Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cell Res ; 34(3): 214-231, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38332199

RESUMEN

Flickering light stimulation has emerged as a promising non-invasive neuromodulation strategy to alleviate neuropsychiatric disorders. However, the lack of a neurochemical underpinning has hampered its therapeutic development. Here, we demonstrate that light flickering triggered an immediate and sustained increase (up to 3 h after flickering) in extracellular adenosine levels in the primary visual cortex (V1) and other brain regions, as a function of light frequency and intensity, with maximal effects observed at 40 Hz frequency and 4000 lux. We uncovered cortical (glutamatergic and GABAergic) neurons, rather than astrocytes, as the cellular source, the intracellular adenosine generation from AMPK-associated energy metabolism pathways (but not SAM-transmethylation or salvage purine pathways), and adenosine efflux mediated by equilibrative nucleoside transporter-2 (ENT2) as the molecular pathway responsible for extracellular adenosine generation. Importantly, 40 Hz (but not 20 and 80 Hz) light flickering for 30 min enhanced non-rapid eye movement (non-REM) and REM sleep for 2-3 h in mice. This somnogenic effect was abolished by ablation of V1 (but not superior colliculus) neurons and by genetic deletion of the gene encoding ENT2 (but not ENT1), but recaptured by chemogenetic inhibition of V1 neurons and by focal infusion of adenosine into V1 in a dose-dependent manner. Lastly, 40 Hz light flickering for 30 min also promoted sleep in children with insomnia by decreasing sleep onset latency, increasing total sleep time, and reducing waking after sleep onset. Collectively, our findings establish the ENT2-mediated adenosine signaling in V1 as the neurochemical basis for 40 Hz flickering-induced sleep and unravel a novel and non-invasive treatment for insomnia, a condition that affects 20% of the world population.


Asunto(s)
Trastornos del Inicio y del Mantenimiento del Sueño , Humanos , Niño , Animales , Ratones , Sueño , Transducción de Señal , Adenosina , Astrocitos
2.
Front Pharmacol ; 14: 1098976, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36969831

RESUMEN

Sleep, torpor, and hibernation are three distinct hypometabolic states. However, they have some similar physiological features, such as decreased core body temperature and slowing heart rate. In addition, the accumulation of adenosine seems to be a common feature before entry into these three states, suggesting that adenosine and its receptors, also known as P1 receptors, may mediate the initiation and maintenance of these states. This review, therefore, summarizes the current research on the roles and possible neurobiological mechanisms of adenosine and P1 receptors in sleep, torpor, and hibernation. Understanding these aspects will give us better prospects in sleep disorders, therapeutic hypothermia, and aerospace medicine.

3.
Front Neurosci ; 16: 982596, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36090271

RESUMEN

GABAergic neurons in the vestibular nuclei (VN) participate in multiple vital vestibular sensory processing allowing for the maintenance and rehabilitation of vestibular functions. However, although the important role of GABA in the central vestibular system has been widely reported, the underlying neural circuits between VN GABAergic neurons and other brain functional regions remain elusive, which limits the further study of the underlying mechanism. Hence, it is necessary to elucidate neural connectivity based on outputs and inputs of GABAergic neurons in the VN. This study employed a modified rabies virus retrograde tracing vector and cre-dependent adeno-associated viruses (AAVs) anterograde tracing vector, combined with a transgenic VGAT-IRES-Cre mice, to map the inputs and outputs of VN GABAergic neurons in the whole brain. We found that 51 discrete brain regions received projections from VN GABAergic neurons in the whole brain, and there were 77 upstream nuclei innervating GABAergic neurons in the VN. These nuclei were mainly located in four brain regions, including the medulla, pons, midbrain, and cerebellum. Among them, VN GABAergic neurons established neural circuits with some functional nuclei in the whole brain, especially regulating balance maintenance, emotion control, pain processing, sleep and circadian rhythm regulation, and fluid homeostasis. Therefore, this study deepens a comprehensive understanding of the whole-brain neural connectivity of VN, providing the neuroanatomical information for further research on the neural mechanism of the co-morbidities with vestibular dysfunction.

4.
Brain Res ; 1790: 147984, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35709891

RESUMEN

Caffeine has been used as a first-line drug for treatment of apnea neonatorum for decades due to its high safety and effectiveness. Studies report that caffeine mainly acts as a blocker of Adenosine Receptors (ARs). However, the mechanism of caffeine in reducing apnea neonatorum in the central nervous system has not been fully explored. Medial parabrachial nucleus (MPB) is part of the respiratory center of the pons that may be related to the activity of caffeine. Previous studies have not explored the effect and mechanism of caffeine on MPB neurons. To elucidate this, the current study used antagonists of A1 and A2a receptors to mimic the effect of caffeine in MPB of mice in vitro using the patch-clamp technique. The firing rates and spontaneous post-synaptic currents were recorded. The findings of the study showed that caffeine excited MPB neurons. Notably, the adenosine A1R antagonist 8-cyclopentyl-1,3-dimethyl-xanthine (CPT) but not the adenosine A2aR antagonist Istradefylline (KW6002) mimicked the exciting effect of caffeine, implying that caffeine excited MPB neurons in mice by blocking A1Rs. Further, the results indicated that caffeine could increase efficiency of synaptic transmission to excite MPB neurons. These findings suggest that A1Rs in MPB may be potential targets for caffeine in reducing apnea neonatorum.


Asunto(s)
Núcleos Parabraquiales , Receptor de Adenosina A1 , Adenosina/farmacología , Animales , Apnea , Cafeína/farmacología , Ratones , Neuronas/metabolismo , Núcleos Parabraquiales/metabolismo , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A2A
5.
Int J Mol Sci ; 23(9)2022 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-35562990

RESUMEN

Sleep and wakefulness are basic behavioral states that require coordination between several brain regions, and they involve multiple neurochemical systems, including neuropeptides. Neuropeptides are a group of peptides produced by neurons and neuroendocrine cells of the central nervous system. Like traditional neurotransmitters, neuropeptides can bind to specific surface receptors and subsequently regulate neuronal activities. For example, orexin is a crucial component for the maintenance of wakefulness and the suppression of rapid eye movement (REM) sleep. In addition to orexin, melanin-concentrating hormone, and galanin may promote REM sleep. These results suggest that neuropeptides play an important role in sleep-wake regulation. These neuropeptides can be divided into three categories according to their effects on sleep-wake behaviors in rodents and humans. (i) Galanin, melanin-concentrating hormone, and vasoactive intestinal polypeptide are sleep-promoting peptides. It is also noticeable that vasoactive intestinal polypeptide particularly increases REM sleep. (ii) Orexin and neuropeptide S have been shown to induce wakefulness. (iii) Neuropeptide Y and substance P may have a bidirectional function as they can produce both arousal and sleep-inducing effects. This review will introduce the distribution of various neuropeptides in the brain and summarize the roles of different neuropeptides in sleep-wake regulation. We aim to lay the foundation for future studies to uncover the mechanisms that underlie the initiation, maintenance, and end of sleep-wake states.


Asunto(s)
Galanina , Neuropéptidos , Galanina/farmacología , Péptidos y Proteínas de Señalización Intracelular/farmacología , Neuropéptidos/metabolismo , Orexinas/farmacología , Sueño/fisiología , Péptido Intestinal Vasoactivo/farmacología
6.
Front Neurosci ; 16: 830474, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35360167

RESUMEN

Background: Hypersomnia is a common and highly impairing symptom marked by pathological excessive sleepiness, which induces suboptimal functioning and poor quality of life. Hypersomnia can be both a primary (e.g., hypersomnolence disorder) and secondary (e.g., tumors, and head trauma) symptom of disorders. However, its underlying mechanisms remain largely unknown. Case Presentation: We report that three clinical cases with lesions around the paraventricular nucleus of the hypothalamus (PVH) area showed excessive daytime sleepiness and a prolonged nocturnal sleep lasting more than 20 h per day. Sleep architecture and subjective daytime sleepiness were examined by polysomnography. These cases were presented with stroke, myelin oligodendrocyte glycoprotein (MOG) antibody associated disorders and neuromyelitis optical spectrum disorder (NMOSD), respectively. Magnetic resonance imaging (MRI) showed lesions around the PVH area in all these three patients. After treatment of their primary disorders, their excessive sleep decreased as the PVH area recovered. Conclusion: Our findings suggest that the PVH may play an essential role in the occurrence of hypersomnia.

7.
Ying Yong Sheng Tai Xue Bao ; 32(12): 4447-4456, 2021 Dec.
Artículo en Chino | MEDLINE | ID: mdl-34951286

RESUMEN

With the acceleration of industrialization, the toxic effect of heavy metal cadmium (Cd) pollution has become prominent. In order to explore the molecular mechanism of the physiological regulation of Caulerpa lentillifera under Cd stress, we analyzed the transcriptome of Cd-stressed (Hcd2+) algae tissues using RNA-Seq. A total of 702 differentially expressed genes (DEGs) were screened between the control and Hcd2+ groups, out of which 257 genes were up-regulated and 445 genes were down-regulated in the Hcd2+ group. We conducted functional annotation and enrichment analysis of the obtained DEGs. The results showed that various biological functions of C. lentillifera were affected under Cd2+stress, which eventually showed growth inhibition. Results of GO enrichment analysis showed that the production and removal of reactive oxygen species (ROS) in C. lentillifera were out of balance and caused oxidative damage such as DNA damage. Results of KEGG enrichment analysis showed that many photosynthesis-related pathways were inhibited, indicating that Cd2+ stress led to disorder of photosynthetic reaction of C. lentillifera.


Asunto(s)
Caulerpa , Metales Pesados , Cadmio/toxicidad , Caulerpa/genética , Perfilación de la Expresión Génica , Metales Pesados/toxicidad , Transcriptoma
8.
Elife ; 102021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34787078

RESUMEN

Hypersomnolence disorder (HD) is characterized by excessive sleep, which is a common sequela following stroke, infection, or tumorigenesis. HD is traditionally thought to be associated with lesions of wake-promoting nuclei. However, lesions of a single wake-promoting nucleus, or even two simultaneously, did not exert serious HD. Therefore, the specific nucleus and neural circuitry for HD remain unknown. Here, we observed that the paraventricular nucleus of the hypothalamus (PVH) exhibited higher c-fos expression during the active period (23:00) than during the inactive period (11:00) in mice. Therefore, we speculated that the PVH, in which most neurons are glutamatergic, may represent one of the key arousal-controlling centers. By using vesicular glutamate transporter 2 (vglut2Cre) mice together with fiber photometry, multichannel electrophysiological recordings, and genetic approaches, we found that PVHvglut2 neurons were most active during wakefulness. Chemogenetic activation of PVHvglut2 neurons induced wakefulness for 9 hr, and photostimulation of PVHvglut2→parabrachial complex/ventral lateral septum circuits immediately drove transitions from sleep to wakefulness. Moreover, lesioning or chemogenetic inhibition of PVHvglut2 neurons dramatically decreased wakefulness. These results indicate that the PVH is critical for arousal promotion and maintenance.


Asunto(s)
Nivel de Alerta/fisiología , Trastornos de Somnolencia Excesiva/fisiopatología , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/fisiopatología , Animales , Masculino , Ratones , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Vigilia
9.
Brain Res ; 1768: 147590, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34310936

RESUMEN

Depression, rapid eye movement (REM) sleep behavior disorder, and altered olfaction are often present in Parkinson's disease. Our previous studies demonstrated the role of the olfactory bulb (OB) in causing REM sleep disturbances in depression. Furthermore, adenosine A2A receptors (A2AR) which are richly expressed in the OB, play an important role in the regulation of REM sleep. Caffeine, an adenosine A1 receptors and A2AR antagonist, and other A2AR antagonists were reported to improve olfactory function and restore age-related olfactory deficits. Therefore, we hypothesized that the A2AR neurons in the OB may regulate olfaction or odor-guided behaviors in mice. In the present study, we employed chemogenetics to specifically activate or inhibit neuronal activity. Then, buried food test and olfactory habituation/dishabituation test were performed to measure the changes in the mice's olfactory ability. We demonstrated that activation of OB neurons or OB A2AR neurons shortened the latency of buried food test and enhanced olfactory habituation to the same odors and dishabituation to different odors; inhibition of these neurons showed the opposite effects. Photostimulation of ChR2-expressing OB A2AR neuron terminals evoked inward current in the olfactory tubercle (OT) and the piriform cortex (Pir), which was blocked by glutamate receptor antagonists 2-amino-5-phosphonopentanoic acid and 6-cyano-7nitroquinoxaline-2,3-dione. Collectively, these results suggest that the OB mediates olfaction via A2AR neurons in mice. Moreover, the excitatory glutamatergic release from OB neurons to the OT and the Pir were found responsible for the olfaction-mediated effects of OB A2AR neurons.


Asunto(s)
Receptor de Adenosina A2A/metabolismo , Olfato/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Odorantes , Bulbo Olfatorio/metabolismo , Corteza Olfatoria/metabolismo , Percepción Olfatoria/fisiología , Corteza Piriforme/metabolismo , Receptor de Adenosina A2A/fisiología
10.
Curr Biol ; 31(9): 1893-1902.e5, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33705720

RESUMEN

Although general anesthesia (GA) enables patients to undergo surgery without consciousness, the precise neural mechanisms underlying this phenomenon have yet to be identified. In addition to many studies over the past two decades implicating the thalamus, cortex, brainstem, and conventional sleep-wake circuits in GA-induced loss of consciousness (LOC), some recent studies have begun to highlight the importance of other brain areas as well. Here, we found that population activities of neurons expressing dopamine D1 receptor (D1R) in the nucleus accumbens (NAc), a critical interface between the basal ganglia and limbic system, began to decrease before sevoflurane-induced LOC and gradually returned after recovery of consciousness (ROC). Chemogenetic activation of NAcD1R neurons delayed induction of and accelerated emergence from sevoflurane GA, whereas chemogenetic inhibition of NAcD1R neurons exerted opposite effects. Moreover, transient activation of NAcD1R neurons induced significant cortical activation and behavioral emergence during continuous steady-state GA with sevoflurane or deep anesthesia state with constant and stable burst-suppression oscillations. Taken together, our findings uncover that NAcD1R neurons modulated states of consciousness associated with sevoflurane GA and may represent an area for targeting GA-induced changes in consciousness and ameliorating related adverse effects.


Asunto(s)
Anestesia , Núcleo Accumbens , Estado de Conciencia , Humanos , Neuronas/metabolismo , Núcleo Accumbens/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Sevoflurano , Inconsciencia
11.
Sleep ; 43(2)2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-31552427

RESUMEN

Light has immediate effects on sleep in rodents, but the neural pathways underlying the effect remain to be elucidated. The intergeniculate leaflet (IGL) containing GABAergic neurons receives direct retinal inputs. We hypothesized that IGL GABAergic neurons may mediate light-induced sleep. EEG/electromyogram recording, immunohistochemistry, electrophysiology, optogenetics, fiber photometry, behavioral tests, and cell-specific destruction were employed to investigate the role of IGL GABAergic neurons in the regulation of acute light-induced sleep. Here, EEG/electromyogram recordings revealed that acute light exposure during the nocturnal active phase in mice induced a significant increase in non-rapid eye movement and rapid eye movement sleep compared with controls. Immunohistochemistry showed that acute light exposure for 2 hours in the active phase induced an increase in c-Fos expression in the IGL, whereas lights-off in the rest phase inhibited it. Patch clamp coupled with optogenetics demonstrated that retinal ganglion cells had monosynaptic functional connections to IGL GABAergic neurons. Calcium activity by fiber photometry in freely behaving mice showed that light exposure increased the activity of IGL GABAergic neurons. Furthermore, lesion of IGL GABAergic neurons by caspase-3 virus significantly attenuated the sleep-promoting effect of light exposure during active phases. Collectively, these results clearly indicated that the IGL is one of key nuclei mediating light-induced sleep in mice.


Asunto(s)
Neuronas GABAérgicas , Cuerpos Geniculados , Animales , Ritmo Circadiano , Ratones , Proteínas Proto-Oncogénicas c-fos , Ratas , Ratas Wistar , Sueño , Núcleo Supraquiasmático
12.
Pain ; 161(2): 288-299, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31651580

RESUMEN

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.


Asunto(s)
Corteza Cerebral/fisiopatología , Ritmo Delta/fisiología , Hiperalgesia/fisiopatología , Neuralgia/fisiopatología , Umbral del Dolor/fisiología , Sueño/fisiología , Inhibidores de Captación Adrenérgica/farmacología , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Antagonistas Colinérgicos/farmacología , Sincronización Cortical/efectos de los fármacos , Sincronización Cortical/fisiología , Ritmo Delta/efectos de los fármacos , Electroencefalografía , Electromiografía , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Giro del Cíngulo/efectos de los fármacos , Giro del Cíngulo/metabolismo , Hiperalgesia/metabolismo , Ratones , Modafinilo/farmacología , Neuralgia/metabolismo , Umbral del Dolor/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Reserpina/farmacología , Nervio Ciático/cirugía , Escopolamina/farmacología , Sueño/efectos de los fármacos , Privación de Sueño/inducido químicamente , Privación de Sueño/fisiopatología , Promotores de la Vigilia/farmacología
13.
Neuropharmacology ; 168: 107923, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31874169

RESUMEN

The olfactory tubercle (OT), an important nucleus in processing sensory information, has been reported to change cortical activity under odor. However, little is known about the physiological role and mechanism of the OT in sleep-wake regulation. The OT expresses abundant adenosine A2A receptors (A2ARs), which are important in sleep regulation. Therefore, we hypothesized that the OT regulates sleep via A2ARs. This study examined sleep-wake profiles through electroencephalography and electromyography recordings with pharmacological and chemogenetic manipulations in freely moving rodents. Compared with their controls, activation of OT A2ARs pharmacologically and OT A2AR neurons via chemogenetics increased non-rapid eye movement sleep for 5 and 3 h, respectively, while blockade of A2ARs decreased non-rapid eye movement sleep. Tracing and electrophysiological studies showed OT A2AR neurons projected to the ventral pallidum and lateral hypothalamus, forming inhibitory innervations. Together, these findings indicate that A2ARs in the OT play an important role in sleep regulation.


Asunto(s)
Agonistas del Receptor de Adenosina A2/farmacología , Tubérculo Olfatorio/metabolismo , Receptor de Adenosina A2A/metabolismo , Sueño/fisiología , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Electroencefalografía/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tubérculo Olfatorio/efectos de los fármacos , Fenetilaminas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Adenosina A2A/genética , Roedores , Sueño/efectos de los fármacos
14.
Handb Exp Pharmacol ; 253: 359-381, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-28646346

RESUMEN

The classic endogenous somnogen adenosine promotes sleep via A1 and A2A receptors. In this chapter, we present an overview of the current knowledge regarding the regulation of adenosine levels, adenosine receptors, and available pharmacologic and genetic tools to manipulate the adenosine system. This is followed by a summary of current knowledge of the role of adenosine and its receptors in the regulation of sleep and wakefulness. Despite strong data implicating numerous brain areas, including the basal forebrain, the tuberomammillary nucleus, the lateral hypothalamus, and the nucleus accumbens, in the adenosinergic control of sleep, the complete neural circuitry in the brain involved in the sleep-promoting effects of adenosine remains unclear. Moreover, the popular demand for natural sleep aids has led to a search for natural compounds that can promote sleep via adenosine receptor activation. Finally, we discuss the effects of caffeine in man and the possible use of more selective adenosine receptor drugs for the treatment of sleep disorders.


Asunto(s)
Adenosina , Sueño , Adenosina/metabolismo , Encéfalo/fisiología , Vigilia/fisiología
15.
Front Neurosci ; 12: 807, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30455627

RESUMEN

The suprachiasmatic nucleus (SCN) is the principal pacemaker driving the circadian rhythms of physiological behaviors. The SCN consists of distinct neurons expressing neuropeptides, including arginine vasopressin (AVP), vasoactive intestinal polypeptide (VIP), gastrin-releasing peptide (GRP), cholecystokinin (CCK), and so on. AVP, VIP, and GRP neurons receive light stimulation from the retina to synchronize endogenous circadian clocks with the solar day, whereas CCK neurons are not directly innervated by retinal ganglion cells and may be involved in the non-photic regulation of the circadian clock. To better understand the function of CCK neurons in non-photic circadian rhythm, it is vital to clarify the direct afferent inputs to CCK neurons in the SCN. Here, we utilized a recently developed rabies virus- and Cre/loxP-based, cell type-specific, retrograde tracing system to map and quantitatively analyze the whole-brain monosynaptic inputs to SCN CCK neurons. We found that SCN CCK neurons received direct inputs from 29 brain nuclei. Among these nuclei, paraventricular nucleus of the hypothalamus (PVH), paraventricular nucleus of the thalamus (PVT), supraoptic nucleus (SON), ventromedial nucleus of the hypothalamus, and seven other nuclei sent numerous inputs to CCK neurons. Moderate inputs originated from the zona incerta, periventricular hypothalamic nucleus, and five other nuclei. A few inputs to CCK neurons originated from the orbital frontal cortex, prelimbic cortex, cingulate cortex, claustrum, and seven other nuclei. In addition, SCN CCK neurons were preferentially innervated by AVP neurons of the ipsilateral PVH and SON rather than their contralateral counterpart, whereas the contralateral PVT sent more projections to CCK neurons than to its ipsilateral counterpart. Taken together, these results expand our knowledge of the specific innervation to mouse SCN CCK neurons and provide an important indication for further investigations on the function of CCK neurons.

16.
Curr Neuropharmacol ; 16(1): 5-16, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28486925

RESUMEN

BACKGROUND: Sleep and epilepsy are mutually related in a complex, bidirectional manner. However, our understanding of this relationship remains unclear. RESULTS: The literatures of the neurobiological basis of the interactions between sleep and epilepsy indicate that non rapid eye movement sleep and idiopathic generalized epilepsy share the same thalamocortical networks. Most of neurotransmitters and neuromodulators such as adenosine, melatonin, prostaglandin D2, serotonin, and histamine are found to regulate the sleep-wake behavior and also considered to have antiepilepsy effects; antiepileptic drugs, in turn, also have effects on sleep. Furthermore, many drugs that regulate the sleep-wake cycle can also serve as potential antiseizure agents. The nonpharmacological management of epilepsy including ketogenic diet, epilepsy surgery, neurostimulation can also influence sleep. CONCLUSION: In this paper, we address the issues involved in these phenomena and also discuss the various therapies used to modify them.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia/fisiopatología , Epilepsia/terapia , Neurotransmisores/uso terapéutico , Sueño/fisiología , Animales , Epilepsia/patología , Humanos , Neurotransmisores/metabolismo , Neurotransmisores/farmacología , Sueño/efectos de los fármacos
17.
Sci Rep ; 7(1): 12678, 2017 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-28978989

RESUMEN

Ethanol has extensive effects on sleep and daytime alertness, causing premature disability and death. Adenosine, as a potent sleep-promoting substance, is involved in many cellular and behavioral responses to ethanol. However, the mechanisms of hypnotic effects of ethanol remain unclear. In this study, we investigated the role of adenosine in ethanol-induced sleep using C57BL/6Slac mice, adenosine A2A receptor (A2AR) knockout mice, and their wild-type littermates. The results showed that intraperitoneal injection of ethanol (3.0 g/kg) at 21:00 decreased the latency to non-rapid eye movement (NREM) sleep and increased the duration of NREM sleep for 5 h. Ethanol dose-dependently increased NREM sleep, which was consistent with decreases in wakefulness in C57BL/6Slac mice compared with their own control. Caffeine (5, 10, or 15 mg/kg), a nonspecific adenosine receptor antagonist, dose-dependently and at high doses completely blocked ethanol-induced NREM sleep when administered 30 min prior to (but not after) ethanol injection. Moreover, ethanol-induced NREM sleep was completely abolished in A2AR knockout mice compared with wild-type mice. These findings strongly indicate that A2AR is a key receptor for the hypnotic effects of ethanol, and pretreatment of caffeine might be a strategy to counter the hypnotic effects of ethanol.


Asunto(s)
Etanol/farmacología , Hipnóticos y Sedantes/farmacología , Receptor de Adenosina A2A/metabolismo , Animales , Cafeína/farmacología , Electroencefalografía , Etanol/administración & dosificación , Ratones Endogámicos C57BL , Latencia del Sueño/efectos de los fármacos , Sueño REM/efectos de los fármacos , Vigilia/efectos de los fármacos
18.
Elife ; 62017 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-29022877

RESUMEN

Dysfunction of the striatum is frequently associated with sleep disturbances. However, its role in sleep-wake regulation has been paid little attention even though the striatum densely expresses adenosine A2A receptors (A2ARs), which are essential for adenosine-induced sleep. Here we showed that chemogenetic activation of A2AR neurons in specific subregions of the striatum induced a remarkable increase in non-rapid eye movement (NREM) sleep. Anatomical mapping and immunoelectron microscopy revealed that striatal A2AR neurons innervated the external globus pallidus (GPe) in a topographically organized manner and preferentially formed inhibitory synapses with GPe parvalbumin (PV) neurons. Moreover, lesions of GPe PV neurons abolished the sleep-promoting effect of striatal A2AR neurons. In addition, chemogenetic inhibition of striatal A2AR neurons led to a significant decrease of NREM sleep at active period, but not inactive period of mice. These findings reveal a prominent contribution of striatal A2AR neuron/GPe PV neuron circuit in sleep control.


Asunto(s)
Globo Pálido/fisiología , Neostriado/fisiología , Neuronas/fisiología , Parvalbúminas/análisis , Receptor de Adenosina A2A/análisis , Sueño , Vigilia , Adenosina/metabolismo , Animales , Mapeo Encefálico , Masculino , Ratones , Microscopía Inmunoelectrónica , Neuronas/química
19.
Nat Commun ; 8(1): 734, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28963505

RESUMEN

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.


Asunto(s)
Núcleo Accumbens/fisiología , Sueño/fisiología , Animales , Ritmo Circadiano , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Motivación , Técnicas de Placa-Clamp , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor de Adenosina A2A/metabolismo , Receptor de Adenosina A2A/fisiología
20.
J Sleep Res ; 26(3): 386-393, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28318068

RESUMEN

Natural helicid (4-formylphenyl-O-ß-d-allopyranoside), a main active constituent from seeds of the Chinese herb Helicia nilagirica, has been reported to exert a sedative, analgesic and hypnotic effect, and is used clinically to treat neurasthenic syndrome, vascular headaches and trigeminal neuralgia. In the current study, mechanical allodynia tests, electroencephalograms, electromyogram recordings and c-Fos expression in neuropathic pain-like model mice of partial sciatic nerve ligation were used to investigate the effect of helicid on neuropathic pain and co-morbid insomnia. Our results showed that helicid at a dose of 100, 200 or 400 mg kg-1 could increase the mechanical threshold by 2.5-, 2.8- and 3.1-fold for 3 h after administration, respectively. Helicid at 200 and 400 mg kg-1 given at 07:00 hours increased the amount of non-rapid eye movement sleep in a 3-h period by 1.27- and 1.35-fold in partial sciatic nerve ligated mice. However, helicid (400 mg kg-1 ) given at 21:00 hours did not change the sleep pattern in normal mice. Immunohistochemical study showed that helicid (400 mg kg-1 ) administration could reverse the increase of c-Fos expression in the neurons of the rostral anterior cingulate cortex and tuberomammillary nucleus, and the decrease of c-Fos expression in the ventrolateral preoptic area caused by partial sciatic nerve ligation. These results indicate that helicid is an effective agent for both neuropathic pain and sleep disturbances in partial sciatic nerve ligated mice.


Asunto(s)
Benzaldehídos/uso terapéutico , Neuralgia/complicaciones , Neuralgia/tratamiento farmacológico , Trastornos del Inicio y del Mantenimiento del Sueño/etiología , Trastornos del Inicio y del Mantenimiento del Sueño/prevención & control , Analgésicos/administración & dosificación , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Benzaldehídos/administración & dosificación , Benzaldehídos/farmacología , Modelos Animales de Enfermedad , Electroencefalografía , Electromiografía , Genes fos/genética , Giro del Cíngulo/efectos de los fármacos , Giro del Cíngulo/metabolismo , Hiperalgesia/diagnóstico , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/farmacología , Hipnóticos y Sedantes/uso terapéutico , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Ligadura , Masculino , Ratones , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , Nervio Ciático , Sueño/efectos de los fármacos , Sueño/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA