Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.084
Filter
1.
Biochem Biophys Res Commun ; 720: 150072, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38749187

ABSTRACT

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.


Subject(s)
Circadian Rhythm , Ephrins , Mushroom Bodies , Neuroglia , Neurons , Signal Transduction , Sleep , Animals , Neuroglia/metabolism , Sleep/physiology , Sleep/genetics , Circadian Rhythm/physiology , Neurons/metabolism , Ephrins/metabolism , Ephrins/genetics , Mushroom Bodies/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Receptors, Eph Family/metabolism , Receptors, Eph Family/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/physiology , Drosophila melanogaster/genetics , Drosophila/metabolism
2.
Clin Nutr ; 43(6): 1544-1550, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38754306

ABSTRACT

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.


Subject(s)
Genetic Predisposition to Disease , Irritable Bowel Syndrome , Life Style , Humans , Irritable Bowel Syndrome/genetics , Irritable Bowel Syndrome/epidemiology , Female , Male , Prospective Studies , Middle Aged , Incidence , United Kingdom/epidemiology , Risk Factors , Adult , Proportional Hazards Models , Aged , Sleep/genetics , Healthy Lifestyle , Diet/statistics & numerical data
3.
Cell Rep ; 43(5): 114192, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38703367

ABSTRACT

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.


Subject(s)
Galanin , Neurons , Preoptic Area , Sleep Deprivation , Animals , Galanin/metabolism , Galanin/genetics , Neurons/metabolism , Preoptic Area/metabolism , Mice , Sleep Deprivation/metabolism , Sleep Deprivation/genetics , Male , RNA-Seq , Mice, Inbred C57BL , Sleep/genetics , Sleep/physiology , Single-Cell Analysis
4.
Nat Commun ; 15(1): 3685, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693116

ABSTRACT

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.


Subject(s)
Behavior, Animal , Drosophila melanogaster , Locomotion , Sleep , Animals , Sleep/physiology , Sleep/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Locomotion/physiology , Locomotion/genetics , Behavior, Animal/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Social Behavior , Male
5.
Front Endocrinol (Lausanne) ; 15: 1264410, 2024.
Article in English | MEDLINE | ID: mdl-38737549

ABSTRACT

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.


Subject(s)
Mendelian Randomization Analysis , Sleep , Testosterone , Humans , Male , Testosterone/blood , Sleep/genetics , Sleep/physiology , Sex Hormone-Binding Globulin/genetics , Sex Hormone-Binding Globulin/metabolism , Middle Aged , Circadian Rhythm/genetics , Polymorphism, Single Nucleotide , Aged , Chronotype
6.
Chronobiol Int ; 41(5): 757-766, 2024 May.
Article in English | MEDLINE | ID: mdl-38695651

ABSTRACT

Delayed sleep phase disorder and advanced sleep phase disorder cause disruption of the circadian clock and present with extreme morning/evening chronotype with unclear role of the genetic etiology, especially for delayed sleep phase disorder. To assess if genotyping can aid in clinical diagnosis, we examined the presence of genetic variants in circadian clock genes previously linked to both sleep disorders in Slovenian patient cohort. Based on Morning-evening questionnaire, we found 15 patients with extreme chronotypes, 13 evening and 2 morning, and 28 controls. Sanger sequencing was used to determine the presence of carefully selected candidate SNPs in regions of the CSNK1D, PER2/3 and CRY1 genes. In a patient with an extreme morning chronotype and a family history of circadian sleep disorder we identified two heterozygous missense variants in PER3 gene, c.1243C>G (NM_001377275.1 (p.Pro415Ala)) and c.1250A>G (NM_001377275.1 (p.His417Arg)). The variants were significantly linked to Advanced sleep phase disorder and were also found in proband's father with extreme morningness. Additionally, a rare SNP was found in PER2 gene in a patient with clinical picture of Delayed sleep phase disorder. The novel variant in PER2 (NM_022817.3):c.1901-218 G>T was found in proband's parent with eveningness, indicating an autosomal dominant inheritance. We identified a family with autosomal dominant inheritance of two PER3 heterozygous variants that can be linked to Advanced sleep phase disorder. We revealed also a rare hereditary form of Delayed sleep phase disorder with a new PER2 variant with autosomal dominant inheritance, shedding the light into the genetic causality.


Subject(s)
Circadian Clocks , Period Circadian Proteins , Polymorphism, Single Nucleotide , Sleep Disorders, Circadian Rhythm , Humans , Period Circadian Proteins/genetics , Male , Female , Adult , Middle Aged , Sleep Disorders, Circadian Rhythm/genetics , Circadian Clocks/genetics , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Genetic Predisposition to Disease , Slovenia , Pedigree , Sleep/genetics , Sleep/physiology , Young Adult
7.
Front Public Health ; 12: 1381482, 2024.
Article in English | MEDLINE | ID: mdl-38784581

ABSTRACT

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.


Subject(s)
Causality , Frailty , Genome-Wide Association Study , Mendelian Randomization Analysis , Sleep , Humans , Frailty/genetics , Sleep/physiology , Sleep/genetics , Male , Female , Aged , Risk Factors , Middle Aged , Sleep Wake Disorders/genetics , Sleep Wake Disorders/epidemiology
8.
Dev Psychobiol ; 66(2)2024 Feb.
Article in English | MEDLINE | ID: mdl-38601952

ABSTRACT

Maternal exposure to childhood adversity is associated with detrimental health outcomes throughout the lifespan and may have implications for offspring. Evidence links maternal adverse childhood experiences (ACEs) to detrimental birth outcomes, yet the impact on the infant's epigenome is unclear. Moreover, maternal sleep habits during pregnancy may influence this association. Here, we explore whether restless sleep during pregnancy moderates the association between exposure to maternal childhood adversity and infant epigenetic age acceleration in 332 mother-infant dyads (56% female; 39% Black; 25% Hispanic). During the 2nd trimester, mothers self-reported childhood adversity and past-week restless sleep; DNA methylation from umbilical vein endothelial cells was used to estimate five epigenetic clocks. Multivariable linear regression was used to test study hypotheses. Despite no evidence of main effects, there was evidence of an interaction between maternal ACEs and restless sleep in predicting infant epigenetic age acceleration using the EPIC Gestational Age clock. Only infants whose mothers reported exposure to both ACEs and restless sleep demonstrated accelerated epigenetic aging. Results provide preliminary evidence that maternal childhood adversity and sleep may influence the infant epigenome.


Subject(s)
Adverse Childhood Experiences , Infant , Pregnancy , Humans , Female , Male , Endothelial Cells , Mothers , Aging , Epigenesis, Genetic , Sleep/genetics
9.
Curr Opin Neurobiol ; 86: 102874, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38582021

ABSTRACT

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.


Subject(s)
Sleep , Humans , Sleep/physiology , Sleep/genetics , Animals , Energy Metabolism/physiology , Energy Metabolism/genetics , Circadian Clocks/physiology , Circadian Clocks/genetics , Circadian Rhythm/physiology , Circadian Rhythm/genetics
10.
Cell Rep Med ; 5(5): 101534, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38670100

ABSTRACT

Thalamocortical (TC) circuits are essential for sensory information processing. Clinical and preclinical studies of autism spectrum disorders (ASDs) have highlighted abnormal thalamic development and TC circuit dysfunction. However, mechanistic understanding of how TC dysfunction contributes to behavioral abnormalities in ASDs is limited. Here, our study on a Shank3 mouse model of ASD reveals TC neuron hyperexcitability with excessive burst firing and a temporal mismatch relationship with slow cortical rhythms during sleep. These TC electrophysiological alterations and the consequent sensory hypersensitivity and sleep fragmentation in Shank3 mutant mice are causally linked to HCN2 channelopathy. Restoring HCN2 function early in postnatal development via a viral approach or lamotrigine (LTG) ameliorates sensory and sleep problems. A retrospective case series also supports beneficial effects of LTG treatment on sensory behavior in ASD patients. Our study identifies a clinically relevant circuit mechanism and proposes a targeted molecular intervention for ASD-related behavioral impairments.


Subject(s)
Autism Spectrum Disorder , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Nerve Tissue Proteins , Thalamus , Animals , Thalamus/metabolism , Thalamus/pathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Mice , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/physiopathology , Autism Spectrum Disorder/pathology , Lamotrigine/pharmacology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Channelopathies/genetics , Channelopathies/metabolism , Channelopathies/pathology , Humans , Disease Models, Animal , Male , Neurons/metabolism , Female , Mice, Inbred C57BL , Mutation/genetics , Sleep/physiology , Sleep/drug effects , Sleep/genetics , Potassium Channels
11.
Geriatr Gerontol Int ; 24(6): 537-545, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38639007

ABSTRACT

AIM: Despite limited evidence regarding the impact of sleep quality on sarcopenia, it is widely recognized as being associated with various diseases. This study aimed to explore the causal relationship between sleep traits and sarcopenia-related traits. METHODS: This study utilized a two-sample bidirectional Mendelian randomization analysis. Genetic genome-wide summary data of sleep quality indicators, including chronotype, morning wake-up time, sleep duration, daytime napping, insomnia and daytime dozing, were used. Data on sarcopenia-related traits, such as appendicular lean mass, grip strength of both hands, walking pace and waist circumference, were collected from a large cohort study. The primary method used was the inverse-variance weighted analysis. RESULTS: A causal association was found between chronotype and appendicular lean mass (odds ratio [OR] 1.019, 95% confidence interval [CI] 1.016-1.211, P = 0.021). Napping during the day was connected with walking pace (OR 0.879, 95% CI 0.834-0.928, P = 2.289 × 10-6) and waist circumference (OR 1.234, 95% CI 1.081-1.408, P = 0.002). Insomnia was related to lower grip strength of the right hand (OR 0.844, 95% CI 0.747-0.954, P = 0.007), left hand (OR 0.836, 95% CI 0.742-0.943, P = 0.003), as well as walking pace (OR 0.871, 95% CI 0.798-0.951, P = 0.002). Furthermore, the reverse Mendelian randomization analysis showed associations between certain sarcopenia-related traits and poor sleep quality. CONCLUSIONS: Some sleep traits were associated with the occurrence of sarcopenia. These findings emphasized the significance of prioritizing sleep quality as a preventive measure against sarcopenia. Geriatr Gerontol Int 2024; 24: 537-545.


Subject(s)
Hand Strength , Mendelian Randomization Analysis , Sarcopenia , Humans , Sarcopenia/genetics , Sarcopenia/epidemiology , Male , Hand Strength/physiology , Female , Aged , Sleep Quality , Waist Circumference , Cohort Studies , Sleep Initiation and Maintenance Disorders/genetics , Sleep Initiation and Maintenance Disorders/epidemiology , Sleep/physiology , Sleep/genetics , Middle Aged
12.
Exp Neurol ; 376: 114775, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38604438

ABSTRACT

OBJECTIVE: Sleep-related hypermotor epilepsy (SHE) is a focal epilepsy syndrome characterized by seizures that predominantly occur during sleep. The pathogenesis of these seizures remains unclear. We previously detected rare variants in GABRG2, which encodes the γ2 subunit of γ-aminobutyric acid type A receptor (GABAAR), in patients with SHE and demonstrated that these variants impaired GABAAR function in vitro. However, the mechanisms by which GABRG2 variants contribute to seizure attacks during sleep remain unclear. METHODS: In this study, we designed a knock-in (KI) mouse expressing the mouse Gabrg2 T316N variant, corresponding to human GABRG2 T317N variant, using CRISPR/Cas9. Continuous video-electroencephalogram monitoring and in vivo multichannel electrophysiological recordings were performed to explore seizure susceptibility to pentylenetetrazol (PTZ), alterations in the sleep-wake cycle, spontaneous seizure patterns, and synchronized activity in the motor thalamic nuclei (MoTN) and secondary motor cortex (M2). Circadian variations in the expression of total, membrane-bound, and synaptic GABAAR subunits were also investigated. RESULTS: No obvious changes in gross morphology were detected in Gabrg2T316N/+ mice compared to their wild-type (Gabrg2+/+) littermates. Gabrg2T316N/+ mice share key phenotypes with patients, including sleep fragmentation and spontaneous seizures during sleep. Gabrg2T316N/+ mice showed increased susceptibility to PTZ-induced seizures and higher mortality after seizures. Synchronization of the local field potentials between the MoTN and M2 was abnormally enhanced in Gabrg2T316N/+ mice during light phase, when sleep dominates, accompanied by increased local activities in the MoTN and M2. Interestingly, in Gabrg2+/+ mice, GABAAR γ2 subunits showed a circadian increase on the neuronal membrane and synaptosomes in the transition from dark phase to light phase, which was absent in Gabrg2T316N/+ mice. CONCLUSION: We generated a new SHE mouse model and provided in vivo evidence that rare variants of GABRG2 contribute to seizure attacks during sleep in SHE.


Subject(s)
Cerebral Cortex , Epilepsy , Receptors, GABA-A , Thalamus , Animals , Female , Male , Mice , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Electroencephalography , Epilepsy/genetics , Epilepsy/physiopathology , Gene Knock-In Techniques , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Sleep/physiology , Sleep/genetics , Thalamus/metabolism , Thalamus/pathology
13.
Sci Rep ; 14(1): 9585, 2024 04 26.
Article in English | MEDLINE | ID: mdl-38671284

ABSTRACT

This study aimed to investigate the probable existence of a causal relationship between sleep phenotypes and proliferative diabetic retinopathy (PDR). Single nucleotide polymorphisms associated with sleep phenotypes were selected as instrumental variables at the genome-wide significance threshold (P < 5 × 10-8). Inverse-variance weighted was applied as the primary Mendelian randomization (MR) analysis method, and MR Egger regression, weighted median, simple mode, and weighted mode methods were used as complementary analysis methods to estimate the causal association between sleep phenotypes and PDR. Results indicated that genetically predicted sleep phenotypes had no causal effects on PDR risk after Bonferroni correction (P = 0.05/10) [Chronotype: P = 0.143; Daytime napping: P = 0.691; Daytime sleepiness: P = 0.473; Insomnia: P = 0.181; Long sleep duration: P = 0.671; Morning person:P = 0.113; Short sleep duration: P = 0.517; Obstructive sleep apnea: P = 0.091; Sleep duration: P = 0.216; and snoring: P = 0.014]. Meanwhile, there are no reverse causality for genetically predicted PDR on sleep phenotypes [Chronotype: P = 0.100; Daytime napping: P = 0.146; Daytime sleepiness: P = 0.469; Insomnia: P = 0.571; Long sleep duration: P = 0.779; Morning person: P = 0.040; Short sleep duration: P = 0.875; Obstructive sleep apnea: P = 0.628; Sleep duration: P = 0.896; and snoring: P = 0.047]. This study's findings did not support the causal effect of between sleep phenotypes and PDR. Whereas, longitudinal studies can further verify results validation.


Subject(s)
Diabetic Retinopathy , Mendelian Randomization Analysis , Phenotype , Polymorphism, Single Nucleotide , Sleep , Humans , Diabetic Retinopathy/genetics , Sleep/genetics , Risk Factors , Genetic Predisposition to Disease , Genome-Wide Association Study
14.
PeerJ ; 12: e17119, 2024.
Article in English | MEDLINE | ID: mdl-38525273

ABSTRACT

Background: Studies have shown that chronic exposure to job stress may increase the risk of sleep disturbances and that hypothalamic‒pituitary‒adrenal (HPA) axis gene polymorphisms may play an important role in the psychopathologic mechanisms of sleep disturbances. However, the interactions among job stress, gene polymorphisms and sleep disturbances have not been examined from the perspective of the HPA axis. This study aimed to know whether job stress is a risk factor for sleep disturbances and to further explore the effect of the HPA axis gene × job stress interaction on sleep disturbances among railway workers. Methods: In this cross-sectional study, 671 participants (363 males and 308 females) from the China Railway Fuzhou Branch were included. Sleep disturbances were evaluated with the Pittsburgh Sleep Quality Index (PSQI), and job stress was measured with the Effort-Reward Imbalance scale (ERI). Generalized multivariate dimensionality reduction (GMDR) models were used to assess gene‒environment interactions. Results: We found a significant positive correlation between job stress and sleep disturbances (P < 0.01). The FKBP5 rs1360780-T and rs4713916-A alleles and the CRHR1 rs110402-G allele were associated with increased sleep disturbance risk, with adjusted ORs (95% CIs) of 1.75 [1.38-2.22], 1.68 [1.30-2.18] and 1.43 [1.09-1.87], respectively. However, the FKBP5 rs9470080-T allele was a protective factor against sleep disturbances, with an OR (95% CI) of 0.65 [0.51-0.83]. GMDR analysis indicated that under job stress, individuals with the FKBP5 rs1368780-CT, rs4713916-GG, and rs9470080-CT genotypes and the CRHR1 rs110402-AA genotype had the greatest risk of sleep disturbances. Conclusions: Individuals carrying risk alleles who experience job stress may be at increased risk of sleep disturbances. These findings may provide new insights into stress-related sleep disturbances in occupational populations.


Subject(s)
Gene-Environment Interaction , Occupational Stress , Male , Female , Humans , Hypothalamo-Hypophyseal System , Cross-Sectional Studies , Pituitary-Adrenal System , Polymorphism, Genetic/genetics , Occupational Stress/epidemiology , Sleep/genetics
15.
J Biol Rhythms ; 39(3): 270-281, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38425306

ABSTRACT

The association between circadian rhythms and diseases has been well established, while the association with mental health is less explored. Given the heritable nature of circadian rhythms, this study aimed to investigate the relationship between genes underlying circadian rhythms and mental health outcomes, as well as a possible gene-environment correlation for circadian rhythms. Polygenic scores (PGSs) represent the genetic predisposition to develop a certain trait or disease. In a sample from the Netherlands Twin Register (N = 14,021), PGSs were calculated for two circadian rhythm measures: morningness and relative amplitude (RA). The PGSs were used to predict mental health outcomes such as subjective happiness, quality of life, and depressive symptoms. In addition, we performed the same prediction analysis in a within-family design in a subset of dizygotic twins. The PGS for morningness significantly predicted morningness (R2 = 1.55%) and depressive symptoms (R2 = 0.22%). The PGS for RA significantly predicted general health (R2 = 0.12%) and depressive symptoms (R2 = 0.20%). Item analysis of the depressive symptoms showed that 4 out of 14 items were significantly associated with the PGSs. Overall, the results showed that people with a genetic predisposition of being a morning person or with a high RA are likely to have fewer depressive symptoms. The four associated depressive symptoms described symptoms related to decision-making, energy, and feeling worthless or inferior, rather than sleep. Based on our findings future research should include a substantial role for circadian rhythms in depression research and should further explore the gene-environment correlation in circadian rhythms.


Subject(s)
Circadian Rhythm , Depression , Multifactorial Inheritance , Quality of Life , Humans , Circadian Rhythm/genetics , Male , Female , Depression/genetics , Adult , Middle Aged , Netherlands , Twins, Dizygotic/genetics , Mental Health , Sleep/genetics , Sleep/physiology , Genetic Predisposition to Disease , Aged , Young Adult , Chronotype
16.
Eur J Clin Invest ; 54(6): e14189, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38429948

ABSTRACT

BACKGROUND: Disturbances in habitual sleep have been associated with multiple age-associated diseases. However, the biological mechanisms underpinning these associations remain largely unclear. We assessed the possible involvement of the circulating immune system by determining the associations between sleep traits and white blood cell counts using multivariable-adjusted linear regression and Mendelian randomization. METHODS: Cross-sectional multivariable-adjusted linear regression analyses were done using participants within the normal range of total white blood cell counts (>4.5 × 109 and <11.0 × 109/µL) from UK Biobank. For the sleep traits, we examined (short and long) sleep duration, chronotype, insomnia symptoms and daytime dozing. Two-sample Mendelian randomization analyses were done using instruments for sleep traits derived from European-ancestry participants from UK Biobank (over 410,000 participants) and using SNP-outcome data derived from European-ancestry participants from the Blood Cell Consortium (N = 563,946) to which no data from UK Biobank contributed. RESULTS: Using data from 357,656 participants (mean [standard deviation] age: 56.5 [8.1] years, and 44.4% men), we did not find evidence that disturbances in any of the studied sleep traits were associated with differences in blood cell counts (total, lymphocytes, neutrophiles, eosinophiles and basophiles). Also, we did not find associations between disturbances in any of the studied sleep traits and white blood cell counts using Mendelian Randomization. CONCLUSION: Based on the results from two different methodologies, disturbances in habitual sleep are unlikely to cause changes in blood cell counts and thereby differences in blood cell counts are unlikely to be underlying the observed sleep-disease associations.


Subject(s)
Mendelian Randomization Analysis , Sleep , Humans , Male , Female , Middle Aged , Leukocyte Count , Cross-Sectional Studies , Sleep/genetics , Sleep/physiology , Aged , Sleep Initiation and Maintenance Disorders/genetics , Sleep Initiation and Maintenance Disorders/epidemiology , Linear Models , Polymorphism, Single Nucleotide , Adult , Multivariate Analysis
17.
BMC Geriatr ; 24(1): 168, 2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38368347

ABSTRACT

BACKGROUND: Observational studies have suggested that sedentary behaviors and sleep status are associated with frailty. However, it remains unclear whether these associations are causal. METHODS: Using summary statistics from genome-wide association studies, we evaluated the causal effect of modifiable risk factors, including leisure sedentary behaviors and sleep status on the frailty index (FI) using two-sample univariable and multivariable Mendelian randomization (MR) analyses. Genetic correlations were tested between the correlated traits. RESULTS: We identified potential causal associations between the time spent watching television (ß = 0.26, 95% confidence interval [CI]: 0.21-0.31, P = 3.98e-25), sleep duration (ß = -0.18, 95%CI: -0.26, -0.10; P = 6.04e-06), and daytime napping (ß = 0.29, 95%CI: 0.18-0.41, P = 2.68e-07) and the FI based on the inverse-variance-weighted method. The estimates were consistent across robust and multivariate MR analyses. Linkage disequilibrium score regression detected a genetic correlation between the time spent watching television (Rg = 0.43, P = 6.46e-48), sleep duration (Rg = -0.20, P = 5.29e-10), and daytime napping (Rg = 0.25, P = 3.34e-21) and the FI. CONCLUSIONS: Genetic predispositions to time spent watching television and daytime napping were positively associated with the FI, while sleep duration was negatively associated with the FI. Our findings offer key insights into factors influencing biological aging and suggest areas for interventions to promote healthy aging and slow down the aging process.


Subject(s)
Frailty , Humans , Genome-Wide Association Study , Mendelian Randomization Analysis , Sedentary Behavior , Sleep/genetics , Leisure Activities
18.
Genes Brain Behav ; 23(1): e12885, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38359178

ABSTRACT

Genome-wide association studies (GWAS) have been important for characterizing the genetic component and enhancing our understanding of the biological aetiology of both neuropsychiatric disorders and sleep-related phenotypes such as chronotype, which is our preference for morning or evening time. Mendelian randomization (MR) is a post-GWAS analysis that is used to infer causal relationships between potential risk factors and outcomes. MR uses genetic variants as instrumental variants for exposures to study the effect on outcomes. This review details the main results from GWAS of neuropsychiatric disorders and sleep-related phenotypes, and the application of MR to investigate their bidirectional relationship. The main results from MR studies of neuropsychiatric disorders and sleep-related phenotypes are summarized. These MR studies have identified 37 causal relationships between neuropsychiatric disorders and sleep-related phenotypes. MR studies identified evidence of a causal role for five neuropsychiatric disorders and symptoms (attention deficit hyperactivity disorder, bipolar disorder, depressive symptoms, major depressive disorder and schizophrenia) on sleep-related phenotypes and evidence of a causal role for five sleep-related phenotypes (daytime napping, insomnia, morning person, long sleep duration and sleep duration) on risk for neuropsychiatric disorders. These MR results show a bidirectional relationship between neuropsychiatric disorders and sleep-related phenotypes and identify potential risk factors for follow-up studies.


Subject(s)
Depressive Disorder, Major , Genome-Wide Association Study , Humans , Chronotype , Mendelian Randomization Analysis , Sleep/genetics
19.
Prev Med ; 180: 107893, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38342383

ABSTRACT

PURPOSE: The association between sleep and myopia in children and adolescents has been reported, yet it remains controversial and inconclusive. This study aimed to investigate the influence of different sleep traits on the risk of myopia using meta-analytical and Mendelian randomization (MR) techniques. METHODS: The literature search was performed in August 31, 2023 based on PubMed, Embase, Web of Science, and Cochrane library. The meta-analysis of observational studies reporting the relationship between sleep and myopia was conducted. MR analyses were carried out to assess the causal impact of genetic pre-disposition for sleep traits on myopia. RESULTS: The results of the meta-analysis indicated a significant association between the risk of myopia and both short sleep duration [odds ratio (OR) = 1.23, 95% confidence interval (CI) = 1.08-1.42, P = 0.003] and long sleep duration (OR = 0.75, 95% CI = 0.66-0.86, P < 0.001). MR analyses revealed no significant causal associations of genetically determined sleep traits with myopia, including chronotype, sleep duration, short sleep duration and long sleep duration (all P > 0.05). CONCLUSIONS: No evidence was found to support a causal relationship between sleep traits and myopia. While sleep may not independently predict the risk of myopia, the potential impact of sleep on the occurrence and development of myopia cannot be disregarded.


Subject(s)
Mendelian Randomization Analysis , Myopia , Child , Adolescent , Humans , Sleep/genetics , Myopia/epidemiology , Myopia/genetics , Odds Ratio , Phenotype , Genome-Wide Association Study
20.
J Med Genet ; 61(6): 586-589, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38350721

ABSTRACT

Pogo transposable element-derived protein with ZNF domain (POGZ) gene encodes a chromatin regulator and rare variants on this gene have been associated with a broad spectrum of neurodevelopmental disorders, such as White-Sutton syndrome. Patient clinical manifestations frequently include developmental delay, autism spectrum disorder and obesity. Sleep disturbances are also commonly observed in these patients, yet the biological pathways which link sleep traits to the POGZ-associated syndrome remain unclear. We screened for sleep implications among individuals with causative POGZ variants previously described. Sleep disturbances were observed in 52% of patients, and being obese was not observed as a risk factor for sleep problems. Next, we identified genes associated with sleep-associated traits among the POGZ regulatory targets, aiming to uncover the molecular pathways that, when disrupted by POGZ loss of function, contribute to the aetiology of sleep phenotypes in these patients. The intersect between POGZ targets and sleep-related genes was used in a pathway enrichment analysis. Relevant pathways among these overlapping genes are involved in the regulation of circadian rhythm, tau protein binding, ATPase activator activity. This study may represent the beginning for novel functional investigations on shared molecular mechanisms between sleep disturbances and rare developmental syndromes related to POGZ and its regulatory targets.


Subject(s)
Neurodevelopmental Disorders , Phenotype , Sleep Wake Disorders , Humans , Neurodevelopmental Disorders/genetics , Sleep Wake Disorders/genetics , Male , Female , Sleep/genetics , Child , Child, Preschool , Circadian Rhythm/genetics , DNA-Binding Proteins , Cell Cycle Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...