 2022 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-35649726

RESUMEN

The lateral preoptic (LPO) hypothalamus is a center for NREM and REM sleep induction and NREM sleep homeostasis. Although LPO is needed for NREM sleep, we found that calcium signals were, surprisingly, highest in REM sleep. Furthermore, and equally surprising, NMDA receptors in LPO were the main drivers of excitation. Deleting the NMDA receptor GluN1 subunit from LPO abolished calcium signals in all cells and produced insomnia. Mice of both sexes had highly fragmented NREM sleep-wake patterns and could not generate conventionally classified REM sleep. The sleep phenotype produced by deleting NMDA receptors depended on where in the hypothalamus the receptors were deleted. Deleting receptors from the anterior hypothalamic area (AHA) did not influence sleep-wake states. The sleep fragmentation originated from NMDA receptors on GABA neurons in LPO. Sleep fragmentation could be transiently overcome with sleeping medication (zolpidem) or sedatives (dexmedetomidine; Dex). By contrast, fragmentation persisted under high sleep pressure produced by sleep deprivation (SD), mice had a high propensity to sleep but woke up. By analyzing changes in δ power, sleep homeostasis (also referred to as "sleep drive") remained intact after NMDA receptor ablation. We suggest NMDA glutamate receptor activation stabilizes firing of sleep-on neurons and that mechanisms of sleep maintenance differ from that of the sleep drive itself.SIGNIFICANCE STATEMENT Insomnia is a common affliction. Most insomniacs feel that they do not get enough sleep, but in fact, often have good amounts of sleep. Their sleep, however, is fragmented, and sufferers wake up feeling unrefreshed. It is unknown how sleep is maintained once initiated. We find that in mice, NMDA-type glutamate receptors in the hypothalamus are the main drivers of excitation and are required for a range of sleep properties: they are, in fact, needed for both sustained NREM sleep periods, and REM sleep generation. When NMDA receptors are selectively reduced from inhibitory preoptic (PO) neurons, mice have normal total amounts of sleep but high sleep-wake fragmentation, providing a model for studying intractable insomnia.


Asunto(s)
Trastornos del Inicio y del Mantenimiento del Sueño , Sueño REM , Animales , Calcio , Electroencefalografía , Femenino , Hipotálamo , Masculino , Ratones , N-Metilaspartato , Receptores de N-Metil-D-Aspartato , Sueño/fisiología , Privación de Sueño , Sueño REM/fisiología , Vigilia/fisiología
4.
Science ; 377(6601): 63-72, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35771921

RESUMEN

In mice, social defeat stress (SDS), an ethological model for psychosocial stress, induces sleep. Such sleep could enable resilience, but how stress promotes sleep is unclear. Activity-dependent tagging revealed a subset of ventral tegmental area γ-aminobutyric acid (GABA)-somatostatin (VTAVgat-Sst) cells that sense stress and drive non-rapid eye movement (NREM) and REM sleep through the lateral hypothalamus and also inhibit corticotropin-releasing factor (CRF) release in the paraventricular hypothalamus. Transient stress enhances the activity of VTAVgat-Sst cells for several hours, allowing them to exert their sleep effects persistently. Lesioning of VTAVgat-Sst cells abolished SDS-induced sleep; without it, anxiety and corticosterone concentrations remained increased after stress. Thus, a specific circuit allows animals to restore mental and body functions by sleeping, potentially providing a refined route for treating anxiety disorders.


Asunto(s)
Resiliencia Psicológica , Sueño , Derrota Social , Estrés Psicológico , Área Tegmental Ventral , Animales , Hormona Liberadora de Corticotropina/metabolismo , Área Hipotalámica Lateral/fisiopatología , Ratones , Sueño REM , Somatostatina/metabolismo , Estrés Psicológico/fisiopatología , Área Tegmental Ventral/fisiopatología , Ácido gamma-Aminobutírico/metabolismo
5.
Front Neurosci ; 15: 709825, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34720852

RESUMEN

When mice are exposed to external warmth, nitric oxide synthase (NOS1) neurons in the median and medial preoptic (MnPO/MPO) hypothalamus induce sleep and concomitant body cooling. However, how these neurons regulate baseline sleep and body temperature is unknown. Using calcium photometry, we show that NOS1 neurons in MnPO/MPO are predominantly NREM and REM active, especially at the boundary of wake to NREM transitions, and in the later parts of REM bouts, with lower activity during wakefulness. In addition to releasing nitric oxide, NOS1 neurons in MnPO/MPO can release GABA, glutamate and peptides. We expressed tetanus-toxin light-chain in MnPO/MPO NOS1 cells to reduce vesicular release of transmitters. This induced changes in sleep structure: over 24 h, mice had less NREM sleep in their dark (active) phase, and more NREM sleep in their light (sleep) phase. REM sleep episodes in the dark phase were longer, and there were fewer REM transitions between other vigilance states. REM sleep had less theta power. Mice with synaptically blocked MnPO/MPO NOS1 neurons were also warmer than control mice at the dark-light transition (ZT0), as well as during the dark phase siesta (ZT16-20), where there is usually a body temperature dip. Also, at this siesta point of cooled body temperature, mice usually have more NREM, but mice with synaptically blocked MnPO/MPO NOS1 cells showed reduced NREM sleep at this time. Overall, MnPO/MPO NOS1 neurons promote both NREM and REM sleep and contribute to chronically lowering body temperature, particularly at transitions where the mice normally enter NREM sleep.

6.
Mol Psychiatry ; 26(9): 5213-5228, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-32555422

RESUMEN

The ventral tegmental area (VTA), an important source of dopamine, regulates goal- and reward-directed and social behaviors, wakefulness, and sleep. Hyperactivation of dopamine neurons generates behavioral pathologies. But any roles of non-dopamine VTA neurons in psychiatric illness have been little explored. Lesioning or chemogenetically inhibiting VTA GABAergic (VTAVgat) neurons generated persistent wakefulness with mania-like qualities: locomotor activity was increased; sensitivity to D-amphetamine was heightened; immobility times decreased on the tail suspension and forced swim tests; and sucrose preference increased. Furthermore, after sleep deprivation, mice with lesioned VTAVgat neurons did not catch up on lost sleep, even though they were starting from a sleep-deprived baseline, suggesting that sleep homeostasis was bypassed. The mania-like behaviors, including the sleep loss, were reversed by valproate, and re-emerged when treatment was stopped. Lithium salts and lamotrigine, however, had no effect. Low doses of diazepam partially reduced the hyperlocomotion and fully recovered the immobility time during tail suspension. The mania like-behaviors mostly depended on dopamine, because giving D1/D2/D3 receptor antagonists reduced these behaviors, but also partially on VTAVgat projections to the lateral hypothalamus (LH). Optically or chemogenetically inhibiting VTAVgat terminals in the LH elevated locomotion and decreased immobility time during the tail suspension and forced swimming tests. VTAVgat neurons help set an animal's (and perhaps human's) mental and physical activity levels. Inputs inhibiting VTAVgat neurons intensify wakefulness (increased activity, enhanced alertness and motivation), qualities useful for acute survival. In the extreme, however, decreased or failed inhibition from VTAVgat neurons produces mania-like qualities (hyperactivity, hedonia, decreased sleep).


Asunto(s)
Neuronas GABAérgicas , Área Tegmental Ventral , Animales , Neuronas Dopaminérgicas , Área Hipotalámica Lateral , Manía , Ratones
7.
Curr Biol ; 29(19): 3315-3322.e3, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31543455

RESUMEN

Our urge to sleep increases with time spent awake, until sleep becomes inescapable. The sleep following sleep deprivation is longer and deeper, with an increased power of delta (0.5-4 Hz) oscillations, a phenomenon termed sleep homeostasis [1-4]. Although widely expressed genes regulate sleep homeostasis [1, 4-10] and the process is tracked by somnogens and phosphorylation [1, 3, 7, 11-14], at the circuit level sleep homeostasis has remained mysterious. Previously, we found that sedation induced with α2-adrenergic agonists (e.g., dexmedetomidine) and sleep homeostasis both depend on the preoptic (PO) hypothalamus [15, 16]. Dexmedetomidine, increasingly used for long-term sedation in intensive care units [17], induces a non-rapid-eye-movement (NREM)-like sleep but with undesirable hypothermia [18, 19]. Within the PO, various neuronal subtypes (e.g., GABA/galanin and glutamate/NOS1) induce NREM sleep [20-22] and concomitant body cooling [21, 22]. This could be because NREM sleep's restorative effects depend on lower body temperature [23, 24]. Here, we show that mice with lesioned PO galanin neurons have reduced sleep homeostasis: in the recovery sleep following sleep deprivation there is a diminished increase in delta power, and the mice catch up little on lost sleep. Furthermore, dexmedetomidine cannot induce high-power delta oscillations or sustained hypothermia. Some hours after dexmedetomidine administration to wild-type mice there is a rebound in delta power when they enter normal NREM sleep, reminiscent of emergence from torpor. This delta rebound is reduced in mice lacking PO galanin neurons. Thus, sleep homeostasis and dexmedetomidine-induced sedation require PO galanin neurons and likely share common mechanisms.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Dexmedetomidina/farmacología , Galanina/metabolismo , Hipnóticos y Sedantes/farmacología , Neuronas/fisiología , Privación de Sueño/metabolismo , Sueño/fisiología , Animales , Femenino , Homeostasis , Masculino , Ratones , Neuronas/efectos de los fármacos , Sueño/efectos de los fármacos
9.
Sleep ; 42(5)2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30722053

RESUMEN

Acute chemogenetic inhibition of histamine (HA) neurons in adult mice induced nonrapid eye movement (NREM) sleep with an increased delta power. By contrast, selective genetic lesioning of HA neurons with caspase in adult mice exhibited a normal sleep-wake cycle overall, except at the diurnal start of the lights-off period, when they remained sleepier. The amount of time spent in NREM sleep and in the wake state in mice with lesioned HA neurons was unchanged over 24 hr, but the sleep-wake cycle was more fragmented. Both the delayed increase in wakefulness at the start of the night and the sleep-wake fragmentation are similar phenotypes to histidine decarboxylase knockout mice, which cannot synthesize HA. Chronic loss of HA neurons did not affect sleep homeostasis after sleep deprivation. However, the chronic loss of HA neurons or chemogenetic inhibition of HA neurons did notably reduce the ability of the wake-promoting compound modafinil to sustain wakefulness. Thus, part of modafinil's wake-promoting actions arise through the HA system.


Asunto(s)
Histamina/genética , Modafinilo/uso terapéutico , Neuronas/efectos de los fármacos , Privación de Sueño/genética , Promotores de la Vigilia/uso terapéutico , Vigilia/efectos de los fármacos , Animales , Electroencefalografía/efectos de los fármacos , Electroencefalografía/métodos , Vectores Genéticos/administración & dosificación , Histamina/deficiencia , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modafinilo/farmacología , Neuronas/fisiología , Sueño/efectos de los fármacos , Sueño/fisiología , Privación de Sueño/tratamiento farmacológico , Privación de Sueño/metabolismo , Vigilia/fisiología , Promotores de la Vigilia/farmacología
10.
Nat Neurosci ; 22(1): 106-119, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30559475

RESUMEN

We screened for novel circuits in the mouse brain that promote wakefulness. Chemogenetic activation experiments and electroencephalogram recordings pointed to glutamatergic/nitrergic (NOS1) and GABAergic neurons in the ventral tegmental area (VTA). Activating glutamatergic/NOS1 neurons, which were wake- and rapid eye movement (REM) sleep-active, produced wakefulness through projections to the nucleus accumbens and the lateral hypothalamus. Lesioning the glutamate cells impaired the consolidation of wakefulness. By contrast, activation of GABAergic VTA neurons elicited long-lasting non-rapid-eye-movement-like sleep resembling sedation. Lesioning these neurons produced an increase in wakefulness that persisted for at least 4 months. Surprisingly, these VTA GABAergic neurons were wake- and REM sleep-active. We suggest that GABAergic VTA neurons may limit wakefulness by inhibiting the arousal-promoting VTA glutamatergic and/or dopaminergic neurons and through projections to the lateral hypothalamus. Thus, in addition to its contribution to goal- and reward-directed behaviors, the VTA has a role in regulating sleep and wakefulness.


Asunto(s)
Neuronas GABAérgicas/fisiología , Ácido Glutámico/metabolismo , Neuronas/fisiología , Sueño/fisiología , Área Tegmental Ventral/fisiología , Vigilia/fisiología , Animales , Neuronas Dopaminérgicas/fisiología , Ratones , Óxido Nítrico Sintasa de Tipo I/metabolismo , Sueño REM/fisiología , Área Tegmental Ventral/metabolismo
11.
Curr Biol ; 28(14): 2263-2273.e4, 2018 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-30017485

RESUMEN

Mammals, including humans, prepare for sleep by nesting and/or curling up, creating microclimates of skin warmth. To address whether external warmth induces sleep through defined circuitry, we used c-Fos-dependent activity tagging, which captures populations of activated cells and allows them to be reactivated to test their physiological role. External warming tagged two principal groups of neurons in the median preoptic (MnPO)/medial preoptic (MPO) hypothalamic area. GABA neurons located mainly in MPO produced non-rapid eye movement (NREM) sleep but no body temperature decrease. Nitrergic-glutamatergic neurons in MnPO-MPO induced both body cooling and NREM sleep. This circuitry explains how skin warming induces sleep and why the maximal rate of core body cooling positively correlates with sleep onset. Thus, the pathways that promote NREM sleep, reduced energy expenditure, and body cooling are inextricably linked, commanded by the same neurons. This implies that one function of NREM sleep is to lower brain temperature and/or conserve energy.


Asunto(s)
Regulación de la Temperatura Corporal/fisiología , Neuronas/fisiología , Área Preóptica/fisiología , Sueño/fisiología , Adaptación Fisiológica , Animales , Frío , Calor , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-fos/metabolismo
12.
Curr Biol ; 28(4): 580-587.e5, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-29398217

RESUMEN

The lateral habenula has been widely studied for its contribution in generating reward-related behaviors [1, 2]. We have found that this nucleus plays an unexpected role in the sedative actions of the general anesthetic propofol. The lateral habenula is a glutamatergic, excitatory hub that projects to multiple targets throughout the brain, including GABAergic and aminergic nuclei that control arousal [3-5]. When glutamate release from the lateral habenula in mice was genetically blocked, the ability of propofol to induce sedation was greatly diminished. In addition to this reduced sensitivity to propofol, blocking output from the lateral habenula caused natural non-rapid eye movement (NREM) sleep to become highly fragmented, especially during the rest ("lights on") period. This fragmentation was largely reversed by the dual orexinergic antagonist almorexant. We conclude that the glutamatergic output from the lateral habenula is permissive for the sedative actions of propofol and is also necessary for the consolidation of natural sleep.


Asunto(s)
Ácido Glutámico/metabolismo , Habénula/efectos de los fármacos , Hipnóticos y Sedantes/farmacología , Vías Nerviosas/efectos de los fármacos , Propofol/farmacología , Anestésicos Intravenosos/metabolismo , Animales , Células HEK293 , Habénula/fisiología , Humanos , Masculino , Ratones , Vías Nerviosas/fisiología
13.
J Neurosci ; 36(44): 11171-11184, 2016 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-27807161

RESUMEN

Zolpidem, a GABAA receptor-positive modulator, is the gold-standard drug for treating insomnia. Zolpidem prolongs IPSCs to decrease sleep latency and increase sleep time, effects that depend on α2 and/or α3 subunit-containing receptors. Compared with natural NREM sleep, zolpidem also decreases the EEG power, an effect that depends on α1 subunit-containing receptors, and which may make zolpidem-induced sleep less optimal. In this paper, we investigate whether zolpidem needs to potentiate only particular GABAergic pathways to induce sleep without reducing EEG power. Mice with a knock-in F77I mutation in the GABAA receptor γ2 subunit gene are zolpidem-insensitive. Using these mice, GABAA receptors in the frontal motor neocortex and hypothalamic (tuberomammillary nucleus) histaminergic-neurons of γ2I77 mice were made selectively sensitive to zolpidem by genetically swapping the γ2I77 subunits with γ2F77 subunits. When histamine neurons were made selectively zolpidem-sensitive, systemic administration of zolpidem shortened sleep latency and increased sleep time. But in contrast to the effect of zolpidem on wild-type mice, the power in the EEG spectra of NREM sleep was not decreased, suggesting that these EEG power-reducing effects of zolpidem do not depend on reduced histamine release. Selective potentiation of GABAA receptors in the frontal cortex by systemic zolpidem administration also reduced sleep latency, but less so than for histamine neurons. These results could help with the design of new sedatives that induce a more natural sleep. SIGNIFICANCE STATEMENT: Many people who find it hard to get to sleep take sedatives. Zolpidem (Ambien) is the most widely prescribed "sleeping pill." It makes the inhibitory neurotransmitter GABA work better at its receptors throughout the brain. The sleep induced by zolpidem does not resemble natural sleep because it produces a lower power in the brain waves that occur while we are sleeping. We show using mouse genetics that zolpidem only needs to work on specific parts and cell types of the brain, including histamine neurons in the hypothalamus, to induce sleep but without reducing the power of the sleep. This knowledge could help in the design of sleeping pills that induce a more natural sleep.


Asunto(s)
Neocórtex/fisiología , Neuronas/fisiología , Piridinas/administración & dosificación , Receptores de GABA-A/metabolismo , Sueño/efectos de los fármacos , Sueño/fisiología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Histamínicos/administración & dosificación , Hipnóticos y Sedantes/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Neocórtex/citología , Neocórtex/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Fármacos Inductores del Sueño/administración & dosificación , Zolpidem
14.
Neuron ; 87(1): 164-78, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26094607

RESUMEN

Histaminergic neurons in the tuberomammilary nucleus (TMN) of the hypothalamus form a widely projecting, wake-active network that sustains arousal. Yet most histaminergic neurons contain GABA. Selective siRNA knockdown of the vesicular GABA transporter (vgat, SLC32A1) in histaminergic neurons produced hyperactive mice with an exceptional amount of sustained wakefulness. Ablation of the vgat gene throughout the TMN further sharpened this phenotype. Optogenetic stimulation in the caudate-putamen and neocortex of "histaminergic" axonal projections from the TMN evoked tonic (extrasynaptic) GABAA receptor Cl(-) currents onto medium spiny neurons and pyramidal neurons. These currents were abolished following vgat gene removal from the TMN area. Thus wake-active histaminergic neurons generate a paracrine GABAergic signal that serves to provide a brake on overactivation from histamine, but could also increase the precision of neocortical processing. The long range of histamine-GABA axonal projections suggests that extrasynaptic inhibition will be coordinated over large neocortical and striatal areas.


Asunto(s)
Histamina/metabolismo , Área Hipotalámica Lateral/metabolismo , Neocórtex/metabolismo , Neostriado/metabolismo , Inhibición Neural/fisiología , Neuronas/metabolismo , Vigilia/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Axones , Técnicas de Silenciamiento del Gen , Ratones , Inhibición Neural/genética , Optogenética , Células Piramidales/metabolismo , Receptores de GABA-A/metabolismo , Transmisión Sináptica , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética , Vigilia/genética
15.
Nat Neurosci ; 18(4): 553-561, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25706476

RESUMEN

Do sedatives engage natural sleep pathways? It is usually assumed that anesthetic-induced sedation and loss of righting reflex (LORR) arise by influencing the same circuitry to lesser or greater extents. For the α2 adrenergic receptor agonist dexmedetomidine, we found that sedation and LORR were in fact distinct states, requiring different brain areas: the preoptic hypothalamic area and locus coeruleus (LC), respectively. Selective knockdown of α2A adrenergic receptors from the LC abolished dexmedetomidine-induced LORR, but not sedation. Instead, we found that dexmedetomidine-induced sedation resembled the deep recovery sleep that follows sleep deprivation. We used TetTag pharmacogenetics in mice to functionally mark neurons activated in the preoptic hypothalamus during dexmedetomidine-induced sedation or recovery sleep. The neuronal ensembles could then be selectively reactivated. In both cases, non-rapid eye movement sleep, with the accompanying drop in body temperature, was recapitulated. Thus, α2 adrenergic receptor-induced sedation and recovery sleep share hypothalamic circuitry sufficient for producing these behavioral states.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Sedación Profunda , Dexmedetomidina/farmacología , Hipnóticos y Sedantes/farmacología , Hipotálamo/efectos de los fármacos , Sueño/efectos de los fármacos , Animales , Electroencefalografía , Hipotálamo/fisiología , Hipotermia/inducido químicamente , Locus Coeruleus/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Farmacogenética
16.
Curr Biol ; 24(23): 2838-44, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25454592

RESUMEN

Circadian clocks allow anticipation of daily environmental changes. The suprachiasmatic nucleus (SCN) houses the master clock, but clocks are also widely expressed elsewhere in the body. Although some peripheral clocks have established roles, it is unclear what local brain clocks do. We tested the contribution of one putative local clock in mouse histaminergic neurons in the tuberomamillary nucleus to the regulation of the sleep-wake cycle. Histaminergic neurons are silent during sleep, and start firing after wake onset; the released histamine, made by the enzyme histidine decarboxylase (HDC), enhances wakefulness. We found that hdc gene expression varies with time of day. Selectively deleting the Bmal1 (also known as Arntl or Mop3) clock gene from histaminergic cells removes this variation, producing higher HDC expression and brain histamine levels during the day. The consequences include more fragmented sleep, prolonged wake at night, shallower sleep depth (lower nonrapid eye movement [NREM] δ power), increased NREM-to-REM transitions, hindered recovery sleep after sleep deprivation, and impaired memory. Removing BMAL1 from histaminergic neurons does not, however, affect circadian rhythms. We propose that for mammals with polyphasic/nonwake consolidating sleep, the local BMAL1-dependent clock directs appropriately timed declines and increases in histamine biosynthesis to produce an appropriate balance of wake and sleep within the overall daily cycle of rest and activity specified by the SCN.


Asunto(s)
Factores de Transcripción ARNTL/fisiología , Neuronas/metabolismo , Sueño/fisiología , Animales , Ritmo Circadiano/fisiología , Regulación de la Expresión Génica , Histamina/metabolismo , Histidina Descarboxilasa/genética , Histidina Descarboxilasa/metabolismo , Ratones Noqueados , Ratones Transgénicos , Privación de Sueño , Núcleo Supraquiasmático/fisiología
17.
Eur J Neurosci ; 40(1): 2311-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24674448

RESUMEN

How external stimuli prevent the onset of sleep has been little studied. This is usually considered to be a non-specific type of phenomenon. However, the hypnotic drug dexmedetomidine, an agonist at α2 adrenergic receptors, has unusual properties that make it useful for investigating this question. Dexmedetomidine is considered to produce an 'arousable' sleep-like state, so that patients or animals given dexmedetomidine become alert following modest stimulation. We hypothesized that it might be more difficult to make mice unconscious with dexmedetomidine if there was a sufficient external stimulus. Employing a motorized rotating cylinder, which provided a continuous and controlled arousal stimulus, we quantitatively measured the ability of such a stimulus to prevent dexmedetomidine loss of righting reflex in two inbred strains of mice (C57BL/6 and 129X1). We found that whereas the C57BL/6 strain required a strong stimulus to prevent dexmedetomidine-induced hypnosis, the 129X1 strain stayed awake even with minimal stimuli. Remarkably, this could be calibrated as a simple threshold trait, i.e. a binary 'yes-no' response, which after crossing the two mouse strains behaved as a dominant-like trait. We carried out a genome-wide linkage analysis on the F2 progeny to determine if the ability of a stimulus to prevent dexmedetomidine hypnosis could be mapped to one or more chromosomal regions. We identified a locus on chromosome 4 with an associated Logarithm of Odds score exceeding the pre-established threshold level. These results show that complex traits, such as the ability of a stimulus to reverse drug-induced hypnosis, may have precise genetic determinants.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Dexmedetomidina/farmacología , Sueño/genética , Vigilia/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Cromosomas de los Mamíferos , Electroencefalografía , Genes Dominantes , Estudio de Asociación del Genoma Completo , Hipnóticos y Sedantes/farmacología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Farmacogenética , Estimulación Física , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/genética , Reflejo de Enderezamiento/fisiología , Prueba de Desempeño de Rotación con Aceleración Constante , Sueño/efectos de los fármacos , Sueño/fisiología , Especificidad de la Especie , Vigilia/efectos de los fármacos , Vigilia/fisiología
18.
J Biol Chem ; 282(29): 20977-90, 2007 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-17548360

RESUMEN

Certain two-pore domain K(+) channels are plausible targets for volatile general anesthetics, yet little is known at the molecular level about how these simple agents cause channel activation. The first anesthetic-activated K(+) current I(K(An)) that was characterized was discovered in the mollusk Lymnaea stagnalis and is remarkable for both its sensitivity to general anesthetics and its stereoselective responses to anesthetic enantiomers (Franks, N. P., and Lieb, W. R. (1988) Nature 333, 662-664 and Franks, N. P., and Lieb, W. R. (1991) Science 254, 427-430). Here we report the molecular cloning of a two-pore domain K(+) channel LyTASK from L. stagnalis and show that, when expressed in HEK-293 cells, it displays the same biophysical characteristics as the anesthetic-activated K(+) current I(K(An)). Sequence analysis and functional properties show it to be a member of the TASK family of channels with approximately 47% identity at the amino acid level when compared with human TASK-1 and TASK-3. By using chimeric channel constructs and site-directed mutagenesis we have identified the specific amino acid 159 to be a critical determinant of anesthetic sensitivity, which, when mutated to alanine, essentially eliminates anesthetic activation in the human channels and greatly reduces activation in LyTASK. The L159A mutation in LyTASK disrupts the stereoselective response to isoflurane while having no effect on the pH sensitivity of the channel, suggesting this critical amino acid may form part of an anesthetic binding site.


Asunto(s)
Anestésicos/química , Canales de Potasio/química , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Fenómenos Biofísicos , Biofisica , Línea Celular , Humanos , Lymnaea , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Proteínas Recombinantes de Fusión/química , Homología de Secuencia de Aminoácido , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA