Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61.483
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 186(25): 5500-5516.e21, 2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-38016470

RESUMEN

Most animals require sleep, and sleep loss induces serious pathophysiological consequences, including death. Previous experimental approaches for investigating sleep impacts in mice have been unable to persistently deprive animals of both rapid eye movement sleep (REMS) and non-rapid eye movement sleep (NREMS). Here, we report a "curling prevention by water" paradigm wherein mice remain awake 96% of the time. After 4 days of exposure, mice exhibit severe inflammation, and approximately 80% die. Sleep deprivation increases levels of prostaglandin D2 (PGD2) in the brain, and we found that elevated PGD2 efflux across the blood-brain-barrier-mediated by ATP-binding cassette subfamily C4 transporter-induces both accumulation of circulating neutrophils and a cytokine-storm-like syndrome. Experimental disruption of the PGD2/DP1 axis dramatically reduced sleep-deprivation-induced inflammation. Thus, our study reveals that sleep-related changes in PGD2 in the central nervous system drive profound pathological consequences in the peripheral immune system.


Asunto(s)
Privación de Sueño , Animales , Ratones , Citocinas/metabolismo , Inflamación , Prostaglandina D2 , Sueño/fisiología , Privación de Sueño/genética , Privación de Sueño/metabolismo , Síndrome , Humanos , Ratas , Línea Celular , Tormentas Ciclónicas , Neutrófilos/metabolismo
2.
Cell ; 186(26): 5739-5750.e17, 2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-38070510

RESUMEN

Conscious perception is greatly diminished during sleep, but the underlying circuit mechanism is poorly understood. We show that cortical ignition-a brain process shown to be associated with conscious awareness in humans and non-human primates-is strongly suppressed during non-rapid-eye-movement (NREM) sleep in mice due to reduced cholinergic modulation and rapid inhibition of cortical responses. Brain-wide functional ultrasound imaging and cell-type-specific calcium imaging combined with optogenetics showed that activity propagation from visual to frontal cortex is markedly reduced during NREM sleep due to strong inhibition of frontal pyramidal neurons. Chemogenetic activation and inactivation of basal forebrain cholinergic neurons powerfully increased and decreased visual-to-frontal activity propagation, respectively. Furthermore, although multiple subtypes of dendrite-targeting GABAergic interneurons in the frontal cortex are more active during wakefulness, soma-targeting parvalbumin-expressing interneurons are more active during sleep. Chemogenetic manipulation of parvalbumin interneurons showed that sleep/wake-dependent cortical ignition is strongly modulated by perisomatic inhibition of pyramidal neurons.


Asunto(s)
Electroencefalografía , Parvalbúminas , Sueño , Animales , Ratones , Neuronas Colinérgicas/fisiología , Lóbulo Frontal/metabolismo , Parvalbúminas/metabolismo , Sueño/fisiología , Vigilia/fisiología
3.
Cell ; 186(13): 2911-2928.e20, 2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37269832

RESUMEN

Animals with complex nervous systems demand sleep for memory consolidation and synaptic remodeling. Here, we show that, although the Caenorhabditis elegans nervous system has a limited number of neurons, sleep is necessary for both processes. In addition, it is unclear if, in any system, sleep collaborates with experience to alter synapses between specific neurons and whether this ultimately affects behavior. C. elegans neurons have defined connections and well-described contributions to behavior. We show that spaced odor-training and post-training sleep induce long-term memory. Memory consolidation, but not acquisition, requires a pair of interneurons, the AIYs, which play a role in odor-seeking behavior. In worms that consolidate memory, both sleep and odor conditioning are required to diminish inhibitory synaptic connections between the AWC chemosensory neurons and the AIYs. Thus, we demonstrate in a living organism that sleep is required for events immediately after training that drive memory consolidation and alter synaptic structures.


Asunto(s)
Caenorhabditis elegans , Odorantes , Animales , Caenorhabditis elegans/fisiología , Olfato , Sueño/fisiología , Sinapsis/fisiología
4.
Cell ; 181(6): 1307-1328.e15, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32502393

RESUMEN

The view that sleep is essential for survival is supported by the ubiquity of this behavior, the apparent existence of sleep-like states in the earliest animals, and the fact that severe sleep loss can be lethal. The cause of this lethality is unknown. Here we show, using flies and mice, that sleep deprivation leads to accumulation of reactive oxygen species (ROS) and consequent oxidative stress, specifically in the gut. ROS are not just correlates of sleep deprivation but drivers of death: their neutralization prevents oxidative stress and allows flies to have a normal lifespan with little to no sleep. The rescue can be achieved with oral antioxidant compounds or with gut-targeted transgenic expression of antioxidant enzymes. We conclude that death upon severe sleep restriction can be caused by oxidative stress, that the gut is central in this process, and that survival without sleep is possible when ROS accumulation is prevented. VIDEO ABSTRACT.


Asunto(s)
Tracto Gastrointestinal/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Privación de Sueño/metabolismo , Sueño/fisiología , Animales , Antioxidantes/metabolismo , Drosophila , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Estrés Oxidativo/fisiología
5.
Cell ; 183(7): 1986-2002.e26, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33333022

RESUMEN

Serotonin plays a central role in cognition and is the target of most pharmaceuticals for psychiatric disorders. Existing drugs have limited efficacy; creation of improved versions will require better understanding of serotonergic circuitry, which has been hampered by our inability to monitor serotonin release and transport with high spatial and temporal resolution. We developed and applied a binding-pocket redesign strategy, guided by machine learning, to create a high-performance, soluble, fluorescent serotonin sensor (iSeroSnFR), enabling optical detection of millisecond-scale serotonin transients. We demonstrate that iSeroSnFR can be used to detect serotonin release in freely behaving mice during fear conditioning, social interaction, and sleep/wake transitions. We also developed a robust assay of serotonin transporter function and modulation by drugs. We expect that both machine-learning-guided binding-pocket redesign and iSeroSnFR will have broad utility for the development of other sensors and in vitro and in vivo serotonin detection, respectively.


Asunto(s)
Evolución Molecular Dirigida , Aprendizaje Automático , Serotonina/metabolismo , Algoritmos , Secuencia de Aminoácidos , Amígdala del Cerebelo/fisiología , Animales , Conducta Animal , Sitios de Unión , Encéfalo/metabolismo , Células HEK293 , Humanos , Cinética , Modelos Lineales , Ratones , Ratones Endogámicos C57BL , Fotones , Unión Proteica , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Sueño/fisiología , Vigilia/fisiología
6.
Cell ; 179(2): 514-526.e13, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31585085

RESUMEN

Sleep has been implicated in both memory consolidation and forgetting of experiences. However, it is unclear what governs the balance between consolidation and forgetting. Here, we tested how activity-dependent processing during sleep might differentially regulate these two processes. We specifically examined how neural reactivations during non-rapid eye movement (NREM) sleep were causally linked to consolidation versus weakening of the neural correlates of neuroprosthetic skill. Strikingly, we found that slow oscillations (SOs) and delta (δ) waves have dissociable and competing roles in consolidation versus forgetting. By modulating cortical spiking linked to SOs or δ waves using closed-loop optogenetic methods, we could, respectively, weaken or strengthen consolidation and thereby bidirectionally modulate sleep-dependent performance gains. We further found that changes in the temporal coupling of spindles to SOs relative to δ waves could account for such effects. Thus, our results indicate that neural activity driven by SOs and δ waves have competing roles in sleep-dependent memory consolidation.


Asunto(s)
Encéfalo/fisiología , Ritmo Delta , Consolidación de la Memoria/fisiología , Sueño/fisiología , Animales , Masculino , Ratas , Ratas Long-Evans
7.
Cell ; 177(5): 1293-1307.e16, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-31031008

RESUMEN

The perioculomotor (pIII) region of the midbrain was postulated as a sleep-regulating center in the 1890s but largely neglected in subsequent studies. Using activity-dependent labeling and gene expression profiling, we identified pIII neurons that promote non-rapid eye movement (NREM) sleep. Optrode recording showed that pIII glutamatergic neurons expressing calcitonin gene-related peptide alpha (CALCA) are NREM-sleep active; optogenetic and chemogenetic activation/inactivation showed that they strongly promote NREM sleep. Within the pIII region, CALCA neurons form reciprocal connections with another population of glutamatergic neurons that express the peptide cholecystokinin (CCK). Activation of CCK neurons also promoted NREM sleep. Both CALCA and CCK neurons project rostrally to the preoptic hypothalamus, whereas CALCA neurons also project caudally to the posterior ventromedial medulla. Activation of each projection increased NREM sleep. Together, these findings point to the pIII region as an excitatory sleep center where different subsets of glutamatergic neurons promote NREM sleep through both local reciprocal connections and long-range projections.


Asunto(s)
Hipotálamo/metabolismo , Mesencéfalo/metabolismo , Neuronas/metabolismo , Fases del Sueño/fisiología , Animales , Colecistoquinina/metabolismo , Hipotálamo/citología , Mesencéfalo/citología , Ratones , Ratones Transgénicos , Neuronas/citología , Optogenética
8.
Cell ; 175(5): 1213-1227.e18, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30318147

RESUMEN

Neurons use two main schemes to encode information: rate coding (frequency of firing) and temporal coding (timing or pattern of firing). While the importance of rate coding is well established, it remains controversial whether temporal codes alone are sufficient for controlling behavior. Moreover, the molecular mechanisms underlying the generation of specific temporal codes are enigmatic. Here, we show in Drosophila clock neurons that distinct temporal spike patterns, dissociated from changes in firing rate, encode time-dependent arousal and regulate sleep. From a large-scale genetic screen, we identify the molecular pathways mediating the circadian-dependent changes in ionic flux and spike morphology that rhythmically modulate spike timing. Remarkably, the daytime spiking pattern alone is sufficient to drive plasticity in downstream arousal neurons, leading to increased firing of these cells. These findings demonstrate a causal role for temporal coding in behavior and define a form of synaptic plasticity triggered solely by temporal spike patterns.


Asunto(s)
Plasticidad Neuronal , Sueño/fisiología , Potenciales de Acción , Animales , Relojes Circadianos/fisiología , Drosophila , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Modelos Neurológicos , Neuronas/metabolismo , Optogenética , Canales de Potasio/genética , Canales de Potasio/metabolismo , Canales de Potasio Calcio-Activados/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Transmisión Sináptica
9.
Cell ; 169(2): 203-215.e13, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28388406

RESUMEN

Patterns of daily human activity are controlled by an intrinsic circadian clock that promotes ∼24 hr rhythms in many behavioral and physiological processes. This system is altered in delayed sleep phase disorder (DSPD), a common form of insomnia in which sleep episodes are shifted to later times misaligned with the societal norm. Here, we report a hereditary form of DSPD associated with a dominant coding variation in the core circadian clock gene CRY1, which creates a transcriptional inhibitor with enhanced affinity for circadian activator proteins Clock and Bmal1. This gain-of-function CRY1 variant causes reduced expression of key transcriptional targets and lengthens the period of circadian molecular rhythms, providing a mechanistic link to DSPD symptoms. The allele has a frequency of up to 0.6%, and reverse phenotyping of unrelated families corroborates late and/or fragmented sleep patterns in carriers, suggesting that it affects sleep behavior in a sizeable portion of the human population.


Asunto(s)
Criptocromos/metabolismo , Trastornos del Sueño del Ritmo Circadiano/genética , Ritmo Circadiano , Criptocromos/genética , Exones , Femenino , Eliminación de Gen , Humanos , Masculino , Persona de Mediana Edad , Linaje , Trastornos del Sueño del Ritmo Circadiano/fisiopatología
10.
Annu Rev Neurosci ; 46: 123-143, 2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-36854316

RESUMEN

This review explores the interface between circadian timekeeping and the regulation of brain function by astrocytes. Although astrocytes regulate neuronal activity across many time domains, their cell-autonomous circadian clocks exert a particular role in controlling longer-term oscillations of brain function: the maintenance of sleep states and the circadian ordering of sleep and wakefulness. This is most evident in the central circadian pacemaker, the suprachiasmatic nucleus, where the molecular clock of astrocytes suffices to drive daily cycles of neuronal activity and behavior. In Alzheimer's disease, sleep impairments accompany cognitive decline. In mouse models of the disease, circadian disturbances accelerate astroglial activation and other brain pathologies, suggesting that daily functions in astrocytes protect neuronal homeostasis. In brain cancer, treatment in the morning has been associated with prolonged survival, and gliomas have daily rhythms in gene expression and drug sensitivity. Thus, circadian time is fast becoming critical to elucidating reciprocal astrocytic-neuronal interactions in health and disease.


Asunto(s)
Astrocitos , Relojes Circadianos , Ratones , Animales , Astrocitos/fisiología , Ritmo Circadiano/fisiología , Relojes Circadianos/genética , Sueño , Núcleo Supraquiasmático/metabolismo
11.
Physiol Rev ; 103(3): 2231-2269, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-36731029

RESUMEN

Salt-inducible kinases (SIKs), which comprise a family of three homologous serine-threonine kinases, were first described for their role in sodium sensing but have since been shown to regulate multiple aspects of physiology. These kinases are activated or deactivated in response to extracellular signals that are cell surface receptor mediated and go on to phosphorylate multiple targets including the transcription cofactors CRTC1-3 and the class IIa histone deacetylases (HDACs). Thus, the SIK family conveys signals about the cellular environment to reprogram transcriptional and posttranscriptional processes in response. In this manner, SIKs have been shown to regulate metabolic responses to feeding/fasting, cell division and oncogenesis, inflammation, immune responses, and most recently, sleep and circadian rhythms. Sleep and circadian rhythms are master regulators of physiology and are exquisitely sensitive to regulation by environmental light and physiological signals such as the need for sleep. Salt-inducible kinases have been shown to be central to the molecular regulation of both these processes. Here, we summarize the molecular mechanisms by which SIKs control these different domains of physiology and highlight where there is mechanistic overlap with sleep/circadian rhythm control.


Asunto(s)
Proteínas Serina-Treonina Quinasas , Factores de Transcripción , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Cloruro de Sodio , Ritmo Circadiano , Sueño
12.
Mol Cell ; 81(24): 4979-4993.e7, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34798058

RESUMEN

The characteristics of the sleep drivers and the mechanisms through which sleep relieves the cellular homeostatic pressure are unclear. In flies, zebrafish, mice, and humans, DNA damage levels increase during wakefulness and decrease during sleep. Here, we show that 6 h of consolidated sleep is sufficient to reduce DNA damage in the zebrafish dorsal pallium. Induction of DNA damage by neuronal activity and mutagens triggered sleep and DNA repair. The activity of the DNA damage response (DDR) proteins Rad52 and Ku80 increased during sleep, and chromosome dynamics enhanced Rad52 activity. The activity of the DDR initiator poly(ADP-ribose) polymerase 1 (Parp1) increased following sleep deprivation. In both larva zebrafish and adult mice, Parp1 promoted sleep. Inhibition of Parp1 activity reduced sleep-dependent chromosome dynamics and repair. These results demonstrate that DNA damage is a homeostatic driver for sleep, and Parp1 pathways can sense this cellular pressure and facilitate sleep and repair activity.


Asunto(s)
Conducta Animal , Encéfalo , Daño del ADN , Reparación del ADN , Neuronas , Poli(ADP-Ribosa) Polimerasa-1 , Sueño , Proteínas de Pez Cebra , Animales , Femenino , Masculino , Animales Modificados Genéticamente , Encéfalo/enzimología , Encéfalo/patología , Encéfalo/fisiopatología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Ratones Endogámicos C57BL , Neuronas/enzimología , Neuronas/patología , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/fisiología , Proteína Recombinante y Reparadora de ADN Rad52/genética , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Factores de Tiempo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
13.
Annu Rev Neurosci ; 43: 119-140, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32075519

RESUMEN

While neurons and circuits are almost unequivocally considered to be the computational units and actuators of behavior, a complete understanding of the nervous system must incorporate glial cells. Far beyond a copious but passive substrate, glial influence is inextricable from neuronal physiology, whether during developmental guidance and synaptic shaping or through the trophic support, neurotransmitter and ion homeostasis, cytokine signaling and immune function, and debris engulfment contributions that this class provides throughout an organism's life. With such essential functions, among a growing literature of nuanced roles, it follows that glia are consequential to behavior in adult animals, with novel genetic tools allowing for the investigation of these phenomena in living organisms. We discuss here the relevance of glia for maintaining circadian rhythms and also for serving functions of sleep.


Asunto(s)
Ritmo Circadiano/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Sueño/fisiología , Animales , Drosophila/fisiología , Humanos , Neurotransmisores/metabolismo
14.
Physiol Rev ; 100(2): 805-868, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31804897

RESUMEN

Sleep spindles are burstlike signals in the electroencephalogram (EEG) of the sleeping mammalian brain and electrical surface correlates of neuronal oscillations in thalamus. As one of the most inheritable sleep EEG signatures, sleep spindles probably reflect the strength and malleability of thalamocortical circuits that underlie individual cognitive profiles. We review the characteristics, organization, regulation, and origins of sleep spindles and their implication in non-rapid-eye-movement sleep (NREMS) and its functions, focusing on human and rodent. Spatially, sleep spindle-related neuronal activity appears on scales ranging from small thalamic circuits to functional cortical areas, and generates a cortical state favoring intracortical plasticity while limiting cortical output. Temporally, sleep spindles are discrete events, part of a continuous power band, and elements grouped on an infraslow time scale over which NREMS alternates between continuity and fragility. We synthesize diverse and seemingly unlinked functions of sleep spindles for sleep architecture, sensory processing, synaptic plasticity, memory formation, and cognitive abilities into a unifying sleep spindle concept, according to which sleep spindles 1) generate neural conditions of large-scale functional connectivity and plasticity that outlast their appearance as discrete EEG events, 2) appear preferentially in thalamic circuits engaged in learning and attention-based experience during wakefulness, and 3) enable a selective reactivation and routing of wake-instated neuronal traces between brain areas such as hippocampus and cortex. Their fine spatiotemporal organization reflects NREMS as a physiological state coordinated over brain and body and may indicate, if not anticipate and ultimately differentiate, pathologies in sleep and neurodevelopmental, -degenerative, and -psychiatric conditions.


Asunto(s)
Ondas Encefálicas , Encéfalo/fisiopatología , Cognición , Enfermedades del Sistema Nervioso/fisiopatología , Periodicidad , Fases del Sueño , Trastornos del Sueño-Vigilia/fisiopatología , Animales , Atención , Encéfalo/metabolismo , Humanos , Inteligencia , Memoria , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/psicología , Plasticidad Neuronal , Trastornos del Sueño-Vigilia/genética , Trastornos del Sueño-Vigilia/metabolismo , Trastornos del Sueño-Vigilia/psicología , Factores de Tiempo
15.
Annu Rev Pharmacol Toxicol ; 64: 359-386, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-37708433

RESUMEN

Sleep is essential for human well-being, yet the quality and quantity of sleep reduce as age advances. Older persons (>65 years old) are more at risk of disorders accompanied and/or exacerbated by poor sleep. Furthermore, evidence supports a bidirectional relationship between disrupted sleep and Alzheimer's disease (AD) or related dementias. Orexin/hypocretin neuropeptides stabilize wakefulness, and several orexin receptor antagonists (ORAs) are approved for the treatment of insomnia in adults. Dysregulation of the orexin system occurs in aging and AD, positioning ORAs as advantageous for these populations. Indeed, several clinical studies indicate that ORAs are efficacious hypnotics in older persons and dementia patients and, as in adults, are generally well tolerated. ORAs are likely to be more effective when administered early in sleep/wake dysregulation to reestablish good sleep/wake-related behaviors and reduce the accumulation of dementia-associated proteinopathic substrates. Improving sleep in aging and dementia represents a tremendous opportunity to benefit patients, caregivers, and health systems.


Asunto(s)
Enfermedad de Alzheimer , Antagonistas de los Receptores de Orexina , Humanos , Anciano , Anciano de 80 o más Años , Orexinas/farmacología , Antagonistas de los Receptores de Orexina/farmacología , Antagonistas de los Receptores de Orexina/uso terapéutico , Receptores de Orexina , Sueño/fisiología , Enfermedad de Alzheimer/tratamiento farmacológico
16.
EMBO J ; 42(1): e111485, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36385434

RESUMEN

Sleep intensity is adjusted by the length of previous awake time, and under tight homeostatic control by protein phosphorylation. Here, we establish microglia as a new cellular component of the sleep homeostasis circuit. Using quantitative phosphoproteomics of the mouse frontal cortex, we demonstrate that microglia-specific deletion of TNFα perturbs thousands of phosphorylation sites during the sleep period. Substrates of microglial TNFα comprise sleep-related kinases such as MAPKs and MARKs, and numerous synaptic proteins, including a subset whose phosphorylation status encodes sleep need and determines sleep duration. As a result, microglial TNFα loss attenuates the build-up of sleep need, as measured by electroencephalogram slow-wave activity and prevents immediate compensation for loss of sleep. Our data suggest that microglia control sleep homeostasis by releasing TNFα which acts on neuronal circuitry through dynamic control of phosphorylation.


Asunto(s)
Microglía , Factor de Necrosis Tumoral alfa , Ratones , Animales , Microglía/metabolismo , Fosforilación , Factor de Necrosis Tumoral alfa/metabolismo , Sueño/fisiología , Homeostasis/fisiología
17.
EMBO J ; 42(3): e111304, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36477886

RESUMEN

Parvalbumin-positive neurons (PVs) are the main class of inhibitory neurons in the mammalian central nervous system. By examining diurnal changes in synaptic and neuronal activity of PVs in the supragranular layer of the mouse primary visual cortex (V1), we found that both PV input and output are modulated in a time- and sleep-dependent manner throughout the 24-h day. We first show that PV-evoked inhibition is stronger by the end of the light cycle (ZT12) relative to the end of the dark cycle (ZT0), which is in line with the lower inhibitory input of PV neurons at ZT12 than at ZT0. Interestingly, PV inhibitory and excitatory synaptic transmission slowly oscillate in opposite directions during the light/dark cycle. Although excitatory synapses are predominantly regulated by experience, inhibitory synapses are regulated by sleep, via acetylcholine activating M1 receptors. Consistent with synaptic regulation of PVs, we further show in vivo that spontaneous PV activity displays daily rhythm mainly determined by visual experience, which negatively correlates with the activity cycle of surrounding pyramidal neurons and the dorsal lateral geniculate nucleus-evoked responses in V1. These findings underscore the physiological significance of PV's daily modulation.


Asunto(s)
Neuronas , Parvalbúminas , Animales , Ratones , Parvalbúminas/metabolismo , Neuronas/metabolismo , Células Piramidales/metabolismo , Transmisión Sináptica , Sueño , Mamíferos
18.
Annu Rev Neurosci ; 42: 27-46, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-30699051

RESUMEN

Wakefulness, rapid eye movement (REM) sleep, and non-rapid eye movement (NREM) sleep are characterized by distinct electroencephalogram (EEG), electromyogram (EMG), and autonomic profiles. The circuit mechanism coordinating these changes during sleep-wake transitions remains poorly understood. The past few years have witnessed rapid progress in the identification of REM and NREM sleep neurons, which constitute highly distributed networks spanning the forebrain, midbrain, and hindbrain. Here we propose an arousal-action circuit for sleep-wake control in which wakefulness is supported by separate arousal and action neurons, while REM and NREM sleep neurons are part of the central somatic and autonomic motor circuits. This model is well supported by the currently known sleep and wake neurons. It can also account for the EEG, EMG, and autonomic profiles of wake, REM, and NREM states and several key features of their transitions. The intimate association between the sleep and autonomic/somatic motor control circuits suggests that a primary function of sleep is to suppress motor activity.


Asunto(s)
Nivel de Alerta/fisiología , Modelos Neurológicos , Sueño/fisiología , Animales , Encéfalo/fisiología , Electroencefalografía , Humanos , Red Nerviosa/fisiología , Neuronas/fisiología , Fases del Sueño/fisiología , Vigilia/fisiología
19.
Proc Natl Acad Sci U S A ; 121(13): e2312664121, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38498719

RESUMEN

Sleep is an evolutionarily conserved state that supports brain functions, including synaptic plasticity, in species across the animal kingdom. Here, we examine the neuroanatomical and cell-type distribution of presynaptic scaling in the fly brain after sleep loss. We previously found that sleep loss drives accumulation of the active zone scaffolding protein Bruchpilot (BRP) within cholinergic Kenyon cells of the Drosophila melanogaster mushroom body (MB), but not in other classes of MB neurons. To test whether similar cell type-specific trends in plasticity occur broadly across the brain, we used a flp-based genetic reporter to label presynaptic BRP in cholinergic, dopaminergic, GABAergic, or glutamatergic neurons. We then collected whole-brain confocal image stacks of BRP intensity to systematically quantify BRP, a marker of presynapse abundance, across 37 neuropil regions of the central fly brain. Our results indicate that sleep loss, either by overnight (12-h) mechanical stimulation or chronic sleep disruption in insomniac mutants, broadly elevates cholinergic synapse abundance across the brain, while synapse abundance in neurons that produce other neurotransmitters undergoes weaker, if any, changes. Extending sleep deprivation to 24 h drives brain-wide upscaling in glutamatergic, but not other, synapses. Finally, overnight male-male social pairings induce increased BRP in excitatory synapses despite male-female pairings eliciting more waking activity, suggesting experience-specific plasticity. Within neurotransmitter class and waking context, BRP changes are similar across the 37 neuropil domains, indicating that similar synaptic scaling rules may apply across the brain during acute sleep loss and that sleep need may broadly alter excitatory-inhibitory balance in the central brain.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Femenino , Masculino , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Privación de Sueño/metabolismo , Sinapsis/metabolismo , Encéfalo/metabolismo , Colinérgicos
20.
Proc Natl Acad Sci U S A ; 121(16): e2316150121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38593074

RESUMEN

For nearly a century, evidence has accumulated indicating that the lateral hypothalamus (LH) contains neurons essential to sustain wakefulness. While lesion or inactivation of LH neurons produces a profound increase in sleep, stimulation of inhibitory LH neurons promotes wakefulness. To date, the primary wake-promoting cells that have been identified in the LH are the hypocretin/orexin (Hcrt) neurons, yet these neurons have little impact on total sleep or wake duration across the 24-h period. Recently, we and others have identified other LH populations that increase wakefulness. In the present study, we conducted microendoscopic calcium imaging in the LH concomitant with EEG and locomotor activity (LMA) recordings and found that a subset of LH neurons that express Ca2+/calmodulin-dependent protein kinase IIα (CaMKIIα) are preferentially active during wakefulness. Chemogenetic activation of these neurons induced sustained wakefulness and greatly increased LMA even in the absence of Hcrt signaling. Few LH CaMKIIα-expressing neurons are hypocretinergic or histaminergic while a small but significant proportion are GABAergic. Ablation of LH inhibitory neurons followed by activation of the remaining LH CaMKIIα neurons induced similar levels of wakefulness but blunted the LMA increase. Ablated animals showed no significant changes in sleep architecture but both spontaneous LMA and high theta (8 to 10 Hz) power during wakefulness were reduced. Together, these findings indicate the existence of two subpopulations of LH CaMKIIα neurons: an inhibitory population that promotes locomotion without affecting sleep architecture and an excitatory population that promotes prolonged wakefulness even in the absence of Hcrt signaling.


Asunto(s)
Área Hipotalámica Lateral , Vigilia , Animales , Vigilia/fisiología , Área Hipotalámica Lateral/fisiología , Orexinas/metabolismo , Sueño/fisiología , Neuronas/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA