Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 236
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1379130, 2024.
Article in English | MEDLINE | ID: mdl-38988999

ABSTRACT

Objective: To explore the association between circadian syndrome (CircS) and Metabolic Syndrome (MetS) with psoriasis. Compare the performance of MetS and CircS in predicting psoriasis. Methods: An observational study used data from the NHANES surveys conducted in 2005-2006 and 2009-2014. We constructed three multiple logistic regression models to investigate the relationship between MetS, CircS, and their components with psoriasis. The performance of MetS and CircS in predicting psoriasis was compared using five machine-learning algorithms, and the best-performing model was explained via SHAP. Then, bidirectional Mendelian randomization analyses with the inverse variance weighted (IVW) as the primary method were employed to determine the causal effects of each component. Result: A total of 9,531 participants were eligible for the study. Both the MetS (OR = 1.53, 95%CI: 1.07-2.17, P = 0.02) and CircS (OR = 1.40, 95%CI: 1.02-1.91, P = 0.039) positively correlated with psoriasis. Each CircS algorithmic model performs better than MetS, with Categorical Features+Gradient Boosting for CircS (the area under the precision-recall curve = 0.969) having the best prediction effect on psoriasis. Among the components of CircS, elevated blood pressure, depression symptoms, elevated waist circumference (WC), and short sleep contributed more to predicting psoriasis. Under the IVW methods, there were significant causal relationships between WC (OR = 1.52, 95%CI: 1.34-1.73, P = 1.35e-10), hypertension (OR = 1.68, 95%CI: 1.19-2.37, P = 0.003), depression symptoms (OR = 1.39, 95%CI: 1.17-1.65, P = 1.51e-4), and short sleep (OR = 2.03, 95%CI: 1.21-3.39, p = 0.007) with psoriasis risk. Conclusion: CircS demonstrated superior predictive ability for prevalent psoriasis compared to MetS, with elevated blood pressure, depression symptoms, and elevated WC contributing more to the prediction.


Subject(s)
Machine Learning , Metabolic Syndrome , Nutrition Surveys , Psoriasis , Humans , Metabolic Syndrome/epidemiology , Psoriasis/epidemiology , Male , Female , Middle Aged , Adult , Chronobiology Disorders/epidemiology , Chronobiology Disorders/complications , Aged , Risk Factors
2.
Nutrients ; 16(13)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38999859

ABSTRACT

BACKGROUND: Circadian Syndrome (CircS) encompasses cardiometabolic risk factors and comorbidities, indicating an elevated susceptibility to cardiovascular disease and type 2 diabetes. METHODS: This cross-sectional study aimed to investigate the association between vitamin D levels and each of the following: CircS, metabolic syndrome (MetS), and the individual components of CircS. Data from 14,907 adults who participated in the National Health and Nutrition Examination Survey (NHANES) between 2007 and 2018 were utilized. CircS was defined based on MetS components, alongside depression, short sleep, and non-alcoholic fatty liver disease (NAFLD). RESULTS: Our results indicated that low vitamin D levels exhibited meaningful associations with CircS, with vitamin D deficiency and inadequacy demonstrating 2.21-fold (95% CI 1.78-2.74, p < 0.001) and 1.33-fold (95% CI 1.14-1.54, p < 0.001) increases in CircS odds, respectively. The association between vitamin D deficiency and CircS was stronger than that with MetS. Additionally, a dose-response gradient in odds of CircS components, particularly with short sleep duration, was noted as serum vitamin D levels decreased. CONCLUSIONS: our findings highlight a significant association between low serum vitamin D levels and CircS and its components, particularly with short sleep. This suggests a potentially pivotal role of vitamin D in the pathogenesis of Circadian syndrome.


Subject(s)
Metabolic Syndrome , Vitamin D Deficiency , Vitamin D , Humans , Cross-Sectional Studies , Vitamin D/blood , Vitamin D/analogs & derivatives , Male , Female , Vitamin D Deficiency/blood , Vitamin D Deficiency/epidemiology , Vitamin D Deficiency/complications , Middle Aged , Adult , Metabolic Syndrome/blood , Metabolic Syndrome/epidemiology , Nutrition Surveys , Chronobiology Disorders/blood , Chronobiology Disorders/complications , Risk Factors , Circadian Rhythm/physiology , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/epidemiology
3.
Diabetes Metab Res Rev ; 40(5): e3827, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38837323

ABSTRACT

AIMS: Circadian syndrome (CircS) is considered a better predictor for cardiovascular disease than the metabolic syndrome (MetS). We aim to examine the associations between CircS and MetS with cognition in Chinese adults. METHOD: We used the data of 8546 Chinese adults aged ≥40 years from the 2011 China Health and Retirement Longitudinal Study. MetS was defined using harmonised criteria. CircS included the components of MetS plus short sleep and depression. The cut-off for CircS was set as ≥4. Global cognitive function was assessed during the face-to-face interview. RESULTS: CircS and MetS had opposite associations with the global cognition score and self-reported poor memory. Compared with individuals without the CircS and MetS, the regression coefficients (95%CI) for global cognition score were -1.02 (-1.71 to -0.34) for CircS alone and 0.52 (0.09 to 0.96) for MetS alone in men; -1.36 (-2.00 to -0.72) for CircS alone and 0.60 (0.15 to 1.06) for MetS alone in women. Having CircS alone was 2.53 times more likely to report poor memory in men (95%CI 1.80-3.55) and 2.08 times more likely in women (95%CI 1.54-2.81). In contrast, having MetS alone was less likely to report poor memory (OR 0.64 (0.49-0.84) in men and 0.65 (0.52-0.81) in women). People with CircS and MetS combined were more likely to have self-reported poor memory. CONCLUSIONS: CircS is a strong and better predictor for cognition impairment than MetS in Chinese middle-aged adults. MetS without short sleep and depression is associated with better cognition.


Subject(s)
Cognitive Dysfunction , Metabolic Syndrome , Humans , Metabolic Syndrome/epidemiology , Metabolic Syndrome/psychology , Male , Female , Middle Aged , Cognitive Dysfunction/etiology , Cognitive Dysfunction/epidemiology , China/epidemiology , Longitudinal Studies , Aged , Adult , Prognosis , Chronobiology Disorders/complications , Chronobiology Disorders/epidemiology , Risk Factors , Follow-Up Studies , Circadian Rhythm/physiology
4.
Front Endocrinol (Lausanne) ; 15: 1328139, 2024.
Article in English | MEDLINE | ID: mdl-38742195

ABSTRACT

The topic of human circadian rhythms is not only attracting the attention of clinical researchers from various fields but also sparking a growing public interest. The circadian system comprises the central clock, located in the suprachiasmatic nucleus of the hypothalamus, and the peripheral clocks in various tissues that are interconnected; together they coordinate many daily activities, including sleep and wakefulness, physical activity, food intake, glucose sensitivity and cardiovascular functions. Disruption of circadian regulation seems to be associated with metabolic disorders (particularly impaired glucose tolerance) and cardiovascular disease. Previous clinical trials revealed that disturbance of the circadian system, specifically due to shift work, is associated with an increased risk of type 2 diabetes mellitus. This review is intended to provide clinicians who wish to implement knowledge of circadian disruption in diagnosis and strategies to avoid cardio-metabolic disease with a general overview of this topic.


Subject(s)
Cardiovascular Diseases , Circadian Rhythm , Metabolic Diseases , Humans , Circadian Rhythm/physiology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Metabolic Diseases/physiopathology , Metabolic Diseases/metabolism , Metabolic Diseases/etiology , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/metabolism , Chronobiology Disorders/physiopathology , Chronobiology Disorders/complications
5.
Front Endocrinol (Lausanne) ; 15: 1338110, 2024.
Article in English | MEDLINE | ID: mdl-38737554

ABSTRACT

Introduction: Circadian syndrome (CircS) is proposed as a novel risk cluster based on reduced sleep duration, abdominal obesity, depression, hypertension, dyslipidemia and hyperglycemia. However, the association between CircS and chronic kidney disease (CKD) remains unclear. To investigate the cross-sectional and longitudinal association between CircS and CKD, this study was performed. Methods: A national prospective cohort (China Health and Retirement Longitudinal Study, CHARLS) was used in this study. To define CKD, the estimated glomerular filtration rate (eGFR) was calculated based on the 2012 CKD-EPI creatinine-cystatin C equation. Participants with eGFR <60 mL.min-1/1.73/m2 were diagnosed with CKD. Multivariate binary logistic regression was used to assess the cross-sectional association between CircS and CKD. Subgroup and interactive analyses were performed to determine the interactive effects of covariates. In the sensitivity analysis, the obese population was excluded and another method for calculating the eGFR was used to verify the robustness of previous findings. In addition, participants without CKD at baseline were followed up for four years to investigate the longitudinal relationship between CircS and CKD. Results: A total of 6355 participants were included in this study. In the full model, CircS was positively associated with CKD (OR = 1.28, 95% CI = 1.04-1.59, P < 0.05). As per one increase of CircS components, there was a 1.11-fold (95% CI = 1.04-1.18, P < 0.05) risk of prevalent CKD in the full model. A significant interactive effect of hyperuricemia in the CircS-CKD association (P for interaction < 0.01) was observed. Sensitivity analyses excluding the obese population and using the 2009 CKD-EPI creatinine equation to diagnose CKD supported the positive correlation between CircS and CKD. In the 2011-2015 follow-up cohort, the CircS group had a 2.18-fold risk of incident CKD (95% CI = 1.33-3.58, P < 0.01) in the full model. The OR was 1.29 (95% CI = 1.10-1.51, P < 0.001) with per one increase of CircS components. Conclusion: CircS is a risk factor for CKD and may serve as a predictor of CKD for early identification and intervention.


Subject(s)
Glomerular Filtration Rate , Renal Insufficiency, Chronic , Humans , Male , Female , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/physiopathology , Middle Aged , Follow-Up Studies , Aged , Cross-Sectional Studies , Longitudinal Studies , Prospective Studies , China/epidemiology , Risk Factors , Aging/physiology , Chronobiology Disorders/complications , Chronobiology Disorders/epidemiology
6.
Sci Rep ; 14(1): 10777, 2024 05 11.
Article in English | MEDLINE | ID: mdl-38734687

ABSTRACT

Emerging evidence has documented that circadian rhythm disorders could be related to cardiovascular diseases. However, there is limited knowledge on the direct adverse effects of circadian misalignment on the heart. This study aimed to investigate the effect of chronic circadian rhythm disorder on heart homeostasis in a mouse model of consistent jetlag. The jetlag model was induced in mice by a serial 8-h phase advance of the light cycle using a light-controlled isolation box every 4 days for up to 3 months. Herein, we demonstrated for the first time that chronic circadian rhythm disorder established in the mouse jetlag model could lead to HFpEF-like phenotype such as cardiac hypertrophy, cardiac fibrosis, and cardiac diastolic dysfunction, following the attenuation of the Clock-sGC-cGMP-PKG1 signaling. In addition, clock gene knock down in cardiomyocytes induced hypertrophy via decreased sGC-cGMP-PKG signaling pathway. Furthermore, treatment with an sGC-activator riociguat directly attenuated the adverse effects of jetlag model-induced cardiac hypertrophy, cardiac fibrosis, and cardiac diastolic dysfunction. Our data suggest that circadian rhythm disruption could induce HFpEF-like phenotype through downregulation of the clock-sGC-cGMP-PKG1 signaling pathway. sGC could be one of the molecular targets against circadian rhythm disorder-related heart disease.


Subject(s)
CLOCK Proteins , Chronobiology Disorders , Cyclic GMP , Heart Failure , Soluble Guanylyl Cyclase , Animals , Male , Mice , Chronobiology Disorders/complications , Chronobiology Disorders/metabolism , Circadian Rhythm/physiology , CLOCK Proteins/metabolism , CLOCK Proteins/genetics , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Disease Models, Animal , Heart Failure/metabolism , Heart Failure/etiology , Heart Failure/physiopathology , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Phenotype , Signal Transduction , Soluble Guanylyl Cyclase/metabolism , Stroke Volume
8.
Curr Opin Neurol ; 37(4): 414-420, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38809245

ABSTRACT

PURPOSE OF REVIEW: This review aimed to comprehensively outline sleep and circadian rhythm abnormalities in hyperkinetic movement disorders beyond Parkinson's disease and atypical parkinsonisms, including tremor, dystonia, choreiform movements, tics, and ataxia disorders. RECENT FINDINGS: Insomnia, poor sleep quality, and excessive daytime sleepiness (EDS) are commonly reported in essential tremor, Wilson's disease, tics or Tourette's syndrome, and spinocerebellar ataxia (SCA). REM sleep behavior disorder (RBD) have been observed in Wilson's disease and SCA. A combination of REM and non-REM parasomnias, along with nocturnal stridor with the initiation of sleep and re-entering after awakening, are characterized by undifferentiated Non-REM and poorly structured N2 in anti-IgLON5 disease. Restless legs syndrome (RLS) has been reported commonly in SCAs. Sleep-related dyskinesia has been reported in ADCY5-related disease and GNAO1-related movement disorder. SUMMARY: Sleep problems can manifest as a result of movement disorders, either through direct motor disturbances or secondary nonmotor symptoms. Medication effects must be considered, as certain medications for movement disorders can exacerbate or alleviate sleep disturbances. Distinguishing sleep problems in some diseases might involve pathognomonic symptoms and signs, aiding in the diagnosis of movement disorders.


Subject(s)
Movement Disorders , Sleep Wake Disorders , Humans , Movement Disorders/physiopathology , Movement Disorders/etiology , Sleep Wake Disorders/physiopathology , Sleep Wake Disorders/complications , Parkinson Disease/complications , Parkinson Disease/physiopathology , Chronobiology Disorders/physiopathology , Chronobiology Disorders/complications
9.
Trials ; 25(1): 57, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38229181

ABSTRACT

BACKGROUND: Patients with mental disorders have a higher prevalence of sleep problems than the general population. Sleep problems may include insomnia, circadian rhythm disorders, or hypersomnia. A transdiagnostic approach combining cognitive behavioral therapy for insomnia (CBT-I) with chronotherapy addressing a broad range of sleep problems has shown promising results in a limited number of studies. The aim of the study is to investigate the efficacy of a transdiagnostic sleep intervention for patients with sleep problems comorbid to bipolar disorder, unipolar depression, or attention deficit disorders. The primary hypothesis is that the intervention improves sleep quality compared with a control group. The secondary hypotheses are that the intervention increases subjective and objective sleep efficiency, reduces sleep onset latency, wake after sleep onset, number of awakenings, and severity of insomnia; and that it improves well-being, personal recovery, work ability, and consumption of sleep medication compared with a control group. METHODS: The study is a randomized controlled trial enrolling 88 outpatients with bipolar disorder, major depression, or attention deficit disorder with symptoms of various sleep problems (insomnia, circadian rhythm disorders, or hypersomnia). Patients are allocated to either an intervention group receiving six sessions of transdiagnostic sleep treatment or to a control group receiving a single session of sleep hygiene education. Assessments are made at baseline, at week two, and after 6 weeks in both groups. Actigraphy is performed continuously throughout the 6-week study period for all patients. The primary outcome is changes in the subjective appraisal of sleep quality (Pittsburgh Sleep Quality Index). The secondary outcomes are changes in sleep efficiency, sleep onset latency, wake after sleep onset, number of nocturnal awakenings (based on actigraph and sleep diary data), changes in insomnia severity (Insomnia Severity Index), well-being (WHO-5 Well-Being Index), personal recovery (INSPIRE-O), work ability (Work Ability Index), and consumption of sleep medication (sleep-diaries). DISCUSSION: The study was initiated in 2022 and the inclusion period will continue until mid-2024. The results may have implications for the development and implementation of additional treatment options for patients with mental disorders and comorbid sleep problems. TRIAL REGISTRATION: ClinicalTrials.gov. NCT05406414. Registered on June 6, 2022.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Bipolar Disorder , Chronobiology Disorders , Depressive Disorder, Major , Disorders of Excessive Somnolence , Sleep Initiation and Maintenance Disorders , Humans , Bipolar Disorder/diagnosis , Bipolar Disorder/therapy , Sleep Initiation and Maintenance Disorders/diagnosis , Sleep Initiation and Maintenance Disorders/therapy , Sleep Initiation and Maintenance Disorders/complications , Attention Deficit Disorder with Hyperactivity/complications , Outpatients , Sleep , Depressive Disorder, Major/complications , Disorders of Excessive Somnolence/complications , Chronobiology Disorders/complications , Treatment Outcome , Randomized Controlled Trials as Topic
10.
Curr Hypertens Rep ; 26(1): 31-42, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37837518

ABSTRACT

PURPOSE OF REVIEW: Misalignment between the endogenous biological timing system and behavioral activities (i.e., sleep/wake, eating, activity) contributes to adverse cardiovascular health. In this review, we discuss the effects of recurring circadian misalignment on blood pressure regulation and the implications for hypertension development. Additionally, we highlight emerging therapeutic approaches designed to mitigate the negative cardiovascular consequences elicited by circadian disruption. RECENT FINDINGS: Circadian misalignment elicited by work schedules that require individuals to be awake during the biological night (i.e., shift work) alters 24-h blood pressure rhythms. Mechanistically, circadian misalignment appears to alter blood pressure via changes in autonomic nervous system balance, variations to sodium retention, dysregulation of endothelial vasodilatory responsiveness, and activation of proinflammatory mechanisms. Recurring circadian misalignment produced by a mismatch in sleep timing on free days vs. work days (i.e., social jetlag) appears to have no direct effects on prevailing blood pressure levels in healthy adults; though, circadian disruptions resulting from social jetlag may increase the risk of hypertension through enhanced sympathetic activation and/or obesity. Furthermore, social jetlag assessment may be a useful metric in shift work populations where the magnitude of circadian misalignment may be greater than in the general population. Circadian misalignment promotes unfavorable changes to 24-h blood pressure rhythms, most notably in shift working populations. While light therapy, melatonin supplementation, and the timing of drug administration may improve cardiovascular outcomes, interventions designed to target the effects of circadian misalignment on blood pressure regulation are warranted.


Subject(s)
Chronobiology Disorders , Hypertension , Adult , Humans , Blood Pressure , Circadian Rhythm/physiology , Chronobiology Disorders/complications , Sleep/physiology
11.
Biomed Pharmacother ; 166: 115368, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37647688

ABSTRACT

Diabetic retinopathy (DR) is a severe microvascular complication of diabetes mellitus and a major cause of blindness in young adults. Multiple potential factors influence DR; however, the exact mechanisms are poorly understood. Advanced treatments for DR, including laser therapy, vitrectomy, and intraocular drug injections, slow the disease's progression but fail to cure or reverse visual impairment. Therefore, additional effective methods to prevent and treat DR are required. The biological clock plays a crucial role in maintaining balance in the circadian rhythm of the body. Poor lifestyle habits, such as irregular routines and high-fat diets, may disrupt central and limbic circadian rhythms. Disrupted circadian rhythms can result in altered glucose metabolism and obesity. Misaligned central and peripheral clocks lead to a disorder of the rhythm of glucose metabolism, and chronically high sugar levels lead to the development of DR. We observed a disturbance in clock function in patients with diabetes, and a misaligned clock could accelerate the development of DR. In the current study, we examine the relationship between circadian rhythm disorders, diabetes, and DR. We conclude that: 1) abnormal function of the central clock and peripheral clock leads to abnormal glucose metabolism, further causing DR and 2) diabetes causes abnormal circadian rhythms, further exacerbating DR. Thus, our study presents new insights into the prevention and treatment of DR.


Subject(s)
Chronobiology Disorders , Diabetes Mellitus , Diabetic Retinopathy , Young Adult , Humans , Diabetic Retinopathy/etiology , Chronobiology Disorders/complications , Biological Clocks , Circadian Rhythm , Glucose
12.
Eur J Neurosci ; 57(1): 178-200, 2023 01.
Article in English | MEDLINE | ID: mdl-36342744

ABSTRACT

Parkinson's disease is a neurodegenerative disorder predominately affecting midbrain dopaminergic neurons that results in a broad range of motor and non-motor symptoms. Sleep complaints are among the most common non-motor symptoms, even in the prodromal period. Sleep alterations in Parkinson's disease patients may be associated with dysregulation of circadian rhythms, intrinsic 24-h cycles that control essential physiological functions, or with side effects from levodopa medication and physical and mental health challenges. The impact of circadian dysregulation on sleep disturbances in Parkinson's disease is not fully understood; as such, we review the systems, cellular and molecular mechanisms that may underlie circadian perturbations in Parkinson's disease. We also discuss the potential benefits of chronobiology-based personalized medicine in the management of Parkinson's disease both in terms of behavioural and pharmacological interventions. We propose that a fuller understanding of circadian clock function may shed important new light on the aetiology and symptomatology of the disease and may allow for improvements in the quality of life for the millions of people with Parkinson's disease.


Subject(s)
Chronobiology Disorders , Neurodegenerative Diseases , Parkinson Disease , Humans , Quality of Life , Chronobiology Disorders/complications , Sleep/physiology , Circadian Rhythm/physiology
13.
Nervenarzt ; 93(9): 873-881, 2022 Sep.
Article in German | MEDLINE | ID: mdl-35881187

ABSTRACT

BACKGROUND: Numerous symptoms of bipolar disorder are regulated by the circadian rhythm. Because of this association it is assumed that disruption of the circadian rhythm may be part of the pathomechanism of bipolar disorder. OBJECTIVES: A comparison and subsequent critical discussion of the current data situation on chronobiological aspects of bipolar disorder are presented. METHODS: A narrative literature search was carried out and the main findings are presented in a summarized form. RESULTS: There are a large number of animal and human studies investigating the connection between disorders of the circadian rhythm and bipolar disorder. Especially chronotype, the environmental factor light and sleep disorders seem to be associated with the development of bipolar disorder. CONCLUSIONS: The neurobiology of bipolar disorder shows numerous chronobiological aspects. There is evidence for a direct connection of disruption of the circadian rhythm and development and progression of bipolar disorder; however, at present there is no proof for the specificity of these findings for bipolar disorder. Future studies should consolidate the evidence on the impact of disorders of the circadian rhythm on the pathomechanism of bipolar disorder.


Subject(s)
Bipolar Disorder , Chronobiology Disorders , Sleep Wake Disorders , Animals , Bipolar Disorder/complications , Bipolar Disorder/diagnosis , Chronobiology Disorders/complications , Chronobiology Disorders/diagnosis , Circadian Rhythm , Humans , Sleep , Sleep Wake Disorders/diagnosis
14.
Sci Rep ; 12(1): 2434, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35165331

ABSTRACT

Emerging evidence suggests that disruption of circadian rhythmicity contributes to development of comorbid depression, cardiovascular diseases (CVD), and type 2 diabetes mellitus (T2DM). Physical exercise synchronizes the circadian system and has ameliorating effects on the depression- and anxiety-like phenotype induced by circadian disruption in mice and sand rats. We explored the beneficial effects of voluntary wheel running on daily rhythms, and the development of depression, T2DM, and CVD in a diurnal animal model, the fat sand rat (Psammomys obesus). Voluntary exercise strengthened general activity rhythms, improved memory and lowered anxiety- and depressive-like behaviors, enhanced oral glucose tolerance, and decreased plasma insulin levels and liver weight. Animals with access to a running wheel had larger heart weight and heart/body weight ratio, and thicker left ventricular wall. Our results demonstrate that exercising ameliorates pathological-like daily rhythms in activity and blood glucose levels, glucose tolerance and depressive- and anxiety-like behaviors in the sand rat model, supporting the important role of physical activity in modulating the "circadian syndrome" and circadian rhythm-related diseases. We suggest that the utilization of a diurnal rodent animal model may offer an effective way to further explore metabolic, cardiovascular, and affective-like behavioral changes related to chronodisruption and their underlying mechanisms.


Subject(s)
Cardiovascular Diseases/complications , Cardiovascular Diseases/therapy , Chronobiology Disorders/complications , Chronobiology Disorders/therapy , Circadian Rhythm , Depression/complications , Depression/therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/therapy , Exercise Therapy/methods , Physical Conditioning, Animal/methods , Animals , Anxiety/complications , Anxiety/physiopathology , Anxiety/therapy , Blood Glucose/analysis , Cardiovascular Diseases/physiopathology , Chronobiology Disorders/physiopathology , Depression/physiopathology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Gerbillinae , Glucose Tolerance Test , Insulin/blood , Locomotion , Male , Rats , Suprachiasmatic Nucleus/physiopathology , Treatment Outcome
15.
Food Funct ; 13(3): 1048-1061, 2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35050270

ABSTRACT

Circadian rhythm is essential to human physiological homeostasis and health. The oscillation of host circadian rhythm affects the composition and function of intestinal microbiota, meanwhile, the normal operation of host circadian rhythm depends on the diurnal changes of intestinal microbiota. The imbalance of intestinal micro-ecology or the disorder of host circadian rhythm may lead to psychiatric disorders, while the intervention of plant polysaccharides is a possible way to alleviate circadian rhythm disturbance and the related psychiatric diseases. This review discusses the interaction between host circadian rhythm and intestinal microbiota and their effects on psychiatric disorders, and proposes a possible strategy of plant polysaccharides to alleviate circadian rhythm disorders and related psychiatric disorders by regulating intestinal micro-ecology.


Subject(s)
Chronobiology Disorders/complications , Chronobiology Disorders/metabolism , Gastrointestinal Microbiome/physiology , Mental Disorders/complications , Mental Disorders/metabolism , Plants/metabolism , Polysaccharides/metabolism , Animals , Chronobiology Disorders/physiopathology , Circadian Rhythm , Cues , Homeostasis , Humans , Intestines/metabolism , Intestines/physiopathology , Mental Disorders/physiopathology , Mice , Polysaccharides/physiology
16.
Encephale ; 48(3): 325-334, 2022 Jun.
Article in French | MEDLINE | ID: mdl-34916075

ABSTRACT

INTRODUCTION: Sleep disorders are prevalent in patients with a neurocognitive disorder, and diagnosis and treatment in these patients remain challenging in clinical practice. METHODS: This narrative review offers a systematic approach to diagnose and treat sleep disorders in neurocognitive disorders. RESULTS: Alzheimer's disease is often associated with circadian rhythm disorders, chronic insomnia, and sleep apnea-hypopnea syndrome. Alpha-synucleinopathies (e.g., Parkinson's disease and Lewy body dementia) are often associated with a rapid eye movement sleep behavior disorder, restless legs syndrome, chronic insomnia, and sleep apnea-hypopnea syndrome. A focused history allows to diagnose most sleep disorders. Clinicians should ensure to gather the following information in all patients with a neurocognitive disorder: (1) the presence of difficulties falling asleep or staying asleep, (2) the impact of sleep disturbances on daily functioning (fatigue, sleepiness and other daytime consequences), and (3) abnormal movements in sleep. Sleep diaries and questionnaires can assist clinicians in screening for specific sleep disorders. Polysomnography is recommended if a rapid eye movement sleep behavior disorder or a sleep apnea-hypopnea syndrome are suspected. Sleep complaints should prompt clinicians to ensure that comorbidities interfering with sleep are properly managed. The main treatment for moderate to severe obstructive sleep apnea-hypopnea syndrome remains continuous positive airway pressure, as its efficacy has been demonstrated in patients with neurocognitive disorders. Medications should also be reviewed, and time of administration should be optimized (diuretics and stimulating medications in the morning, sedating medications in the evening). Importantly, cholinesterase inhibitors (especially donepezil) may trigger insomnia. Switching to morning dosing or to an alternative drug may help. Cognitive-behavioral therapy for insomnia is indicated to treat chronic insomnia in neurocognitive disorders. False beliefs regarding sleep should be addressed with the patient and their caregiver. The sleep environment should be optimized (decrease light exposure at night, minimize noise, avoid taking vital signs, etc.). Sleep restriction can be considered as patients with a neurocognitive disorder often spend too much time in bed. The need for naps should be assessed case by case as naps may contribute to insomnia in some patients but allow others to complete their diurnal activities. Trazodone (50mg) may also be used under certain circumstances in chronic insomnia. Recent evidence does not support a role for exogenous melatonin in patients with a neucognitive disorder and insomnia. Patients in long-term care facilities are often deprived of an adequate diurnal exposure to light. Increasing daytime exposure to light may improve sleep and mood. Patients with circadian rhythm disorders can also benefit from light therapy (morning bright light therapy in case of phase delay and evening bright light therapy in case of phase advance). Rapid eye movement sleep behavior disorder can lead to violent behaviors, and the sleeping environment should be secured (e.g., mattress on the floor, remove surrounding objects). Medication exacerbating this disorder should be stopped if possible. High dose melatonin (6 to 18mg) or low dose clonazepam (0.125-0.25mg) at bedtime may be used to reduce symptoms. Melatonin is preferred in first-line as it is generally well tolerated with few side effects. Patients with restless legs syndrome should be investigated for iron deficiency. Medication decreasing dopaminergic activity should be reduced or stopped if possible. Behavioral strategies such as exercise and leg massages may be beneficial. Low-dose dopamine agonists (such as pramipexole 0.125mg two hours before bedtime) can be used to treat the condition, but a prolonged treatment may paradoxically worsen the symptoms. Alpha-2-delta calcium channel ligands can also be used while monitoring for the risk of falls. CONCLUSION: Multiple and sustained nonpharmacological approaches are recommended for the treatment of sleep disturbances in patients with neurocognitive disorder. Pharmacological indications remain limited, and further randomized clinical trials integrating a multimodal approach are warranted to evaluate the treatment of sleep disorders in specific neurocognitive disorders.


Subject(s)
Alzheimer Disease , Chronobiology Disorders , Melatonin , REM Sleep Behavior Disorder , Restless Legs Syndrome , Sleep Apnea Syndromes , Sleep Initiation and Maintenance Disorders , Sleep Wake Disorders , Alzheimer Disease/complications , Alzheimer Disease/therapy , Chronobiology Disorders/chemically induced , Chronobiology Disorders/complications , Chronobiology Disorders/drug therapy , Humans , Melatonin/therapeutic use , REM Sleep Behavior Disorder/chemically induced , REM Sleep Behavior Disorder/complications , REM Sleep Behavior Disorder/drug therapy , Restless Legs Syndrome/complications , Restless Legs Syndrome/diagnosis , Restless Legs Syndrome/drug therapy , Sleep , Sleep Apnea Syndromes/chemically induced , Sleep Apnea Syndromes/complications , Sleep Apnea Syndromes/drug therapy , Sleep Initiation and Maintenance Disorders/drug therapy , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/therapy
17.
Front Endocrinol (Lausanne) ; 12: 723918, 2021.
Article in English | MEDLINE | ID: mdl-34745000

ABSTRACT

Background: Hepatic lipid metabolism regulates biliary composition and influences the formation of cholesterol gallstones. The genes Hmgcr and Cyp7a1, which encode key liver enzymes, are regulated by circadian rhythm-related transcription factors. We aimed to investigate the effect of circadian rhythm disruption on hepatic cholesterol and bile acid metabolism and the incidence of cholesterol stone formation. Methods: Adult male C57BL/6J mice were fed either a lithogenic diet (LD) only during the sleep phase (time-restricted lithogenic diet feeding, TRF) or an LD ad libitum (non-time-restricted lithogenic diet feeding, nTRF) for 4 weeks. Food consumption, body mass gain, and the incidence of gallstones were assessed. Circulating metabolic parameters, lipid accumulation in the liver, the circadian expression of hepatic clock and metabolic genes, and the gut microbiota were analyzed. Results: TRF caused a dysregulation of the circadian rhythm in the mice, characterized by significant differences in the circadian expression patterns of clock-related genes. In TRF mice, the circadian rhythms in the expression of genes involved in bile acid and cholesterol metabolism were disrupted, as was the circadian rhythm of the gut microbiota. These changes were associated with high biliary cholesterol content, which promoted gallstone formation in the TRF mice. Conclusion: Disordered circadian rhythm is associated with abnormal hepatic bile acid and cholesterol metabolism in mice, which promotes gallstone formation.


Subject(s)
Chronobiology Disorders/complications , Gallstones/etiology , Gastrointestinal Microbiome , Lipid Metabolism , Liver/metabolism , Animals , Cholesterol/metabolism , Chronobiology Disorders/etiology , Chronobiology Disorders/metabolism , Chronobiology Disorders/microbiology , Circadian Rhythm/physiology , Diet/adverse effects , Gallstones/metabolism , Gallstones/microbiology , Gastrointestinal Microbiome/physiology , Lipid Metabolism/physiology , Male , Mice , Mice, Inbred C57BL , Time Factors
18.
J Mol Histol ; 52(4): 799-808, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34131827

ABSTRACT

Defective autophagy in vascular smooth muscle cells (VSMCs) in response to oxidative stress can lead to cellular apoptosis and plaque instability. Previous studies have revealed that the circadian clock system is involved in autophagic regulation and plaque progression. However, the mechanism by which circadian rhythmicity influences VSMC autophagy and plaque stability remains unclear. Our study described the circadian profiles in atheromatous plaques and verified the role of circadian misalignment in VSMC autophagy and apoptosis. We found that the mRNA expression levels of circadian locomotor output cycles protein kaput (CLOCK) and Beclin 1 were significantly decreased in unstable plaques compared with stable plaques. No significant differences were observed in other circadian rhythm genes. VSMCs treated with oxidized low-density lipoprotein (ox-LDL, 80 µg/ml) exhibited abnormal circadian rhythmicity and impaired autophagy, as evidenced by consistent decreases in CLOCK and Beclin 1 expression, suggesting a correlation between CLOCK and autophagy. CLOCK protein expression was inhibited by ox-LDL, accompanied by defective autophagy and an increased apoptosis rates (P < 0.05). Administration of rapamycin (10 nM) reversed the effect of ox-LDL on VSMC autophagy and apoptosis. Finally, CLOCK silencing led to a considerable decrease in autophagy. VSMCs with stable CLOCK silencing also showed an increased apoptosis rate. In addition, gene silencing of CLOCK in VSMCs counteracted the effects of moderate rapamycin concentrations on autophagy and apoptosis. In conclusion, these findings suggested that the CLOCK-dependent rapamycin signaling pathway is a critical mediator in ox-LDL-induced VSMCs with defective autophagy that exacerbates plaque destabilization.


Subject(s)
Apoptosis , Autophagy , Beclin-1/genetics , CLOCK Proteins/genetics , Chronobiology Disorders/complications , Gene Expression Regulation/physiology , Muscle, Smooth, Vascular/pathology , Aorta/cytology , Blotting, Western , Cadaverine/analogs & derivatives , Cadaverine/metabolism , Cells, Cultured , Humans , Lipoproteins, LDL/pharmacology , Male , Middle Aged , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Transfection
19.
Curr Top Behav Neurosci ; 48: 133-147, 2021.
Article in English | MEDLINE | ID: mdl-33547594

ABSTRACT

Symptoms of affective disorders encompass a range of changes to biological processes such as sleep and appetite. These processes are regulated over a 24-h cycle known as the circadian rhythm. Sleep is a particularly useful marker of this rhythm as it is readily measurable and functionally significant. Sleep disturbance is common in bipolar affective disorder and may act as a marker, and precipitant, of relapse. Circadian rhythms are modulated by environmental and social cues and have been shown to be influenced by treatment in BPAD. As such understanding of circadian rhythms may lead to a better understanding of the pathophysiology of BPAD and its treatment. This chapter will explore the neurobiology of the circadian clock and the putative role of circadian rhythm dysregulation in the pathophysiology and treatment of bipolar affective disorder (BPAD).


Subject(s)
Bipolar Disorder , Chronobiology Disorders , Chronobiology Disorders/complications , Circadian Rhythm , Humans , Mood Disorders , Sleep
20.
Life Sci ; 262: 118512, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33010281

ABSTRACT

Circadian rhythms play an important role in a wide range of human physiology and pathology. Individuals increasingly experience situations such as night-shift work schedules, likely leading to circadian disruption. Recent studies have also demonstrated that patients with other diseases often show symptoms of circadian disruption as manifested by the sleep-wake cycle and other biological rhythms. Circadian disruption often results in changes to the phase, period, and amplitude of the sleep-wake cycle, melatonin rhythm, and core body temperature. Several cardiometabolic, psychiatric, and neurodegenerative diseases are closely related to circadian disruption. Several interventions are also available, including phototherapy, exogenous melatonin, and exercise. The cumulative findings suggest that circadian disruption can increase risk for some cardiometabolic diseases. Circadian disruption also acts as a concomitant symptom of several psychiatric and neurodegenerative diseases. More attention should be paid to evaluating the impact of circadian disruption on these related diseases, as well as the benefits of the mitigation interventions for both circadian disruption and related diseases.


Subject(s)
Chronobiology Disorders/physiopathology , Circadian Rhythm/physiology , Animals , Chronobiology Disorders/complications , Chronobiology Disorders/therapy , Exercise/physiology , Humans , Melatonin/administration & dosage , Phototherapy/methods
SELECTION OF CITATIONS
SEARCH DETAIL