Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.093
Filtrar
1.
Ren Fail ; 46(2): 2387430, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39132818

RESUMEN

OBJECTIVES: Extensive researches highlight the detrimental impact of sleep disorders such as insomnia and insufficient sleep duration on kidney function. However, establishing a clear causal relationship between insomnia, sleep duration, and kidney function remains challenging. This study aims to estimate this relationship using Mendelian randomization (MR). METHODS: Independent genetic variants strongly associated with insomnia (N = 462,341) and sleep duration (N = 460,099) were selected as instrumental variables from corresponding genome-wide association studies (GWAS). Kidney function parameters, including serum creatinine, estimated glomerular filtration rate by cystatin C (eGFRcys), acute renal failure (ARF), chronic renal failure (CRF), kidney injury molecule-1, neutrophil gelatinase associated lipocalin, microalbuminuria, cystatin C, and ß2 microglobulin, were derived from GWAS databases. A two-sample MR study was conducted to assess the causal relationship between sleep disorders and kidney function, and multivariable MR was used to identify potential mediators. The inverse-variance weighted was used as the primary estimate. RESULTS: MR analysis found robust evidence indicating that insomnia and short sleep duration were associated with an increased risk of elevated serum creatinine, regardless of adjusting for obesity. Causal links between sleep duration and eGFRcys or cystatin C were also identified. While genetically predicted insomnia and sleep duration were found to potentially impact ARF, CRF, microalbuminuria, and ß2 microglobulin, the p-values in multivariable MR analysis became nonsignificant. No pleiotropy was detected. CONCLUSIONS: This study demonstrates a causal impact of insomnia on the risk of elevated serum creatinine and a positive effect of sleep duration on serum creatinine, eGFRcys, and cystatin C. Our findings also suggest their potential indirect effects on ARF, CRF, microalbuminuria, and ß2 microglobulin mediated by obesity.


Asunto(s)
Creatinina , Estudio de Asociación del Genoma Completo , Tasa de Filtración Glomerular , Análisis de la Aleatorización Mendeliana , Trastornos del Inicio y del Mantenimiento del Sueño , Sueño , Humanos , Trastornos del Inicio y del Mantenimiento del Sueño/genética , Creatinina/sangre , Sueño/genética , Cistatina C/sangre , Cistatina C/genética , Riñón/fisiopatología , Lesión Renal Aguda/genética , Lesión Renal Aguda/etiología , Polimorfismo de Nucleótido Simple , Duración del Sueño
2.
Int J Mol Sci ; 25(14)2024 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-39062963

RESUMEN

Parkinson's disease (PD) is characterized by a long prodromal period, during which patients often have sleep disturbances. The histaminergic system and circadian rhythms play an important role in the regulation of the sleep-wake cycle. Changes in the functioning of these systems may be involved in the pathogenesis of early stages of PD and may be age-dependent. Here, we have analyzed changes in the expression of genes associated with the regulation of the sleep-wake cycle (Hnmt, Hrh1, Hrh3, Per1, Per2, and Chrm3) in the substantia nigra (SN) and striatum of normal male mice of different ages, as well as in young and adult male mice with an MPTP-induced model of the early symptomatic stage (ESS) of PD. Age-dependent expression analysis in normal mouse brain tissue revealed changes in Hrh3, Per1, Per2, and Chrm3 genes in adult mice relative to young mice. When gene expression was examined in mice with the MPTP-induced model of the ESS of PD, changes in the expression of all studied genes were found only in the SN of adult mice with the ESS model of PD. These data suggest that age is a significant factor influencing changes in the expression of genes associated with sleep-wake cycle regulation in the development of PD.


Asunto(s)
Ritmo Circadiano , Animales , Ratones , Masculino , Ritmo Circadiano/genética , Regulación de la Expresión Génica , Ratones Endogámicos C57BL , Sueño/genética , Envejecimiento/genética , Modelos Animales de Enfermedad , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Sustancia Negra/metabolismo , Cuerpo Estriado/metabolismo , Intoxicación por MPTP/genética , Intoxicación por MPTP/metabolismo , Factores de Edad , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Vigilia
3.
Nature ; 632(8023): 139-146, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38961289

RESUMEN

Brain computation performed by billions of nerve cells relies on a sufficient and uninterrupted nutrient and oxygen supply1,2. Astrocytes, the ubiquitous glial neighbours of neurons, govern brain glucose uptake and metabolism3,4, but the exact mechanisms of metabolic coupling between neurons and astrocytes that ensure on-demand support of neuronal energy needs are not fully understood5,6. Here we show, using experimental in vitro and in vivo animal models, that neuronal activity-dependent metabolic activation of astrocytes is mediated by neuromodulator adenosine acting on astrocytic A2B receptors. Stimulation of A2B receptors recruits the canonical cyclic adenosine 3',5'-monophosphate-protein kinase A signalling pathway, leading to rapid activation of astrocyte glucose metabolism and the release of lactate, which supplements the extracellular pool of readily available energy substrates. Experimental mouse models involving conditional deletion of the gene encoding A2B receptors in astrocytes showed that adenosine-mediated metabolic signalling is essential for maintaining synaptic function, especially under conditions of high energy demand or reduced energy supply. Knockdown of A2B receptor expression in astrocytes led to a major reprogramming of brain energy metabolism, prevented synaptic plasticity in the hippocampus, severely impaired recognition memory and disrupted sleep. These data identify the adenosine A2B receptor as an astrocytic sensor of neuronal activity and show that cAMP signalling in astrocytes tunes brain energy metabolism to support its fundamental functions such as sleep and memory.


Asunto(s)
Adenosina , Astrocitos , Encéfalo , Metabolismo Energético , Neuronas , Transducción de Señal , Animales , Femenino , Masculino , Ratones , Ratas , Adenosina/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Encéfalo/citología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glucosa/metabolismo , Hipocampo/metabolismo , Hipocampo/citología , Ácido Láctico/metabolismo , Ratones Endogámicos C57BL , Plasticidad Neuronal , Neuronas/metabolismo , Receptor de Adenosina A2B/deficiencia , Receptor de Adenosina A2B/efectos de los fármacos , Receptor de Adenosina A2B/genética , Receptor de Adenosina A2B/metabolismo , Reconocimiento en Psicología/fisiología , Sueño/genética , Sueño/fisiología , Sinapsis/metabolismo
4.
Sci Rep ; 14(1): 17746, 2024 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-39085554

RESUMEN

Globally, breast cancer continues to be the leading cause of cancer-related incidence and mortality among females. Research has shown that sleep patterns significantly influence tumor onset and progression. In this research, the association was examined through the application of a two-sample Mendelian randomization (MR) approach. For the analysis of seven sleep patterns, genetic tools were sourced from both the UK Biobank and 23andMe, including morning/evening person (chronotype) n = 177,604, morning person (chronotype) n = 248,094, daytime dozing/sleepiness n = 193,472, getting up in the morning n = 193,717, and sleeplessness n = 193,987; sleep duration n = 192,810; and nap during the day n = 166,853. The Breast Cancer Association Consortium (BCAC) supplied genome-wide association studies (GWAS) data, including 133,384 breast cancer cases and 113,789 controls, alongside subtype-specific data with 106,278 cases and 91,477 controls. We discovered that chronotype encompasses both morning and evening types contributes to the risk of overall breast cancer. While daytime dozing and morning person (chronotype) are linked to a lower risk of breast cancer in general, In subtype-specific analyses, morning person (chronotype) was negatively associated with luminal B, HER2-negative-like, and daytime dozing was negatively correlated with luminal A-like, luminal B-like, and HER2-enriched-like. The study corroborates that chronotype is a danger element for breast cancer, aligning with previous observational findings. The association between being a morning person (chronotype) or having daytime dozing and a decreased risk of breast cancer underscores the significance of sleep patterns in formulating strategies for cancer prevention.


Asunto(s)
Neoplasias de la Mama , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Sueño , Humanos , Neoplasias de la Mama/genética , Neoplasias de la Mama/epidemiología , Femenino , Sueño/genética , Factores de Riesgo , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple
5.
J Exp Child Psychol ; 246: 105994, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38991312

RESUMEN

Early-life positive and adverse parental factors, such as positive parent personality and parental stress, affect the environmental context in which children develop and may influence individual differences in children's sleep health. This study examined the moderating role of early-life parental factors in the heritability (i.e., the extent to which individual differences are due to genetic influences) of objectively assessed childhood sleep duration. A total of 351 families from the Arizona Twin Project were studied. Primary caregivers (95% mothers) reported on multiple dimensions of stress and facets of their own personality when the twins were 12 months old. Seven years later (Mage = 8.43 years, SD = 0.68), families completed a home visit, and twins (51% female; 57% White, 29% Hispanic; 30% monozygotic, 39% same-sex dizygotic, 31% other-sex dizygotic) wore actigraph watches to assess their sleep, with caregivers completing similar assessments on their personality attributes and stress. Early-life positive parent personality moderated the heritability of sleep duration (Δ-2LL [-2 log likelihood] = 2.54, Δdf = 2, p = .28), such that as positive parent personality increased, the heritability of duration decreased. Early-life parental stress also moderated the genetic contribution to sleep duration (Δ-2LL = 2.02, Δdf = 2, p = .36), such that as stress increased, the heritability of duration increased. Concurrent positive parent personality and parental stress composites showed similar patterns of findings. Results highlight the likely contribution of parent positive traits and adverse experiences to the etiology of children's sleep health, with genetic influences on children's sleep more prominent in "riskier" environments. Understanding how genetics and environments work together to influence the etiology of sleep may inform prevention programs.


Asunto(s)
Sueño , Humanos , Masculino , Femenino , Niño , Sueño/genética , Sueño/fisiología , Padres/psicología , Personalidad/genética , Estrés Psicológico/psicología , Estrés Psicológico/genética , Interacción Gen-Ambiente , Arizona , Lactante , Actigrafía , Duración del Sueño
6.
J Neurodev Disord ; 16(1): 39, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39014349

RESUMEN

BACKGROUND: Sleep disturbances are a prevalent and complex comorbidity in neurodevelopmental disorders (NDDs). Dup15q syndrome (duplications of 15q11.2-13.1) is a genetic disorder highly penetrant for NDDs such as autism and intellectual disability and it is frequently accompanied by significant disruptions in sleep patterns. The 15q critical region harbors genes crucial for brain development, notably UBE3A and a cluster of gamma-aminobutyric acid type A receptor (GABAAR) genes. We previously described an electrophysiological biomarker of the syndrome, marked by heightened beta oscillations (12-30 Hz) in individuals with Dup15q syndrome, akin to electroencephalogram (EEG) alterations induced by allosteric modulation of GABAARs. Those with Dup15q syndrome exhibited increased beta oscillations during the awake resting state and during sleep, and they showed profoundly abnormal NREM sleep. This study aims to assess the translational validity of these EEG signatures and to delve into their neurobiological underpinnings by quantifying sleep physiology in chromosome-engineered mice with maternal (matDp/ + mice) or paternal (patDp/ + mice) inheritance of the full 15q11.2-13.1-equivalent duplication, and mice with duplication of just the UBE3A gene (Ube3a overexpression mice; Ube3a OE mice) and comparing the sleep metrics with their respective wildtype (WT) littermate controls. METHODS: We collected 48-h EEG/EMG recordings from 35 (23 male, 12 female) 12-24-week-old matDp/ + , patDp/ + , Ube3a OE mice, and their WT littermate controls. We quantified baseline sleep, sleep fragmentation, spectral power dynamics during sleep states, and recovery following sleep deprivation. Within each group, distinctions between Dup15q mutant mice and WT littermate controls were evaluated using analysis of variance (ANOVA) and student's t-test. The impact of genotype and time was discerned through repeated measures ANOVA, and significance was established at p < 0.05. RESULTS: Our study revealed that across brain states, matDp/ + mice mirrored the elevated beta oscillation phenotype observed in clinical EEGs from individuals with Dup15q syndrome. Time to sleep onset after light onset was significantly reduced in matDp/ + and Ube3a OE mice. However, NREM sleep between Dup15q mutant and WT littermate mice remained unaltered, suggesting a divergence from the clinical presentation in humans. Additionally, while increased beta oscillations persisted in matDp/ + mice after 6-h of sleep deprivation, recovery NREM sleep remained unaltered in all groups, thus suggesting that these mice exhibit resilience in the fundamental processes governing sleep-wake regulation. CONCLUSIONS: Quantification of mechanistic and translatable EEG biomarkers is essential for advancing our understanding of NDDs and their underlying pathophysiology. Our study of sleep physiology in the Dup15q mice underscores that the beta EEG biomarker has strong translational validity, thus opening the door for pre-clinical studies of putative drug targets, using the biomarker as a translational measure of drug-target engagement. The unaltered NREM sleep may be due to inherent differences in neurobiology between mice and humans. These nuanced distinctions highlight the complexity of sleep disruptions in Dup15q syndrome and emphasize the need for a comprehensive understanding that encompasses both shared and distinct features between murine models and clinical populations.


Asunto(s)
Cromosomas Humanos Par 15 , Modelos Animales de Enfermedad , Electroencefalografía , Animales , Ratones , Cromosomas Humanos Par 15/genética , Masculino , Femenino , Trastornos del Sueño-Vigilia/genética , Trastornos del Sueño-Vigilia/fisiopatología , Sueño/fisiología , Sueño/genética , Trisomía/fisiopatología , Trisomía/genética , Aberraciones Cromosómicas , Discapacidad Intelectual
7.
Cell Syst ; 15(7): 595-596, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39024922

RESUMEN

Rhythmic gene expression can originate not only from the autonomous rhythm of clock genes but likely also from sleep-wake cycles. Jan and colleagues used a novel model-based approach to dissect these individual effects and found that both factors contribute to gene expression rhythms, varying in degree within and across tissues.


Asunto(s)
Ritmo Circadiano , Sueño , Sueño/genética , Sueño/fisiología , Ritmo Circadiano/genética , Ritmo Circadiano/fisiología , Humanos , Animales , Regulación de la Expresión Génica/genética , Relojes Circadianos/genética , Relojes Biológicos/genética
8.
Cell Syst ; 15(7): 610-627.e8, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-38986625

RESUMEN

Analyses of gene-expression dynamics in research on circadian rhythms and sleep homeostasis often describe these two processes using separate models. Rhythmically expressed genes are, however, likely to be influenced by both processes. We implemented a driven, damped harmonic oscillator model to estimate the contribution of circadian- and sleep-wake-driven influences on gene expression. The model reliably captured a wide range of dynamics in cortex, liver, and blood transcriptomes taken from mice and humans under various experimental conditions. Sleep-wake-driven factors outweighed circadian factors in driving gene expression in the cortex, whereas the opposite was observed in the liver and blood. Because of tissue- and gene-specific responses, sleep deprivation led to a long-lasting intra- and inter-tissue desynchronization. The model showed that recovery sleep contributed to these long-lasting changes. The results demonstrate that the analyses of the daily rhythms in gene expression must take the complex interactions between circadian and sleep-wake influences into account. A record of this paper's transparent peer review process is included in the supplemental information.


Asunto(s)
Ritmo Circadiano , Sueño , Vigilia , Ritmo Circadiano/genética , Ritmo Circadiano/fisiología , Animales , Humanos , Sueño/genética , Sueño/fisiología , Ratones , Vigilia/fisiología , Vigilia/genética , Regulación de la Expresión Génica/genética , Hígado/metabolismo , Transcriptoma/genética , Privación de Sueño/genética , Privación de Sueño/fisiopatología , Masculino , Homeostasis/genética
9.
Sci Rep ; 14(1): 14962, 2024 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-38942746

RESUMEN

Self-reported shorter/longer sleep duration, insomnia, and evening preference are associated with hyperglycaemia in observational analyses, with similar observations in small studies using accelerometer-derived sleep traits. Mendelian randomization (MR) studies support an effect of self-reported insomnia, but not others, on glycated haemoglobin (HbA1c). To explore potential effects, we used MR methods to assess effects of accelerometer-derived sleep traits (duration, mid-point least active 5-h, mid-point most active 10-h, sleep fragmentation, and efficiency) on HbA1c/glucose in European adults from the UK Biobank (UKB) (n = 73,797) and the MAGIC consortium (n = 146,806). Cross-trait linkage disequilibrium score regression was applied to determine genetic correlations across accelerometer-derived, self-reported sleep traits, and HbA1c/glucose. We found no causal effect of any accelerometer-derived sleep trait on HbA1c or glucose. Similar MR results for self-reported sleep traits in the UKB sub-sample with accelerometer-derived measures suggested our results were not explained by selection bias. Phenotypic and genetic correlation analyses suggested complex relationships between self-reported and accelerometer-derived traits indicating that they may reflect different types of exposure. These findings suggested accelerometer-derived sleep traits do not affect HbA1c. Accelerometer-derived measures of sleep duration and quality might not simply be 'objective' measures of self-reported sleep duration and insomnia, but rather captured different sleep characteristics.


Asunto(s)
Acelerometría , Glucemia , Hemoglobina Glucada , Análisis de la Aleatorización Mendeliana , Sueño , Humanos , Hemoglobina Glucada/análisis , Hemoglobina Glucada/metabolismo , Sueño/genética , Sueño/fisiología , Glucemia/análisis , Masculino , Femenino , Persona de Mediana Edad , Adulto , Autoinforme , Anciano , Trastornos del Inicio y del Mantenimiento del Sueño/genética
10.
Adv Genet ; 111: 497-535, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38908905

RESUMEN

Due to the multifactorial and complex nature of rest, we focus on phenotypes related to sleep. Sleep regulation is a multifactorial process. In this chapter, we focus on those phenotypes inherent to sleep that are highly prevalent in the population, and that can be modulated by lifestyle, such as sleep quality and duration, insomnia, restless leg syndrome and daytime sleepiness. We, therefore, leave in the background those phenotypes that constitute infrequent pathologies or for which the current level of scientific evidence does not favour the implementation of practical approaches of this type. Similarly, the regulation of sleep quality is intimately linked to the regulation of the circadian rhythm. Although this relationship is discussed in the sections that require it, the in-depth study of circadian rhythm regulation at the molecular level deserves a separate chapter, and this is how it is dealt with in this volume.


Asunto(s)
Ritmo Circadiano , Trastornos del Inicio y del Mantenimiento del Sueño , Sueño , Humanos , Sueño/genética , Sueño/fisiología , Ritmo Circadiano/genética , Ritmo Circadiano/fisiología , Trastornos del Inicio y del Mantenimiento del Sueño/genética , Síndrome de las Piernas Inquietas/genética , Fenotipo , Animales , Calidad del Sueño
11.
Genes (Basel) ; 15(6)2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38927705

RESUMEN

Recent research has highlighted associations between sleep and microbial taxa and pathways. However, the causal effect of these associations remains unknown. To investigate this, we performed a bidirectional two-sample Mendelian randomization (MR) analysis using summary statistics of genome-wide association studies (GWAS) from 412 gut microbiome traits (N = 7738) and GWAS studies from seven sleep-associated traits (N = 345,552 to 386,577). We employed multiple MR methods to assess causality, with Inverse Variance Weighted (IVW) as the primary method, alongside a Bonferroni correction ((p < 2.4 × 10-4) to determine significant causal associations. We further applied Cochran's Q statistical analysis, MR-Egger intercept, and Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO) for heterogeneity and pleiotropy assessment. IVW estimates revealed 79 potential causal effects of microbial taxa and pathways on sleep-related traits and 45 inverse causal relationships, with over half related to pathways, emphasizing their significance. The results revealed two significant causal associations: genetically determined relative abundance of pentose phosphate decreased sleep duration (p = 9.00 × 10-5), and genetically determined increase in fatty acid level increased the ease of getting up in the morning (p = 8.06 × 10-5). Sensitivity analyses, including heterogeneity and pleiotropy tests, as well as a leave-one-out analysis of single nucleotide polymorphisms, confirmed the robustness of these relationships. This study explores the potential causal relationships between sleep and microbial taxa and pathways, offering novel insights into their complex interplay.


Asunto(s)
Microbioma Gastrointestinal , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Sueño , Humanos , Microbioma Gastrointestinal/genética , Sueño/genética , Polimorfismo de Nucleótido Simple , Causalidad
12.
Medicine (Baltimore) ; 103(24): e38318, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38875424

RESUMEN

Observational studies have revealed that several sleep traits can impact ovarian function in women. However, there is no evidence suggesting associations between sleep traits and age at natural menopause (ANM). The objective of this study was to investigate the causal relationship between sleep traits (insomnia, sleep duration, daytime sleepiness) and ANM from the perspective of genetic variation. We selected the single-nucleotide polymorphisms from large-scale genome-wide association studies as instrumental variables and conducted a two-sample Mendelian randomization (MR) analysis on these single-nucleotide polymorphisms, including inverse variance weighting, MR-Egger, weighted median, simple mode and weighted mode. The Steiger test was employed to verify the correct causal directionality. The robustness of the MR analysis was examined through Cochran's Q test, horizontal pleiotropy test, and leave-one-out analysis. The results indicated that insomnia was causally associated with ANM (inverse variance weighting: ß = -0.982; 95% CI: -1.852 to -0.111, P = .027), with other analyses confirming the robustness of this finding. Steiger test and reverse MR Analysis validated the absence of a reverse causal association between the two. However, sleep duration and daytime sleepiness did not exhibit a causal effect on ANM. In summary, this study provides initial evidence that insomnia can contribute to an earlier onset of ANM. Nevertheless, further clinical studies are needed to elucidate these findings.


Asunto(s)
Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Menopausia , Polimorfismo de Nucleótido Simple , Sueño , Humanos , Menopausia/genética , Femenino , Sueño/genética , Sueño/fisiología , Trastornos del Inicio y del Mantenimiento del Sueño/genética , Trastornos del Inicio y del Mantenimiento del Sueño/epidemiología , Factores de Edad , Persona de Mediana Edad
13.
Bone ; 186: 117168, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38878990

RESUMEN

BACKGROUND: Unhealthy sleep behaviors are associated with higher risks of osteoporosis (OP), while prospective evidence is limited. This study aimed to prospectively investigate this association, quantify the attributable burden of OP incidence reduction due to unhealthy sleep behaviors, and explore potential modifications by genetic risk factors. METHODS: This longitudinal cohort study was conducted utilizing data from the UK Biobank, comprising 293,164 participants initially free of OP and with requisite sleep behaviors data at baseline. We followed the participants after recruitment until November 30, 2022, to ascertain incident OP. We assessed the associations of five sleep behaviors including sleep duration, chronotype, insomnia, daytime napping, and morning wake-up difficulties, as well as sleep behavior patterns identified based on the above sleep behaviors, with the risk of OP, using Cox models adjusted for multiple confounders. The analyses were then performed separately among individuals with different OP susceptibility, indexed by standard polygenetic risk scores(PRS) for OP. Our secondary outcome was OP with pathologic fracture. Subgroup and sensitivity analyses were performed. Additionally, attributable risk percent in the exposed population (AR%) and population attributable fraction (PAF) of sleep behaviors were calculated. RESULTS: Over a median follow-up of 13.7 years, 8253 new-onset OP cases were documented. Unhealthy sleep behaviors, such as long or short sleep duration, insomnia, daytime napping, morning wake-up difficulties, and unhealthy sleep patterns, were associated with elevated risks of OP (HRs ranging from 1.14 to 1.46, all P-value <0.001) compared to healthy sleep behaviors. Similar associations were observed for OP with pathologic fractures. Insomnia exhibited the largest AR% of 39.98 % (95%CI: 36.46, 43.31) and PAF of 33.25 % (95%CI: 30.00, 36.34) among healthy sleep patterns and components. A statistically significant multiplicative interaction was noted between sleep behaviors and OP PRS on OP risk (all P-interaction <0.001). CONCLUSIONS: Four unhealthy sleep behaviors and sleep behavior patterns were associated to increased OP risk, with insomnia contributing the most to OP incidence, while genetic risk for OP modified this association. These findings underscore the crucial role of adhering to healthy sleep behaviors for effective OP prevention.


Asunto(s)
Predisposición Genética a la Enfermedad , Osteoporosis , Sueño , Humanos , Femenino , Estudios Prospectivos , Osteoporosis/genética , Osteoporosis/epidemiología , Masculino , Sueño/fisiología , Sueño/genética , Factores de Riesgo , Persona de Mediana Edad , Incidencia , Estudios Longitudinales , Anciano , Adulto
14.
Hepatol Int ; 18(4): 1158-1167, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38888882

RESUMEN

BACKGROUND: The associations between sleep patterns or behaviors and the risk of cirrhosis and the influence of genetic susceptibility on these associations among NAFLD participants remain inadequately elucidated. METHODS: This study conducted a prospective follow-up of 112,196 NAFLD participants diagnosed at baseline from the UK Biobank cohort study. Five sleep behaviors were collected to measure a healthy sleep score. Five genetic variants were used to construct a polygenic risk score. We used Cox proportional hazard model to assess hazard ratios (HR) and 95% confidence intervals (CIs) for incidence of cirrhosis. RESULTS: During the follow-up, 592 incident cirrhosis cases were documented. Healthy sleep pattern was associated with reduced risk of cirrhosis in a dose-response manner (ptrend < 0.001). Participants with favourable sleep score (versus unfavourable sleep score) had an HR of 0.55 for cirrhosis risk (95% CI 0.39-0.78). Multivariable-adjusted HRs (95% CIs) of cirrhosis incidence for NAFLDs with no frequent insomnia, sleeping for 7-8 h per day, and no excessive daytime dozing behaviors were 0.73 (0.61-0.87), 0.79 (0.66-0.93), and 0.69 (0.50-0.95), respectively. Compared with participants with favourable sleep pattern and low genetic risk, those with unfavourable sleep pattern and high genetic risk had higher risks of cirrhosis incidence (HR 3.16, 95% CI 1.88-5.33). In addition, a significant interaction between chronotype and genetic risk was detected for the incidence of cirrhosis (p for multiplicative interaction = 0.004). CONCLUSION: An association was observed between healthy sleep pattern and decreased risk of cirrhosis among NAFLD participants, regardless of low or high genetic risk.


Asunto(s)
Predisposición Genética a la Enfermedad , Cirrosis Hepática , Enfermedad del Hígado Graso no Alcohólico , Sueño , Humanos , Femenino , Masculino , Cirrosis Hepática/genética , Cirrosis Hepática/epidemiología , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/epidemiología , Estudios Prospectivos , Sueño/genética , Sueño/fisiología , Incidencia , Reino Unido/epidemiología , Anciano , Factores de Riesgo , Adulto , Estudios de Seguimiento
15.
Cell Rep ; 43(5): 114192, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38703367

RESUMEN

The preoptic area of the hypothalamus (POA) is essential for sleep regulation. However, the cellular makeup of the POA is heterogeneous, and the molecular identities of the sleep-promoting cells remain elusive. To address this question, this study compares mice during recovery sleep following sleep deprivation to mice allowed extended sleep. Single-nucleus RNA sequencing (single-nucleus RNA-seq) identifies one galanin inhibitory neuronal subtype that shows upregulation of rapid and delayed activity-regulated genes during recovery sleep. This cell type expresses higher levels of growth hormone receptor and lower levels of estrogen receptor compared to other galanin subtypes. single-nucleus RNA-seq also reveals cell-type-specific upregulation of purinergic receptor (P2ry14) and serotonin receptor (Htr2a) during recovery sleep in this neuronal subtype, suggesting possible mechanisms for sleep regulation. Studies with RNAscope validate the single-nucleus RNA-seq findings. Thus, the combined use of single-nucleus RNA-seq and activity-regulated genes identifies a neuronal subtype functionally involved in sleep regulation.


Asunto(s)
Galanina , Neuronas , Área Preóptica , Privación de Sueño , Animales , Galanina/metabolismo , Galanina/genética , Neuronas/metabolismo , Área Preóptica/metabolismo , Ratones , Privación de Sueño/metabolismo , Privación de Sueño/genética , Masculino , RNA-Seq , Ratones Endogámicos C57BL , Sueño/genética , Sueño/fisiología , Análisis de la Célula Individual
16.
Front Endocrinol (Lausanne) ; 15: 1264410, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38737549

RESUMEN

Low testosterone levels in men have been linked to decreased physical and mental function, as well as a reduced quality of life. Previous prospective observational studies have suggested an association between testosterone and sleep traits, but the causality of this relationship remains unclear. We aimed to explore the potential causal link between genetically determined sleep traits and testosterone levels in men using Mendelian randomization (MR) analysis from the UK Biobank dataset. Our exposures were genetic variants associated with sleep traits (chronotype and sleep duration), whereas our outcomes were traits of sex steroid hormones (total testosterone, TT; bioavailable testosterone, BAT; and sex hormone-binding globulin, SHBG). We employed inverse variance weighted (IVW) and weighted median (WM) methods to assess the causal associations. The IVW method offers a robust estimate of causality, whereas the WM method provides reliable results even when some genetic variants are invalid instruments. Our main analysis involving sex steroid hormones and chronotype identified 155 chronotype-related variants. The primary findings from the analysis, which used chronotype as the exposure and sex steroid hormones as the outcomes, showed that a genetically predicted chronotype score was significantly associated with an increased levels of TT (association coefficient ß, 0.08; 95% confidence interval [CI], 0.02-0.14; P = 0.008) and BAT (ß, 0.08; 95% CI, 0.02-0.14; P = 0.007), whereas there was no significant association with SHBG (ß, 0.01; 95% CI, -0.02-0.03; P = 0.64). Meanwhile, MR analysis of sex steroid hormones and sleep duration was performed, and 69 variants associated with sleep duration were extracted. There were no significant association between sleep duration and sex steroid hormones (TT, P = 0.91; BAT, P = 0.82; and SHBG, P = 0.95). Our data support a causal association between chronotype and circulating testosterone levels in men. These findings underscore a potential causal relationship between chronotype and testosterone levels in men, suggesting that lifestyle adjustments are crucial for men's health. Recognizing factors that influence testosterone is essential. One limitation of this study is the use of one-sample MR, which can introduce potential bias due to non-independence of genetic associations for exposure and outcome. In conclusion, our findings indicate that a morning preference is correlated with circulating testosterone levels, emphasizing the potential impact of lifestyle habits on testosterone levels in men.


Asunto(s)
Análisis de la Aleatorización Mendeliana , Sueño , Testosterona , Humanos , Masculino , Testosterona/sangre , Sueño/genética , Sueño/fisiología , Globulina de Unión a Hormona Sexual/genética , Globulina de Unión a Hormona Sexual/metabolismo , Persona de Mediana Edad , Ritmo Circadiano/genética , Polimorfismo de Nucleótido Simple , Anciano , Cronotipo
17.
Nat Commun ; 15(1): 3685, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38693116

RESUMEN

Sleep, locomotor and social activities are essential animal behaviors, but their reciprocal relationships and underlying mechanisms remain poorly understood. Here, we elicit information from a cutting-edge large-language model (LLM), generative pre-trained transformer (GPT) 3.5, which interprets 10.2-13.8% of Drosophila genes known to regulate the 3 behaviors. We develop an instrument for simultaneous video tracking of multiple moving objects, and conduct a genome-wide screen. We have identified 758 fly genes that regulate sleep and activities, including mre11 which regulates sleep only in the presence of conspecifics, and NELF-B which regulates sleep regardless of whether conspecifics are present. Based on LLM-reasoning, an educated signal web is modeled for understanding of potential relationships between its components, presenting comprehensive molecular signatures that control sleep, locomotor and social activities. This LLM-aided strategy may also be helpful for addressing other complex scientific questions.


Asunto(s)
Conducta Animal , Drosophila melanogaster , Locomoción , Sueño , Animales , Sueño/fisiología , Sueño/genética , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Locomoción/fisiología , Locomoción/genética , Conducta Animal/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Conducta Social , Masculino
18.
Front Public Health ; 12: 1381482, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38784581

RESUMEN

Background: Research based on observation has demonstrated a relationship between sleep traits and frailty; however, it remains uncertain if this correlation indicates causation. The purpose of this study was to look at the causal relationship that exists between frailty and sleep traits. Method: Using summaries from a genome-wide association study of self-reported sleep features and frailty index, we performed a bidirectional Mendelian randomization (MR) analysis. Examining the causal relationships between seven sleep-related traits and frailty was the goal. The major method used to calculate effect estimates was the inverse-variance weighted method, supplemented by the weighted median and MR-Egger approaches. The study investigated pleiotropy and heterogeneity using several methodologies, such as the MR-Egger intercept, the MR-PRESSO approach, and the Cochran's Q test. We took multivariate Mendelian randomization and genetic correlations between related traits to enhance the confidence of the results. Furthermore, we used MRlap to correct for any estimation bias due to sample overlap. Results: Insomnia, napping during the day, and sleep apnea syndrome exhibited a positive connection with the frailty index in forward MR analysis. Conversely, there is a negative link between getting up in the morning, snoring and sleep duration with the frailty index. During the reverse MR analysis, the frailty index exhibited a positive correlation with insomnia, napping during the day, and sleep apnea syndrome, while demonstrating a negative correlation with sleep duration. There was no direct correlation between snoring, chronotype, and frailty. In MVMR analyses, the causal effect of sleep characteristics on frailty indices remained consistent after adjusting for potential confounders including BMI, smoking, and triglycerides. Conclusion: The findings of our investigation yield novel evidence that substantiates the notion of a bidirectional causal connection between sleep traits and frailty. Through the optimization of sleep, it is potentially feasible to hinder, postpone, or even reverse the state of frailty, and we proposed relevant interventions.


Asunto(s)
Causalidad , Fragilidad , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Sueño , Humanos , Fragilidad/genética , Sueño/fisiología , Sueño/genética , Masculino , Femenino , Anciano , Factores de Riesgo , Persona de Mediana Edad , Trastornos del Sueño-Vigilia/genética , Trastornos del Sueño-Vigilia/epidemiología
19.
Clin Nutr ; 43(6): 1544-1550, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38754306

RESUMEN

Few prospective studies have investigated the joint effect of lifestyle factors and genetic susceptibility on the risk of irritable bowel syndrome (IBS). This study aims to evaluate the associations of lifestyle and genetic factors with incident IBS in the UK Biobank. We analyzed data from 481,057 participants (54% female) without prevalent IBS at enrollment in the UK Biobank. An overall healthy lifestyle was defined using six modifiable lifestyle factors, including smoking, body mass index (BMI), sleep duration, diet, physical activity, and alcohol consumption, and hence categorized into 'favorable', 'intermediate', and 'unfavorable' lifestyles. A Cox proportional hazard model was used to investigate the association between a healthy lifestyle and incident IBS. Furthermore, we constructed a polygenic risk score (PRS) for IBS and assessed whether lifestyle modified the effect of genetics on the development of IBS. During a median follow-up of 12.1 years, 8645 incident IBS were ascertained. Specifically, among the six modifiable lifestyle factors, adequate sleep demonstrates the greatest protective effect (hazard ratio [HR]: 0.72, 95% CI: 0.69,0.75) against IBS. Compared with a favorable lifestyle, an unfavorable lifestyle was associated with a 56% (95% CI: 46%-67%) increased risk of IBS (P = 8.99 × 10-40). The risk of incident IBS was 12% (95% CI: 4%-21%) higher among those at high genetic risk compared with those at low genetic risk (P = 0.005). When considering the joint effect of lifestyle and genetic susceptibility, the HR nearly doubled among individuals with high genetic risk and unfavorable lifestyle (HR: 1.80; 95% CI:1.51-2.15; P = 3.50 × 10-11) compared to those with low genetic risk and favorable lifestyle. No multiplicative or addictive interaction was observed between lifestyle and genetics. The findings from this study indicated that lifestyle and genetic factors were independently associated with the risk of incident IBS. All these results implicated a possible clinical strategy of lowering the incidence of IBS by advocating a healthy lifestyle.


Asunto(s)
Predisposición Genética a la Enfermedad , Síndrome del Colon Irritable , Estilo de Vida , Humanos , Síndrome del Colon Irritable/genética , Síndrome del Colon Irritable/epidemiología , Femenino , Masculino , Estudios Prospectivos , Persona de Mediana Edad , Incidencia , Reino Unido/epidemiología , Factores de Riesgo , Adulto , Modelos de Riesgos Proporcionales , Anciano , Sueño/genética , Estilo de Vida Saludable , Dieta/estadística & datos numéricos
20.
Biochem Biophys Res Commun ; 720: 150072, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38749187

RESUMEN

The Eph receptor, a prototypically large receptor protein tyrosine kinase, interacts with ephrin ligands, forming a bidirectional signaling system that impacts diverse brain functions. Eph receptors and ephrins mediate forward and reverse signaling, affecting neurogenesis, axon guidance, and synaptic signaling. While mammalian studies have emphasized their roles in neurogenesis and synaptic plasticity, the Drosophila counterparts are less studied, especially in glial cells, despite structural similarities. Using RNAi to modulate Eph/ephrin expression in Drosophila neurons and glia, we studied their roles in brain development and sleep and circadian behavior. Knockdown of neuronal ephrin disrupted mushroom body development, while glial knockdown had minimal impact. Surprisingly, disrupting ephrin in neurons or glial cells altered sleep and circadian rhythms, indicating a direct involvement in these behaviors independent from developmental effects. Further analysis revealed distinct sleep phenotypes between neuronal and glial knockdowns, underscoring the intricate interplay within the neural circuits that govern behavior. Glia-specific knockdowns showed altered sleep patterns and reduced circadian rhythmicity, suggesting an intricate role of glia in sleep regulation. Our findings challenge simplistic models of Eph/ephrin signaling limited to neuron-glia communication and emphasize the complexity of the regulatory networks modulating behavior. Future investigations targeting specific glial subtypes will enhance our understanding of Eph/ephrin signaling's role in sleep regulation across species.


Asunto(s)
Ritmo Circadiano , Efrinas , Cuerpos Pedunculados , Neuroglía , Neuronas , Transducción de Señal , Sueño , Animales , Neuroglía/metabolismo , Sueño/fisiología , Sueño/genética , Ritmo Circadiano/fisiología , Neuronas/metabolismo , Efrinas/metabolismo , Efrinas/genética , Cuerpos Pedunculados/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Receptores de la Familia Eph/metabolismo , Receptores de la Familia Eph/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiología , Drosophila melanogaster/genética , Drosophila/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA