Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Curr Opin Neurobiol ; 86: 102874, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38582021

RESUMO

The metabolic signals that regulate sleep and the metabolic functions that occur during sleep are active areas of research. Prior studies have focused on sugars and nucleotides but new genetic evidence suggests novel functions of lipid and amino acid metabolites in sleep. Additional genetic studies of energetic signaling pathways and the circadian clock transcription factor network have increased our understanding of how sleep responds to changes in the metabolic state. This review focuses on key recent insights from genetic experiments in humans and model organisms to improve our understanding of the interrelationship between metabolism and sleep.


Assuntos
Sono , Humanos , Sono/fisiologia , Sono/genética , Animais , Metabolismo Energético/fisiologia , Metabolismo Energético/genética , Relógios Circadianos/fisiologia , Relógios Circadianos/genética , Ritmo Circadiano/fisiologia , Ritmo Circadiano/genética
2.
Artigo em Inglês | MEDLINE | ID: mdl-38669479

RESUMO

Healthy sleep is vital for humans to achieve optimal health and longevity. Poor sleep and sleep disorders are strongly associated with increased morbidity and mortality. However, the importance of good sleep continues to be underrecognized. Mechanisms regulating sleep and its functions in humans remain mostly unclear even after decades of dedicated research. Advancements in gene sequencing techniques and computational methodologies have paved the way for various genetic analysis approaches, which have provided some insights into human sleep genetics. This review summarizes our current knowledge of the genetic basis underlying human sleep traits and sleep disorders. We also highlight the use of animal models to validate genetic findings from human sleep studies and discuss potential molecular mechanisms and signaling pathways involved in the regulation of human sleep.

3.
bioRxiv ; 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38352599

RESUMO

The circadian clock synchronizes metabolic and behavioral cycles with the rotation of the Earth by integrating environmental cues, such as light. Nutrient content also regulates the clock, though how and why this environmental signal affects the clock remains incompletely understood. Here, we elucidate a role for nutrient in regulating circadian alignment to seasonal photoperiods. High fat diet (HFD) promoted entrainment to a summer light cycle and inhibited entrainment to a winter light cycle by phosphorylating PER2 on serine 662. PER2-S662 phospho-mimetic mutant mice were incapable of entraining to a winter photoperiod, while PER2-S662 phospho-null mutant mice were incapable of entraining to a summer photoperiod, even in the presence of HFD. Multi-omic experimentation in conjunction with isocaloric hydrogenated-fat feeding, revealed a role for polyunsaturated fatty acids in nutrient-dependent seasonal entrainment. Altogether, we identify the mechanism whereby nutrient content shifts circadian rhythms to anticipate seasonal photoperiods in which that nutrient state predominates. HIGHLIGHTS: High fat diet promotes entrainment to summer but inhibits entrainment to winter.Calorie restriction promotes entrainment to winter but inhibits entrainment to summer.PER2-S662 phosphorylation is required for nutritional regulation of seasonal circadian entrainment.Dietary polyunsaturated fatty acids regulate seasonal circadian entrainment.

4.
Proc Natl Acad Sci U S A ; 121(9): e2320276121, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38381789

RESUMO

Neuropeptide S (NPS) was postulated to be a wake-promoting neuropeptide with unknown mechanism, and a mutation in its receptor (NPSR1) causes the short sleep duration trait in humans. We investigated the role of different NPS+ nuclei in sleep/wake regulation. Loss-of-function and chemogenetic studies revealed that NPS+ neurons in the parabrachial nucleus (PB) are wake-promoting, whereas peri-locus coeruleus (peri-LC) NPS+ neurons are not important for sleep/wake modulation. Further, we found that a NPS+ nucleus in the central gray of the pons (CGPn) strongly promotes sleep. Fiber photometry recordings showed that NPS+ neurons are wake-active in the CGPn and wake/REM-sleep active in the PB and peri-LC. Blocking NPS-NPSR1 signaling or knockdown of Nps supported the function of the NPS-NPSR1 pathway in sleep/wake regulation. Together, these results reveal that NPS and NPS+ neurons play dichotomous roles in sleep/wake regulation at both the molecular and circuit levels.


Assuntos
Neuropeptídeos , Sono , Humanos , Sono/fisiologia , Ponte/fisiologia , Locus Cerúleo/fisiologia , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
5.
Mov Disord ; 39(3): 486-497, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38197134

RESUMO

BACKGROUND: Spinocerebellar ataxia type 4 (SCA4) is an autosomal dominant ataxia with invariable sensory neuropathy originally described in a family with Swedish ancestry residing in Utah more than 25 years ago. Despite tight linkage to the 16q22 region, the molecular diagnosis has since remained elusive. OBJECTIVES: Inspired by pathogenic structural variation implicated in other 16q-ataxias with linkage to the same locus, we revisited the index SCA4 cases from the Utah family using novel technologies to investigate structural variation within the candidate region. METHODS: We adopted a targeted long-read sequencing approach with adaptive sampling on the Oxford Nanopore Technologies (ONT) platform that enables the detection of segregating structural variants within a genomic region without a priori assumptions about any variant features. RESULTS: Using this approach, we found a heterozygous (GGC)n repeat expansion in the last coding exon of the zinc finger homeobox 3 (ZFHX3) gene that segregates with disease, ranging between 48 and 57 GGC repeats in affected probands. This finding was replicated in a separate family with SCA4. Furthermore, the estimation of this GGC repeat size in short-read whole genome sequencing (WGS) data of 21,836 individuals recruited to the 100,000 Genomes Project in the UK and our in-house dataset of 11,258 exomes did not reveal any pathogenic repeats, indicating that the variant is ultrarare. CONCLUSIONS: These findings support the utility of adaptive long-read sequencing as a powerful tool to decipher causative structural variation in unsolved cases of inherited neurological disease. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Assuntos
Ataxia Cerebelar , Ataxias Espinocerebelares , Humanos , Linhagem , Ataxias Espinocerebelares/genética , Ataxia Cerebelar/genética , Éxons , Proteínas de Homeodomínio/genética
6.
Proc Natl Acad Sci U S A ; 120(15): e2221686120, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-37014857

RESUMO

Sleep is essential for our well-being, and chronic sleep deprivation has unfavorable health consequences. We recently demonstrated that two familial natural short sleep (FNSS) mutations, DEC2-P384R and Npsr1-Y206H, are strong genetic modifiers of tauopathy in PS19 mice, a model of tauopathy. To gain more insight into how FNSS variants modify the tau phenotype, we tested the effect of another FNSS gene variant, Adrb1-A187V, by crossing mice with this mutation onto the PS19 background. We found that the Adrb1-A187V mutation helped restore rapid eye movement (REM) sleep and alleviated tau aggregation in a sleep-wake center, the locus coeruleus (LC), in PS19 mice. We found that ADRB1+ neurons in the central amygdala (CeA) sent projections to the LC, and stimulating CeAADRB1+ neuron activity increased REM sleep. Furthermore, the mutant Adrb1 attenuated tau spreading from the CeA to the LC. Our findings suggest that the Adrb1-A187V mutation protects against tauopathy by both mitigating tau accumulation and attenuating tau spreading.


Assuntos
Transtornos do Sono-Vigília , Tauopatias , Camundongos , Animais , Sono REM , Tauopatias/genética , Sono/fisiologia , Locus Cerúleo/metabolismo , Receptores Adrenérgicos , Proteínas tau/genética , Proteínas tau/metabolismo , Camundongos Transgênicos , Modelos Animais de Doenças
7.
Cereb Cortex ; 33(8): 4293-4304, 2023 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-36030380

RESUMO

Neocortical vasoactive intestinal polypeptide-expressing (VIP+) interneurons display highly diverse morpho-electrophysiological and molecular properties. To begin to understand the function of VIP+ interneurons in cortical circuits, they must be clearly and comprehensively classified into distinct subpopulations based on specific molecular markers. Here, we utilized patch-clamp RT-PCR (Patch-PCR) to simultaneously obtain the morpho-electric properties and mRNA profiles of 155 VIP+ interneurons in layers 2 and 3 (L2/3) of the mouse somatosensory cortex. Using an unsupervised clustering method, we identified 3 electrophysiological types (E-types) and 2 morphological types (M-types) of VIP+ interneurons. Joint clustering based on the combined electrophysiological and morphological features resulted in 3 morpho-electric types (ME-types). More importantly, we found these 3 ME-types expressed distinct marker genes: ~94% of Sncg+ cells were ME-type 1, 100% of Mybpc1+ cells were ME-type 2, and ~78% of Parm1+ were ME-type 3. By clarifying the properties of subpopulations of cortical L2/3 VIP+ interneurons, this study establishes a basis for future investigations aiming to elucidate their physiological roles.


Assuntos
Córtex Somatossensorial , Peptídeo Intestinal Vasoativo , Animais , Camundongos , Fenômenos Eletrofisiológicos , Interneurônios/fisiologia , Córtex Somatossensorial/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Proteínas de Neoplasias/metabolismo , gama-Sinucleína/metabolismo , Proteína de Ligação a Androgênios/metabolismo
8.
Proc Natl Acad Sci U S A ; 119(34): e2203266119, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-35901245

RESUMO

Sleep is a necessity for our survival, but its regulation remains incompletely understood. Here, we used a human sleep duration gene to identify a population of cells in the peri-tegmental reticular nucleus (pTRNADRB1) that regulate sleep-wake, uncovering a role for a poorly understood brain area. Although initial ablation in mice led to increased wakefulness, further validation revealed that pTRNADRB1 neuron stimulation strongly promotes wakefulness, even after stimulation offset. Using combinatorial genetics, we found that excitatory pTRNADRB1 neurons promote wakefulness. pTRN neurons can be characterized as anterior- or posterior-projecting neurons based on multiplexed analysis of projections by sequencing (MAPseq) analysis. Finally, we found that pTRNADRB1 neurons promote wakefulness, in part, through projections to the lateral hypothalamus. Thus, human genetic information from a human sleep trait allowed us to identify a role for the pTRN in sleep-wake regulation.


Assuntos
Sono , Tegmento Mesencefálico , Vigília , Animais , Humanos , Região Hipotalâmica Lateral/fisiologia , Camundongos , Neurônios/fisiologia , Sono/fisiologia , Tegmento Mesencefálico/fisiologia , Vigília/fisiologia
9.
iScience ; 25(4): 103964, 2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-35496999

RESUMO

Although numerous studies have demonstrated that poor sleep increases the development of AD, direct evidence elucidating the benefits of good sleep on the AD pathogenesis is lacking. Familial Natural Short Sleepers (FNSS) are genetically wired to have lifelong reduction in nightly sleep duration without evident consequence on cognitive demise, implying that they may have better sleep quality. Here we investigated two FNSS mutations, DEC2-P384R and Npsr1-Y206H, on the development of tau and amyloid pathology in AD-like mouse models. We found that the development of tau pathology is attenuated in the hippocampus of tau mice carrying FNSS mutations. We also found that DEC2-P384R;5XFAD and female Npsr1-Y206H;5XFAD mice exhibit significantly less amyloid plaques than control mice at 6 months of age. Together, these results reveal that these two FNSS alleles are strong genetic modifiers of AD pathology and may confer resilience to the progression of tau pathology and amyloid plaque formation in neurodegeneration.

10.
Int J Ophthalmol ; 15(2): 228-232, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35186681

RESUMO

AIM: To evaluate the accuracy and predictability of ray tracing-assisted intraocular lens (IOL) calculation function in Sirius internal software and further improve the accuracy by optimizing the calculation of predicted lens position (PLP). METHODS: This retrospective study recruited 52 eyes of 49 patients. All of the cases with cataract had undergone phacoemulsification combined with IOL implantation. SRK-T, Haigis formula, and Sirius ray-tracing method were all used for each eye's IOL calculation. The mean absolute value of prediction error (prediction error=predicted refraction-postoperative refraction) was defined as mean absolute prediction error (MAPE) and was determined for each method. Calculation of PLP was optimized by effective lens position (ELP). Optimized PLP was entered to Sirius internal software again to verify whether the method was improved. RESULTS: Compared with SRK-T and Haigis formulas, less accuracy was shown in Sirius ray-tracing method (P=0.001). The ELP of the IOL moved forward compared to PLP (P<0.001). The MAPE of the ELP-inputted Sirius ray-tracing method was reduced. ELP and PLP were well correlated. Taking ELP as y and PLP given by Sirius soft as x, a linear regression formula y=0.1637x+3.1741 was concluded (R2 =0.1066, P=0.018). It was shown that the optimized Sirius ray-tracing method (optimized PLP entered), compared with SRK-T and Haigis formulas, worked with the same accuracy (P=0.038). CONCLUSION: The original Sirius ray tracing method is not satisfactory enough. However, in normal eyes, the optimized Sirius ray-tracing method in IOL calculation was as accurate as SRK-T and Haigis formulas.

11.
Neurobiol Sleep Circadian Rhythms ; 12: 100073, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35028489

RESUMO

Sleep deprivation can generate inflammatory responses in the central nervous system. In turn, this inflammation increases sleep drive, leading to a rebound in sleep duration. Microglia, the innate immune cells found exclusively in the CNS, have previously been found to release inflammatory signals and exhibit altered characteristics in response to sleep deprivation. Together, this suggests that microglia may be partially responsible for the brain's response to sleep deprivation through their inflammatory activity. In this study, we ablated microglia from the mouse brain and assessed resulting sleep, circadian, and sleep deprivation phenotypes. We find that microglia are dispensable for both homeostatic sleep and circadian function and the sleep rebound response to sleep deprivation. However, we uncover a phenomenon by which microglia appear to be essential for the protection of fear-conditioning memories formed during the recovery sleep period following a period of sleep deprivation. This phenomenon occurs potentially through the upregulation of synaptic-homeostasis related genes to protect nascent dendritic spines that may be otherwise removed or downscaled during recovery sleep. These findings further expand the list of known functions for microglia in synaptic modulation.

12.
J Clin Invest ; 131(16)2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34396981

RESUMO

Circadian rhythms, present in most phyla across life, are biological oscillations occurring on a daily cycle. Since the discovery of their molecular foundations in model organisms, many inputs that modify this tightly controlled system in humans have been identified. Polygenic variations and environmental factors influence each person's circadian rhythm, contributing to the trait known as chronotype, which manifests as the degree of morning or evening preference in an individual. Despite normal variation in chronotype, much of society operates on a "one size fits all" schedule that can be difficult to adjust to, especially for certain individuals whose endogenous circadian phase is extremely advanced or delayed. This is a public health concern, as phase misalignment in humans is associated with a number of adverse health outcomes. Additionally, modern technology (such as electric lights and computer, tablet, and phone screens that emit blue light) and lifestyles (such as shift or irregular work schedules) are disrupting circadian consistency in an increasing number of people. Though medical and lifestyle interventions can alleviate some of these issues, growing research on endogenous circadian variability and sensitivity suggests that broader social changes may be necessary to minimize the impact of circadian misalignment on health.


Assuntos
Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Sono/genética , Sono/fisiologia , Humanos , Síndrome do Jet Lag/etiologia , Transtornos do Sono do Ritmo Circadiano/etiologia , Transtornos do Sono do Ritmo Circadiano/genética , Transtornos do Sono do Ritmo Circadiano/fisiopatologia , Tolerância ao Trabalho Programado/fisiologia
13.
J Clin Invest ; 131(2)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33463539
14.
Curr Opin Neurobiol ; 69: 19-24, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33360546

RESUMO

Sleep regulation has a strong genetic component. In this review, we highlight the recent advances in sleep genetics from knockout, point mutation, and GWAS studies. We overview specific genetic effects on REM versus NREM sleep as well as how the implicated genes fall in broad functional categories. Furthermore, we elucidate how genes affect different aspects of sleep including sleep duration, sleep consolidation, recovery sleep, and the circadian timing of sleep, demonstrating that genetic studies can be powerful in understanding how the body regulates sleep.


Assuntos
Sono REM , Sono , Ritmo Circadiano/genética , Eletroencefalografia , Sono/genética
15.
Curr Biol ; 31(1): 13-24.e4, 2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33065013

RESUMO

Sufficient and efficient sleep is crucial for our health. Natural short sleepers can sleep significantly shorter than the average population without a desire for more sleep and without any obvious negative health consequences. In searching for genetic variants underlying the short sleep trait, we found two different mutations in the same gene (metabotropic glutamate receptor 1) from two independent natural short sleep families. In vitro, both of the mutations exhibited loss of function in receptor-mediated signaling. In vivo, the mice carrying the individual mutations both demonstrated short sleep behavior. In brain slices, both of the mutations changed the electrical properties and increased excitatory synaptic transmission. These results highlight the important role of metabotropic glutamate receptor 1 in modulating sleep duration.


Assuntos
Receptores de Glutamato Metabotrópico/genética , Sono/genética , Animais , Análise Mutacional de DNA , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Hipocampo/fisiologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Mutação , Plasticidade Neuronal/fisiologia , Técnicas de Patch-Clamp , Linhagem , Polissonografia , Receptores de Glutamato Metabotrópico/metabolismo , Fatores de Tempo , Sequenciamento do Exoma
16.
Ann Neurol ; 88(4): 830-842, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32715519

RESUMO

OBJECTIVE: The objective of this study was to identify the genetic cause for progressive peripheral nerve disease in a Venezuelan family. Despite the growing list of genes associated with Charcot-Marie-Tooth disease, many patients with axonal forms lack a genetic diagnosis. METHODS: A pedigree was constructed, based on family clinical data. Next-generation sequencing of mitochondrial DNA (mtDNA) was performed for 6 affected family members. Muscle biopsies from 4 family members were used for analysis of muscle histology and ultrastructure, mtDNA sequencing, and RNA quantification. Ultrastructural studies were performed on sensory nerve biopsies from 2 affected family members. RESULTS: Electrodiagnostic testing showed a motor and sensory axonal polyneuropathy. Pedigree analysis revealed inheritance only through the maternal line, consistent with mitochondrial transmission. Sequencing of mtDNA identified a mutation in the mitochondrial tRNAVal (mt-tRNAVal ) gene, m.1661A>G, present at nearly 100% heteroplasmy, which disrupts a Watson-Crick base pair in the T-stem-loop. Muscle biopsies showed chronic denervation/reinnervation changes, whereas biochemical analysis of electron transport chain (ETC) enzyme activities showed reduction in multiple ETC complexes. Northern blots from skeletal muscle total RNA showed severe reduction in abundance of mt-tRNAVal , and mildly increased mt-tRNAPhe , in subjects compared with unrelated age- and sex-matched controls. Nerve biopsies from 2 affected family members demonstrated ultrastructural mitochondrial abnormalities (hyperplasia, hypertrophy, and crystalline arrays) consistent with a mitochondrial neuropathy. CONCLUSION: We identify a previously unreported cause of Charcot-Marie-Tooth (CMT) disease, a mutation in the mt-tRNAVal , in a Venezuelan family. This work expands the list of CMT-associated genes from protein-coding genes to a mitochondrial tRNA gene. ANN NEUROL 2020;88:830-842.


Assuntos
Doença de Charcot-Marie-Tooth/genética , RNA Mitocondrial/genética , RNA de Transferência/genética , Adolescente , Adulto , Idoso de 80 Anos ou mais , Criança , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , Venezuela , Adulto Jovem
17.
Neuropsychopharmacology ; 45(1): 45-54, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31400754

RESUMO

Timing and duration of sleep are controlled by the circadian system, which keeps an ~24-h internal rhythm that entrains to environmental stimuli, and the sleep homeostat, which rises as a function of time awake. There is a normal distribution across the population in how the circadian system aligns with typical day and night resulting in varying circadian preferences called chronotypes. A portion of the variation in the population is controlled by genetics as shown by the single-gene mutations that confer extreme early or late chronotypes. Similarly, there is a normal distribution across the population in sleep duration. Genetic variations have been identified that lead to a short sleep phenotype in which individuals sleep only 4-6.5 h nightly. Negative health consequences have been identified when individuals do not sleep at their ideal circadian timing or are sleep deprived relative to intrinsic sleep need. Whether familial natural short sleepers are at risk of the health consequences associated with a short sleep duration based on population data is not known. More work needs to be done to better assess for an individual's chronotype and degree of sleep deprivation to answer these questions.


Assuntos
Relógios Circadianos/genética , Ritmo Circadiano/genética , Homeostase/genética , Transtornos do Sono-Vigília/genética , Sono/genética , Humanos , Transtornos do Sono-Vigília/fisiopatologia
18.
Eur J Neurosci ; 51(1): 422-428, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-30144347

RESUMO

It has been known for many years that genetic influences account for some of the individual differences in human sleep parameters, but the underlying molecular mechanisms remain unclear. With major advances of molecular biology and the recognition of heritable sleep behaviors in humans over the past 30 years, a number of genetic variants have been identified to be associated with human sleep timing, duration and quality, both in healthy individuals and under pathological conditions. Some of these variants were further validated and characterized in animal models, shedding light on the mechanism of how these variants likely alter sleep in humans, which may provide new insights into developing more effective treatments to improve human sleep.


Assuntos
Transtornos do Sono-Vigília , Sono , Animais , Ritmo Circadiano , Humanos , Individualidade , Modelos Animais , Biologia Molecular , Sono/genética
19.
Sci Transl Med ; 11(514)2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31619542

RESUMO

Sleep is a crucial physiological process for our survival and cognitive performance, yet the factors controlling human sleep regulation remain poorly understood. Here, we identified a missense mutation in a G protein-coupled neuropeptide S receptor 1 (NPSR1) that is associated with a natural short sleep phenotype in humans. Mice carrying the homologous mutation exhibited less sleep time despite increased sleep pressure. These animals were also resistant to contextual memory deficits associated with sleep deprivation. In vivo, the mutant receptors showed increased sensitivity to neuropeptide S exogenous activation. These results suggest that the NPS/NPSR1 pathway might play a critical role in regulating human sleep duration and in the link between sleep homeostasis and memory consolidation.


Assuntos
Consolidação da Memória/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Sono/fisiologia , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação/genética , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase em Tempo Real , Receptores Acoplados a Proteínas G/genética , Sono/genética
20.
Neuron ; 103(6): 1044-1055.e7, 2019 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-31473062

RESUMO

Sleep is crucial for our survival, and many diseases are linked to long-term poor sleep quality. Before we can use sleep to enhance our health and performance and alleviate diseases associated with poor sleep, a greater understanding of sleep regulation is necessary. We have identified a mutation in the ß1-adrenergic receptor gene in humans who require fewer hours of sleep than most. In vitro, this mutation leads to decreased protein stability and dampened signaling in response to agonist treatment. In vivo, the mice carrying the same mutation demonstrated short sleep behavior. We found that this receptor is highly expressed in the dorsal pons and that these ADRB1+ neurons are active during rapid eye movement (REM) sleep and wakefulness. Activating these neurons can lead to wakefulness, and the activity of these neurons is affected by the mutation. These results highlight the important role of ß1-adrenergic receptors in sleep/wake regulation.


Assuntos
Receptores Adrenérgicos beta 1/genética , Sono/genética , Vigília/genética , Animais , Técnicas de Introdução de Genes , Humanos , Camundongos , Mutação , Neurônios/metabolismo , Linhagem , Tegmento Pontino/citologia , Tegmento Pontino/metabolismo , Transtornos do Sono-Vigília/genética , Sono REM/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...