Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 122023 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-37548652

RESUMEN

Sleep is a nearly universal feature of animal behaviour, yet many of the molecular, genetic, and neuronal substrates that orchestrate sleep/wake transitions lie undiscovered. Employing a viral insertion sleep screen in larval zebrafish, we identified a novel gene, dreammist (dmist), whose loss results in behavioural hyperactivity and reduced sleep at night. The neuronally expressed dmist gene is conserved across vertebrates and encodes a small single-pass transmembrane protein that is structurally similar to the Na+,K+-ATPase regulator, FXYD1/Phospholemman. Disruption of either fxyd1 or atp1a3a, a Na+,K+-ATPase alpha-3 subunit associated with several heritable movement disorders in humans, led to decreased night-time sleep. Since atpa1a3a and dmist mutants have elevated intracellular Na+ levels and non-additive effects on sleep amount at night, we propose that Dmist-dependent enhancement of Na+ pump function modulates neuronal excitability to maintain normal sleep behaviour.


Asunto(s)
Sodio , Pez Cebra , Animales , Humanos , Pez Cebra/genética , Pez Cebra/metabolismo , Sodio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Homeostasis , Sueño/genética , Fosfoproteínas/metabolismo
2.
PLoS One ; 18(2): e0278316, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36757918

RESUMEN

With the oncoming age of big data, biologists are encountering more use cases for cloud-based computing to streamline data processing and storage. Unfortunately, cloud platforms are difficult to learn, and there are few resources for biologists to demystify them. We have developed a guide for experimental biologists to set up cloud processing on Amazon Web Services to cheaply outsource data processing and storage. Here we provide a guide for setting up a computing environment in the cloud and showcase examples of using Python and Julia programming languages. We present example calcium imaging data in the zebrafish brain and corresponding analysis using suite2p software. Tools for budget and user management are further discussed in the attached protocol. Using this guide, researchers with limited coding experience can get started with cloud-based computing or move existing coding infrastructure into the cloud environment.


Asunto(s)
Programas Informáticos , Pez Cebra , Animales , Lenguajes de Programación , Almacenamiento y Recuperación de la Información , Nube Computacional
4.
Front Mol Neurosci ; 15: 873520, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35465097

RESUMEN

Sleep disorders and chronic sleep disturbances are common and are associated with cardio-metabolic diseases and neuropsychiatric disorders. Several genetic pathways and neuronal mechanisms that regulate sleep have been described in animal models, but the genes underlying human sleep variation and sleep disorders are largely unknown. Identifying these genes is essential in order to develop effective therapies for sleep disorders and their associated comorbidities. To address this unmet health problem, genome-wide association studies (GWAS) have identified numerous genetic variants associated with human sleep traits and sleep disorders. However, in most cases, it is unclear which gene is responsible for a sleep phenotype that is associated with a genetic variant. As a result, it is necessary to experimentally validate candidate genes identified by GWAS using an animal model. Rodents are ill-suited for this endeavor due to their poor amenability to high-throughput sleep assays and the high costs associated with generating, maintaining, and testing large numbers of mutant lines. Zebrafish (Danio rerio), an alternative vertebrate model for studying sleep, allows for the rapid and cost-effective generation of mutant lines using the CRISPR/Cas9 system. Numerous zebrafish mutant lines can then be tested in parallel using high-throughput behavioral assays to identify genes whose loss affects sleep. This process identifies a gene associated with each GWAS hit that is likely responsible for the human sleep phenotype. This strategy is a powerful complement to GWAS approaches and holds great promise to identify the genetic basis for common human sleep disorders.

5.
Mol Psychiatry ; 27(9): 3777-3793, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35484242

RESUMEN

Salient sensory stimuli are perceived by the brain, which guides both the timing and outcome of behaviors in a context-dependent manner. Light is such a stimulus, which is used in treating mood disorders often associated with a dysregulated hypothalamic-pituitary-adrenal stress axis. Relationships between the emotional valence of light and the hypothalamus, and how they interact to exert brain-wide impacts remain unclear. Employing larval zebrafish with analogous hypothalamic systems to mammals, we show in free-swimming animals that hypothalamic corticotropin releasing factor (CRFHy) neurons promote dark avoidance, and such role is not shared by other hypothalamic peptidergic neurons. Single-neuron projection analyses uncover processes extended by individual CRFHy neurons to multiple targets including sensorimotor and decision-making areas. In vivo calcium imaging uncovers a complex and heterogeneous response of individual CRFHy neurons to the light or dark stimulus, with a reduced overall sum of CRF neuronal activity in the presence of light. Brain-wide calcium imaging under alternating light/dark stimuli further identifies distinct and distributed photic response neuronal types. CRFHy neuronal ablation increases an overall representation of light in the brain and broadly enhances the functional connectivity associated with an exploratory brain state. These findings delineate brain-wide photic perception, uncover a previously unknown role of CRFHy neurons in regulating the perception and emotional valence of light, and suggest that light therapy may alleviate mood disorders through reducing an overall sum of CRF neuronal activity.


Asunto(s)
Hormona Liberadora de Corticotropina , Núcleo Hipotalámico Paraventricular , Animales , Hormona Liberadora de Corticotropina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Calcio , Pez Cebra/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Encéfalo/metabolismo , Percepción , Mamíferos/metabolismo
6.
Bio Protoc ; 12(3): e4313, 2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-35284597

RESUMEN

Over the past decade, zebrafish have emerged as a powerful model for the study of vertebrate sleep and wake behaviors. Experimental evidence has demonstrated behavioral, anatomical, genetic, and pharmacological conservation of sleep between zebrafish and mammals, suggesting that discoveries in zebrafish can inform our understanding of mammalian sleep. Here, we describe a protocol for performing sleep behavioral experiments in larval zebrafish, using a high-throughput video tracking system. We explain how to set up a sleep behavioral experiment and provide guidelines on how to analyze the data. Using this protocol, a typical experiment can be completed in less than five days, and this method provides a scalable platform to perform genetic and pharmacological screens in a simple and cost-effective vertebrate model. By combining high-throughput behavioral assays with several advantageous features of zebrafish, this model system provides new opportunities to make discoveries that clarify the genetic and neurological mechanisms that regulate sleep.

7.
Elife ; 92020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33337320

RESUMEN

Although several sleep-regulating neuronal populations have been identified, little is known about how they interact with each other to control sleep/wake states. We previously identified neuropeptide VF (NPVF) and the hypothalamic neurons that produce it as a sleep-promoting system (Lee et al., 2017). Here we show using zebrafish that npvf-expressing neurons control sleep via the serotonergic raphe nuclei (RN), a hindbrain structure that is critical for sleep in both diurnal zebrafish and nocturnal mice. Using genetic labeling and calcium imaging, we show that npvf-expressing neurons innervate and can activate serotonergic RN neurons. We also demonstrate that chemogenetic or optogenetic stimulation of npvf-expressing neurons induces sleep in a manner that requires NPVF and serotonin in the RN. Finally, we provide genetic evidence that NPVF acts upstream of serotonin in the RN to maintain normal sleep levels. These findings reveal a novel hypothalamic-hindbrain neuronal circuit for sleep/wake control.


Asunto(s)
Hipotálamo/fisiología , Neuronas/metabolismo , Neuropéptidos/metabolismo , Núcleos del Rafe/metabolismo , Sueño/fisiología , Animales , Vías Nerviosas/fisiología , Pez Cebra
8.
Sci Rep ; 10(1): 8352, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32415202

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

9.
Sci Rep ; 10(1): 7632, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32376893

RESUMEN

Prokineticin receptors (PROKR1 and PROKR2) are G protein-coupled receptors which control human central and peripheral reproductive processes. Importantly, allelic variants of PROKR2 in humans are associated with altered migration of GnRH neurons, resulting in congenital hypogonadotropic hypogonadism (CHH), a heterogeneous disease characterized by delayed/absent puberty and/or infertility. Although this association is established in humans, murine models failed to fully recapitulate the reproductive and olfactory phenotypes observed in patients harboring PROKR2 mutations. Here, taking advantage of zebrafish model we investigated the role of prokr1b (ortholog of human PROKR2) during early stages of GnRH neuronal migration. Real-Time PCR and whole mount in situ hybridization assays indicate that prokr1b spatial-temporal expression is consistent with gnrh3. Moreover, knockdown and knockout of prokr1b altered the correct development of GnRH3 fibers, a phenotype that is rescued by injection of prokr1b mRNA. These results suggest that prokr1b regulates the development of the GnRH3 system in zebrafish. Analysis of gonads development and mating experiments indicate that prokr1b is not required for fertility in zebrafish, although its loss determine changes also at the testis level. Altogether, our results support the thesis of a divergent evolution in the control of vertebrate reproduction and provide a useful in vivo model for deciphering the mechanisms underlying the effect of PROKR2 allelic variants on CHH.

10.
Curr Biol ; 30(9): 1639-1648.e3, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32169212

RESUMEN

Sleep is a cross-species phenomenon whose evolutionary and biological function remain poorly understood. Clinical and animal studies suggest that sleep disturbance is significantly associated with disruptions in protein homeostasis-or proteostasis-in the brain, but the mechanism of this link has not been explored. In the cell, the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) pathway modulates proteostasis by transiently inhibiting protein synthesis in response to proteostatic stress. In this study, we examined the role of the PERK pathway in sleep regulation and provide the first evidence that PERK signaling is required to regulate normal sleep in both vertebrates and invertebrates. We show that pharmacological inhibition of PERK reduces sleep in both Drosophila and zebrafish, indicating an evolutionarily conserved requirement for PERK in sleep. Genetic knockdown of PERK activity also reduces sleep in Drosophila, whereas PERK overexpression induces sleep. Finally, we demonstrate that changes in PERK signaling directly impact wake-promoting neuropeptide expression, revealing a mechanism through which proteostatic pathways can affect sleep and wake behavior. Taken together, these results demonstrate that protein synthesis pathways like PERK could represent a general mechanism of sleep and wake regulation and provide greater insight into the relationship between sleep and proteostasis.


Asunto(s)
Evolución Biológica , Sueño/genética , Sueño/fisiología , Proteínas de Pez Cebra/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Cinamatos/farmacología , Drosophila melanogaster , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Neuropéptidos/genética , Neuropéptidos/metabolismo , Transducción de Señal , Tiourea/análogos & derivados , Tiourea/farmacología , Vigilia/genética , Vigilia/fisiología , Pez Cebra , Proteínas de Pez Cebra/genética , eIF-2 Quinasa/genética
11.
Sci Adv ; 5(11): eaax4249, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31763451

RESUMEN

The genetic bases for most human sleep disorders and for variation in human sleep quantity and quality are largely unknown. Using the zebrafish, a diurnal vertebrate, to investigate the genetic regulation of sleep, we found that epidermal growth factor receptor (EGFR) signaling is necessary and sufficient for normal sleep levels and is required for the normal homeostatic response to sleep deprivation. We observed that EGFR signaling promotes sleep via mitogen-activated protein kinase/extracellular signal-regulated kinase and RFamide neuropeptide signaling and that it regulates RFamide neuropeptide expression and neuronal activity. Consistent with these findings, analysis of a large cohort of human genetic data from participants of European ancestry revealed that common variants in genes within the EGFR signaling pathway are associated with variation in human sleep quantity and quality. These results indicate that EGFR signaling and its downstream pathways play a central and ancient role in regulating sleep and provide new therapeutic targets for sleep disorders.


Asunto(s)
Receptores ErbB/genética , Regulación de la Expresión Génica , Neuropéptidos/genética , Transducción de Señal/genética , Sueño/genética , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Evolución Molecular , Quinasas MAP Reguladas por Señal Extracelular/genética , Humanos , Proteínas Quinasas Activadas por Mitógenos/genética
12.
Sci Rep ; 9(1): 12405, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31455847

RESUMEN

The circadian clock ensures that behavioral and physiological processes occur at appropriate times during the 24-hour day/night cycle, and is regulated at both the cellular and organismal levels. To identify pathways acting on intact animals, we performed a small molecule screen using a luminescent reporter of molecular circadian rhythms in zebrafish larvae. We identified both known and novel pathways that affect circadian period, amplitude and phase. Several drugs identified in the screen did not affect circadian rhythms in cultured cells derived from luminescent reporter embryos or in established zebrafish and mammalian cell lines, suggesting they act via mechanisms absent in cell culture. Strikingly, using drugs that promote or inhibit inflammation, as well as a mutant that lacks microglia, we found that inflammatory state affects circadian amplitude. These results demonstrate a benefit of performing drug screens using intact animals and provide novel targets for treating circadian rhythm disorders.


Asunto(s)
Ritmo Circadiano/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente/fisiología , Antiinflamatorios no Esteroideos/farmacología , Quinasa de la Caseína I/antagonistas & inhibidores , Quinasa de la Caseína I/metabolismo , Larva/efectos de los fármacos , Larva/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Glicina/agonistas , Receptores de Glicina/metabolismo , Taurina/farmacología , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/metabolismo
13.
Cell ; 178(4): 850-866.e26, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31398340

RESUMEN

We performed a comprehensive assessment of rare inherited variation in autism spectrum disorder (ASD) by analyzing whole-genome sequences of 2,308 individuals from families with multiple affected children. We implicate 69 genes in ASD risk, including 24 passing genome-wide Bonferroni correction and 16 new ASD risk genes, most supported by rare inherited variants, a substantial extension of previous findings. Biological pathways enriched for genes harboring inherited variants represent cytoskeletal organization and ion transport, which are distinct from pathways implicated in previous studies. Nevertheless, the de novo and inherited genes contribute to a common protein-protein interaction network. We also identified structural variants (SVs) affecting non-coding regions, implicating recurrent deletions in the promoters of DLG2 and NR3C2. Loss of nr3c2 function in zebrafish disrupts sleep and social function, overlapping with human ASD-related phenotypes. These data support the utility of studying multiplex families in ASD and are available through the Hartwell Autism Research and Technology portal.


Asunto(s)
Trastorno del Espectro Autista/genética , Predisposición Genética a la Enfermedad/genética , Linaje , Mapas de Interacción de Proteínas/genética , Animales , Niño , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Guanilato-Quinasas/genética , Humanos , Patrón de Herencia/genética , Aprendizaje Automático , Masculino , Núcleo Familiar , Regiones Promotoras Genéticas/genética , Receptores de Mineralocorticoides/genética , Factores de Riesgo , Proteínas Supresoras de Tumor/genética , Secuenciación Completa del Genoma , Pez Cebra/genética
14.
Cell Rep ; 27(10): 2871-2880.e2, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31167134

RESUMEN

Retinal waves, the spontaneous patterned neural activities propagating among developing retinal ganglion cells (RGCs), instruct the activity-dependent refinement of visuotopic maps. Although it is known that the wave is initiated successively by amacrine cells and bipolar cells, the behavior and function of glia in retinal waves remain unclear. Using multiple in vivo methods in larval zebrafish, we found that Müller glial cells (MGCs) display wave-like spontaneous activities, which start at MGC processes within the inner plexiform layer, vertically spread to their somata and endfeet, and horizontally propagate into neighboring MGCs. MGC waves depend on glutamatergic signaling derived from bipolar cells. Moreover, MGCs express both glia-specific glutamate transporters and the AMPA subtype of glutamate receptors. The AMPA receptors mediate MGC calcium activities during retinal waves, whereas the glutamate transporters modulate the occurrence of retinal waves. Thus, MGCs can sense and regulate retinal waves via AMPA receptors and glutamate transporters, respectively.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/metabolismo , Calcio/metabolismo , Células Ependimogliales/metabolismo , Ácido Glutámico/metabolismo , Neuroglía/metabolismo , Receptores AMPA/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Amacrinas/metabolismo , Células Amacrinas/fisiología , Sistema de Transporte de Aminoácidos X-AG/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos X-AG/genética , Animales , Animales Modificados Genéticamente , Células Ependimogliales/citología , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/fisiología , Ácido Glutámico/farmacología , Larva/efectos de los fármacos , Larva/metabolismo , Larva/fisiología , Neuroglía/citología , Neuroglía/fisiología , Receptores AMPA/antagonistas & inhibidores , Retina/citología , Retina/metabolismo , Retina/fisiología , Células Bipolares de la Retina/metabolismo , Células Bipolares de la Retina/fisiología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/fisiología , Pez Cebra
15.
Neuron ; 103(4): 686-701.e8, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31248729

RESUMEN

The role of serotonin (5-HT) in sleep is controversial: early studies suggested a sleep-promoting role, but eventually the paradigm shifted toward a wake-promoting function for the serotonergic raphe. Here, we provide evidence from zebrafish and mice that the raphe are critical for the initiation and maintenance of sleep. In zebrafish, genetic ablation of 5-HT production by the raphe reduces sleep, sleep depth, and the homeostatic response to sleep deprivation. Pharmacological inhibition or ablation of the raphe reduces sleep, while optogenetic stimulation increases sleep. Similarly, in mice, ablation of the raphe increases wakefulness and impairs the homeostatic response to sleep deprivation, whereas tonic optogenetic stimulation at a rate similar to baseline activity induces sleep. Interestingly, burst optogenetic stimulation induces wakefulness in accordance with previously described burst activity of the raphe during arousing stimuli. These results indicate that the serotonergic system promotes sleep in both diurnal zebrafish and nocturnal rodents. VIDEO ABSTRACT.


Asunto(s)
Ratones/fisiología , Núcleos del Rafe/fisiología , Serotonina/fisiología , Sueño/fisiología , Pez Cebra/fisiología , Animales , Nivel de Alerta/genética , Nivel de Alerta/fisiología , Buspirona/farmacología , Ritmo Circadiano/fisiología , Fenclonina/farmacología , Homeostasis , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Optogenética , Quipazina/farmacología , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/fisiología , Serotonina/biosíntesis , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología , Privación de Sueño/genética , Privación de Sueño/fisiopatología , Triptófano Hidroxilasa/deficiencia , Triptófano Hidroxilasa/genética , Vigilia/genética , Vigilia/fisiología , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
16.
Cell Rep ; 27(1): 115-128.e5, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30943395

RESUMEN

During development, oligodendrocyte progenitor cells (OPCs) migrate extensively throughout the spinal cord. However, their migration is restricted at transition zones (TZs). At these specialized locations, unique glial cells in both zebrafish and mice play a role in preventing peripheral OPC migration, but the mechanisms of this regulation are not understood. To elucidate the mechanisms that mediate OPC segregation at motor exit point (MEP) TZs, we performed an unbiased small-molecule screen. Using chemical screening and in vivo imaging, we discovered that inhibition of A2a adenosine receptors (ARs) causes ectopic OPC migration out of the spinal cord. We provide in vivo evidence that neuromodulation, partially mediated by adenosine, influences OPC migration specifically at the MEP TZ. This work opens exciting possibilities for understanding how OPCs reach their final destinations during development and identifies mechanisms that could promote their migration in disease.


Asunto(s)
Adenosina/farmacología , Movimiento Celular/efectos de los fármacos , Placa Motora/embriología , Neurotransmisores/farmacología , Oligodendroglía/efectos de los fármacos , Médula Espinal/embriología , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/fisiología , Diferenciación Celular/efectos de los fármacos , Embrión no Mamífero , Femenino , Masculino , Placa Motora/citología , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/fisiología , Oligodendroglía/fisiología , Médula Espinal/citología , Médula Espinal/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/fisiología , Pez Cebra/embriología
17.
Science ; 363(6426): 455-456, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30705172
18.
Elife ; 72018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29405117

RESUMEN

The duration of sleep varies dramatically between species, yet little is known about the genetic basis or evolutionary factors driving this variation in behavior. The Mexican cavefish, Astyanax mexicanus, exists as surface populations that inhabit rivers, and multiple cave populations with convergent evolution on sleep loss. The number of Hypocretin/Orexin (HCRT)-positive hypothalamic neurons is increased significantly in cavefish, and HCRT is upregulated at both the transcript and protein levels. Pharmacological or genetic inhibition of HCRT signaling increases sleep in cavefish, suggesting enhanced HCRT signaling underlies the evolution of sleep loss. Ablation of the lateral line or starvation, manipulations that selectively promote sleep in cavefish, inhibit hcrt expression in cavefish while having little effect on surface fish. These findings provide the first evidence of genetic and neuronal changes that contribute to the evolution of sleep loss, and support a conserved role for HCRT in sleep regulation.


Asunto(s)
Characidae/fisiología , Hipotálamo/fisiología , Orexinas/metabolismo , Sueño , Regulación hacia Arriba , Animales , Evolución Biológica , Perfilación de la Expresión Génica , México
19.
Trends Neurosci ; 41(2): 70-72, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29405929

RESUMEN

Prior to the 21st century, genetic mechanisms that regulate sleep were largely unknown. In 1998, de Lecea et al. [1] (Proc. Natl. Acad. Sci. U. S. A. 1998; 95:322-327) and Sakurai et al. [2] (Cell 1998; 92: 573-585) reported the discovery of a gene they named hypocretin and orexin, respectively, which led to a revolution in our understanding of genetic and neuronal mechanisms that regulate sleep.


Asunto(s)
Neuropéptidos/metabolismo , Orexinas/farmacología , Sueño/efectos de los fármacos , Vigilia/efectos de los fármacos , Animales , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuronas/efectos de los fármacos , Sueño/fisiología , Vigilia/fisiología
20.
Curr Biol ; 27(24): 3796-3811.e5, 2017 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-29225025

RESUMEN

Sleep is an essential and evolutionarily conserved behavioral state whose regulation remains poorly understood. To identify genes that regulate vertebrate sleep, we recently performed a genetic screen in zebrafish, and here we report the identification of neuropeptide Y (NPY) as both necessary for normal daytime sleep duration and sufficient to promote sleep. We show that overexpression of NPY increases sleep, whereas mutation of npy or ablation of npy-expressing neurons decreases sleep. By analyzing sleep architecture, we show that NPY regulates sleep primarily by modulating the length of wake bouts. To determine how NPY regulates sleep, we tested for interactions with several systems known to regulate sleep, and provide anatomical, molecular, genetic, and pharmacological evidence that NPY promotes sleep by inhibiting noradrenergic signaling. These data establish NPY as an important vertebrate sleep/wake regulator and link NPY signaling to an established arousal-promoting system.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Proteínas de Peces/metabolismo , Neuropéptido Y/metabolismo , Transducción de Señal , Sueño/fisiología , Vigilia/fisiología , Pez Cebra/fisiología , Animales , Sueño/genética , Vigilia/genética , Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...