Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
J Neurodev Disord ; 14(1): 33, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35610565

ABSTRACT

BACKGROUND: Regulator of calcineurin 1 (RCAN1) is overexpressed in Down syndrome (DS), but RCAN1 levels are also increased in Alzheimer's disease (AD) and normal aging. AD is highly comorbid among individuals with DS and is characterized in part by progressive neurodegeneration that resembles accelerated aging. Importantly, abnormal RCAN1 levels have been demonstrated to promote memory deficits and pathophysiology that appear symptomatic of DS, AD, and aging. Anomalous diurnal rest-activity patterns and circadian rhythm disruptions are also common in DS, AD, and aging and have been implicated in facilitating age-related cognitive decline and AD progression. However, no prior studies have assessed whether RCAN1 dysregulation may also promote the age-associated alteration of rest-activity profiles and circadian rhythms, which could in turn contribute to neurodegeneration in DS, AD, and aging. METHODS: The present study examined the impacts of RCAN1 deficiency and overexpression on the photic entrainment, circadian periodicity, intensity and distribution, diurnal patterning, and circadian rhythmicity of wheel running in young (3-6 months old) and aged (9-14 months old) mice of both sexes. RESULTS: We found that daily RCAN1 levels in the hippocampus and suprachiasmatic nucleus (SCN) of light-entrained young mice are generally constant and that balanced RCAN1 expression is necessary for normal circadian locomotor activity rhythms. While the light-entrained diurnal period was unaltered, RCAN1-null and RCAN1-overexpressing mice displayed lengthened endogenous (free-running) circadian periods like mouse models of AD and aging. In light-entrained young mice, RCAN1 deficiency and overexpression also recapitulated the general hypoactivity, diurnal rest-wake pattern fragmentation, and attenuated amplitudes of circadian activity rhythms reported in DS, preclinical and clinical AD, healthily aging individuals, and rodent models thereof. Under constant darkness, RCAN1-null and RCAN1-overexpressing mice displayed altered locomotor behavior indicating circadian clock dysfunction. Using the Dp(16)1Yey/+ (Dp16) mouse model for DS, which expresses three copies of Rcan1, we found reduced wheel running activity and rhythmicity in both light-entrained and free-running young Dp16 mice like young RCAN1-overexpressing mice. Critically, these diurnal and circadian deficits were rescued in part or entirely by restoring Rcan1 to two copies in Dp16 mice. We also found that RCAN1 deficiency but not RCAN1 overexpression altered protein levels of the clock gene Bmal1 in the SCN. CONCLUSIONS: Collectively, this study's findings suggest that both loss and aberrant gain of RCAN1 precipitate anomalous light-entrained diurnal and circadian activity patterns emblematic of DS, AD, and possibly aging.


Subject(s)
Aging , Alzheimer Disease , Calcium-Binding Proteins , Chronobiology Disorders , DNA-Binding Proteins , Down Syndrome , Muscle Proteins , Aging/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Chronobiology Disorders/genetics , Chronobiology Disorders/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/metabolism , Female , Male , Mice , Motor Activity/physiology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Suprachiasmatic Nucleus/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Bipolar Disord ; 24(3): 232-263, 2022 05.
Article in English | MEDLINE | ID: mdl-34850507

ABSTRACT

AIM: Symptoms of bipolar disorder (BD) include changes in mood, activity, energy, sleep, and appetite. Since many of these processes are regulated by circadian function, circadian rhythm disturbance has been examined as a biological feature underlying BD. The International Society for Bipolar Disorders Chronobiology Task Force (CTF) was commissioned to review evidence for neurobiological and behavioral mechanisms pertinent to BD. METHOD: Drawing upon expertise in animal models, biomarkers, physiology, and behavior, CTF analyzed the relevant cross-disciplinary literature to precisely frame the discussion around circadian rhythm disruption in BD, highlight key findings, and for the first time integrate findings across levels of analysis to develop an internally consistent, coherent theoretical framework. RESULTS: Evidence from multiple sources implicates the circadian system in mood regulation, with corresponding associations with BD diagnoses and mood-related traits reported across genetic, cellular, physiological, and behavioral domains. However, circadian disruption does not appear to be specific to BD and is present across a variety of high-risk, prodromal, and syndromic psychiatric disorders. Substantial variability and ambiguity among the definitions, concepts and assumptions underlying the research have limited replication and the emergence of consensus findings. CONCLUSIONS: Future research in circadian rhythms and its role in BD is warranted. Well-powered studies that carefully define associations between BD-related and chronobiologically-related constructs, and integrate across levels of analysis will be most illuminating.


Subject(s)
Bipolar Disorder , Chronobiology Disorders , Animals , Behavioral Research , Bipolar Disorder/diagnosis , Chronobiology Disorders/genetics , Circadian Rhythm/genetics , Humans , Sleep/physiology
3.
Exp Eye Res ; 211: 108751, 2021 10.
Article in English | MEDLINE | ID: mdl-34478739

ABSTRACT

Diabetic retinopathy is a major complication of chronic hyperglycemia and a leading cause of blindness in developed countries. In the present study the interaction between diabetes and retinal clocks was investigated in mice. It was seen that in the db/db mouse - a widely used animal model of diabetic retinopathy - clock function and circadian regulation of gene expression was disturbed in the retina. Remarkably, elimination of clock function by Bmal1-deficiency mitigates the progression of pathophysiology of the diabetic retina. Thus high-fat diet was seen to induce histopathology and molecular markers associated with diabetic retinopathy in wild type but not in Bmal1-deficient mice. The data of the present study suggest that Bmal1/the retinal clock system is both, a target and an effector of diabetes mellitus in the retina and hence represents a putative therapeutic target in the pathogenesis of diabetic retinopathy.


Subject(s)
Chronobiology Disorders/physiopathology , Circadian Clocks/physiology , Circadian Rhythm/physiology , Diabetic Retinopathy/physiopathology , Animals , Blood Glucose/metabolism , CLOCK Proteins/genetics , Chronobiology Disorders/genetics , Diabetic Retinopathy/genetics , Disease Models, Animal , Female , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , RNA, Messenger/genetics , RNA, Ribosomal, 18S/genetics
4.
J Pineal Res ; 70(3): e12726, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33638890

ABSTRACT

Circadian disruption has been identified as a risk factor for health disorders such as obesity, cardiovascular disease, and cancer. Although epidemiological studies suggest an increased risk of various cancers associated with circadian misalignment due to night shift work, the underlying mechanisms have yet to be elucidated. We sought to investigate the potential mechanistic role that circadian disruption of cancer hallmark pathway genes may play in the increased cancer risk in shift workers. In a controlled laboratory study, we investigated the circadian transcriptome of cancer hallmark pathway genes and associated biological pathways in circulating leukocytes obtained from healthy young adults during a 24-hour constant routine protocol following 3 days of simulated day shift or night shift. The simulated night shift schedule significantly altered the normal circadian rhythmicity of genes involved in cancer hallmark pathways. A DNA repair pathway showed significant enrichment of rhythmic genes following the simulated day shift schedule, but not following the simulated night shift schedule. In functional assessments, we demonstrated that there was an increased sensitivity to both endogenous and exogenous sources of DNA damage after exposure to simulated night shift. Our results suggest that circadian dysregulation of DNA repair may increase DNA damage and potentiate elevated cancer risk in night shift workers.


Subject(s)
Biomarkers, Tumor/genetics , Chronobiology Disorders/etiology , Circadian Rhythm , DNA Damage , DNA Repair , Neoplasms/etiology , Shift Work Schedule/adverse effects , Transcriptome , Activity Cycles , Adult , Chronobiology Disorders/genetics , Chronobiology Disorders/physiopathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasms/genetics , Neoplasms/pathology , Risk Assessment , Risk Factors , Sleep , Time Factors , Young Adult
5.
Med Oncol ; 37(10): 90, 2020 Sep 14.
Article in English | MEDLINE | ID: mdl-32926243

ABSTRACT

Endometrial cancer (EC) is one of the most common gynecologic malignancies, and the incidence rate of night shift among women workers is higher than that in the general population. Circadian rhythm disorder, mainly rhythm gene, is related to various tumor onset, including EC. This study described the sleep/night-shift features of EC patients, explored the mechanism of the circadian clock gene PER and investigated prognostic and functional values of Per1 caused by night shift. A total of 619 subjects were enrolled and divided into two groups according to night-shift duties (rhythm group and control group), analyzed for clinical risk factors and night shift features of endometrial carcinoma. Then samples were randomly selected for sequencing and western blot were performed, and the function of overexpressed PER1 in ishikawa cells was explored. We noticed that severer EC patients experienced night-shift more frequently and with longer durations. A total of 58,174 differentially expressed genes were discovered, mainly rhythm genes and related to up and downstream regulatory genes. Western blot showed that the rhythm group had elevated protein expression of BCAS4, TUBB2B and RSPO4, and decreased expression of PER1 and PER2 in night-shift. In TCGA-EC datasets, PER1 was decreased in the EC patients with a significantly positive correlation with PER2, and higher PER1 expression indicated longer survival, opposite to TUBB2B. The research of overexpressing PER1 gene in EC ishikawa cells found that PER1 can promote apoptosis, expression of TNF-a, IL-6 and PD-1/PD-L1, inhibit the tumor invasion and expression of TUBB2B gene. Together, EC severity was associated with night-shift and rhythm disorders. The rhythm relating factors PER1, TUBB2B and tumor immune factors may regulate the mechanisms of EC onset and progression.


Subject(s)
Endometrial Neoplasms , Period Circadian Proteins/metabolism , Sleep Disorders, Circadian Rhythm , Tubulin/metabolism , Adult , Aged , Chronobiology Disorders/genetics , Chronobiology Disorders/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Female , Gene Regulatory Networks , Humans , Middle Aged , Period Circadian Proteins/genetics , Sleep Disorders, Circadian Rhythm/genetics , Sleep Disorders, Circadian Rhythm/metabolism , Transcriptome , Tubulin/genetics
6.
Nat Commun ; 11(1): 3193, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32581213

ABSTRACT

Breast cancer is the most common type of cancer worldwide and one of the major causes of cancer death in women. Epidemiological studies have established a link between night-shift work and increased cancer risk, suggesting that circadian disruption may play a role in carcinogenesis. Here, we aim to shed light on the effect of chronic jetlag (JL) on mammary tumour development. To do this, we use a mouse model of spontaneous mammary tumourigenesis and subject it to chronic circadian disruption. We observe that circadian disruption significantly increases cancer-cell dissemination and lung metastasis. It also enhances the stemness and tumour-initiating potential of tumour cells and creates an immunosuppressive shift in the tumour microenvironment. Finally, our results suggest that the use of a CXCR2 inhibitor could correct the effect of JL on cancer-cell dissemination and metastasis. Altogether, our data provide a conceptual framework to better understand and manage the effects of chronic circadian disruption on breast cancer progression.


Subject(s)
Breast Neoplasms/pathology , Chronobiology Disorders/complications , Tumor Microenvironment/immunology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Cell Transformation, Neoplastic/drug effects , Chronic Disease , Chronobiology Disorders/genetics , Chronobiology Disorders/immunology , Cytokines/genetics , Female , Gene Expression Regulation , Immunosuppression Therapy , Light Signal Transduction/genetics , Mice , Mice, Transgenic , Neoplasm Metastasis/prevention & control , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/genetics
7.
Pflugers Arch ; 472(5): 513-526, 2020 05.
Article in English | MEDLINE | ID: mdl-32363530

ABSTRACT

Circadian rhythms are generated by the circadian clock, a self-sustained internal timing system that exhibits 24-h rhythms in the body. In mammals, circadian rhythms are driven by a central clock located in suprachiasmatic nucleus and various peripheral clocks located in different tissues and organs of the body. Many cellular, behavioral, and physiological processes are regulated by the circadian clock in coordination with environmental cues. The process of metabolism is also under circadian regulation. Loss of synchronization between the internal clock and environmental zeitgebers results in disruption of the circadian rhythms that seriously impacts metabolic homeostasis leading to changed eating behavior, altered glucose and lipid metabolism, and weight gain. This in turn augments the risk of having various cardio-metabolic disorders such as obesity, diabetes, metabolic syndrome, and cardiovascular disease. This review sheds light on circadian rhythms and their role in metabolism with the identification of gaps in the current knowledge that remain to be explored in these fields. In this review, the molecular mechanisms underlying circadian rhythms have been elaborated first. Then, the focus has been kept on explaining the physiological significance of circadian rhythms in regulating metabolism. Finally, the implications for metabolism when these rhythms are disrupted due to genetic mutations or social and occupational needs enforced by modern lifestyle have been discussed.


Subject(s)
Chronobiology Disorders/metabolism , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Circadian Rhythm , Animals , Chronobiology Disorders/genetics , Chronobiology Disorders/physiopathology , Circadian Rhythm Signaling Peptides and Proteins/genetics , Gastrointestinal Microbiome , Homeostasis , Humans
8.
Pharmacol Biochem Behav ; 193: 172915, 2020 06.
Article in English | MEDLINE | ID: mdl-32224058

ABSTRACT

Endogenous circadian rhythms govern behavior and physiology, while circadian disruption is an environmental factor that impacts cognition by altering the circadian clock at a molecular level. We modeled the effects of 2 sources of circadian disruption - activity occurring during typical rest periods and untimely light exposure - to evaluate the effects of circadian disruption on behavior and underlying neurochemistry. Firstly, adult Long-Evans rats of both sexes were maintained on a 12 h:12 h light:dark cycle and tested using a 5-choice serial reaction time task (5-CSRTT) under 3 conditions: 4 h into the dark phase with no exposure to ambient light during testing (control), 4 h into the dark phase with exposure to ambient light during testing, and 4 h into the light phase. Both models resulted in impulsive behavior and reduced attention compared to control. We established that changes in the diurnal expression pattern occur in the clock gene Period 2 (Per2) in the light phase-tested model. Choline acetyltransferase (Chat) and Dopamine receptor 1 (Drd1) showed rhythmic expression with peak expression during the dark phase regardless of light-testing condition. Next, we performed drug challenges in a new rat cohort to examine the interaction between the cholinergic and dopaminergic neurotransmitter systems in regulating the behavioral changes caused by circadian disruption. We administered the cholinergic agonist nicotine and either the dopamine-1 receptor (DR1) antagonist SCH23390 or the DR2 antagonist eticlopride under the 3 circadian conditions to identify differential drug responses between treatment groups. Rats in both models demonstrated increased sensitivity to nicotine as compared to control, while SCH23390 and eticlopride ameliorated the effect of nicotine on 5-CSRTT performance in both models. Our study is the first to identify detrimental effects of both models of circadian disruption on impulsive behavior, and that the effects of circadian disruption are mediated by an interaction between cholinergic and dopaminergic systems.


Subject(s)
Attention , Chronobiology Disorders/genetics , Circadian Clocks/genetics , Circadian Rhythm/genetics , Impulsive Behavior/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Animals , Benzazepines/pharmacology , Choline O-Acetyltransferase/genetics , Chronobiology Disorders/physiopathology , Cognition , Disease Models, Animal , Dopamine Antagonists/pharmacology , Female , Gene Expression , Male , Period Circadian Proteins/genetics , Rats , Rats, Long-Evans , Reaction Time/drug effects , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/genetics , Salicylamides/pharmacology
9.
J Biol Rhythms ; 35(1): 72-83, 2020 02.
Article in English | MEDLINE | ID: mdl-31726916

ABSTRACT

Circadian clocks regulate multiple physiological processes in the eye, but their requirement for retinal health remains unclear. We previously showed that Drosophila homologs of spliceosome proteins implicated in human retinitis pigmentosa (RP), the most common genetically inherited cause of blindness, have a role in the brain circadian clock. In this study, we report circadian phenotypes in murine models of RP. We found that mice carrying a homozygous H2309P mutation in Pre-mRNA splicing factor 8 (Prpf8) display a lengthened period of the circadian wheel-running activity rhythm. We show also that the daily cycling of circadian gene expression is dampened in the retina of Prpf8-H2309P mice. Surprisingly, molecular rhythms are intact in the eye cup, which includes the retinal pigment epithelium (RPE), even though the RPE is thought to be the primary tissue affected in this form of RP. Downregulation of Prp31, another RNA splicing factor implicated in RP, leads to period lengthening in a human cell culture model. The period of circadian bioluminescence in primary fibroblasts of human RP patients is not significantly altered. Together, these studies link a prominent retinal disorder to circadian deficits, which could contribute to disease pathology.


Subject(s)
Chronobiology Disorders/genetics , Mutation , RNA Splicing Factors/genetics , Retinitis Pigmentosa/complications , Retinitis Pigmentosa/genetics , Adult , Animals , Cells, Cultured , Chronobiology Disorders/etiology , Circadian Rhythm/genetics , Disease Models, Animal , Eye Proteins/genetics , Eye Proteins/metabolism , Fibroblasts/physiology , Humans , Luminescence , Male , Mice , Middle Aged , Retina/pathology , Retinal Pigment Epithelium/physiology , Retinitis Pigmentosa/physiopathology , Skin/cytology
10.
J Agric Food Chem ; 67(31): 8510-8519, 2019 Aug 07.
Article in English | MEDLINE | ID: mdl-31294559

ABSTRACT

Acrylamide, mainly formed in Maillard browning reaction during food processing, causes defects in liver circadian clock and mitochondrial function by inducing oxidative stress. Resveratrol is a polyphenol that has powerful antioxidant and anti-inflammatory activity. However, the preventive effects of resveratrol on acrylamide-triggered oxidative damage and circadian rhythm disorders are unclear at the current stage. The present research revealed that resveratrol pretreatment prevented acrylamide-induced cell death, mitochondrial dysfunction, and inflammatory responses in HepG2 liver cells. Acrylamide significantly triggered disorders of circadian genes transcription and protein expressions including Bmal1 and Cry 1 in primary hepatocytes, which were prevented by resveratrol pretreatment. Moreover, we found that the beneficial effects of resveratrol on stimulating Nrf2/NQO-1 pathway and mitochondrial respiration complex expressions in acrylamide-treated cells were Bmal1-dependent. Similarly, the inhibitory effects of resveratrol on inflammation signaling NF-κB were Cry1-dependent. In conclusion, these results demonstrated resveratrol could be a promising compound in suppressing acrylamide-induced hepatotoxicity and balancing the circadian clock.


Subject(s)
ARNTL Transcription Factors/immunology , Acrylamide/toxicity , Chronobiology Disorders/immunology , Cryptochromes/immunology , Hepatocytes/drug effects , Mitochondria/drug effects , Resveratrol/pharmacology , ARNTL Transcription Factors/genetics , Animals , Chronobiology Disorders/drug therapy , Chronobiology Disorders/genetics , Chronobiology Disorders/physiopathology , Circadian Rhythm/drug effects , Cryptochromes/genetics , Hep G2 Cells , Hepatocytes/immunology , Humans , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/immunology
11.
J Cell Biochem ; 120(10): 16408-16415, 2019 10.
Article in English | MEDLINE | ID: mdl-31310357

ABSTRACT

Aging process in mammals is associated with a decline in amplitude and a long period of circadian behaviors which are regulated by a central circadian regulator in the suprachiasmatic nucleus (SCN) and local oscillators in peripheral tissues. It is unclear whether enhancing clock function can retard aging. Using fibroblasts expressing per2::lucSV and senescent cells, we revealed cycloastragenol (CAG), a natural aglycone derivative from astragaloside IV, as a clock amplitude enhancing small molecule. CAG could activate telomerase to antiaging, but no reports focused on its effects on circadian rhythm disorders in aging mice. Here we analyze the potential effects of CAG on d-galactose-induced aging mice on the circadian behavior and expression of clock genes. For this purpose, CAG (20 mg/kg orally), was administered daily to d-galactose (150 mg/kg, subcutaneous) mice model of aging for 6 weeks. An actogram analysis of free-running activity of these mice showed that CAG significantly enhances the locomotor activity. We further found that CAG increase expressions of per2 and bmal1 genes in liver and kidney of aging mouse. Furthermore, CAG enhanced clock protein BMAL1 and PER2 levels in aging mouse liver and SCN. Our results indicated that the CAG could restore the behavior of circadian rhythm in aging mice induced by d-galactose. These data of present study suggested that CAG could be used as a novel therapeutic strategy for the treatment of age-related circadian rhythm disruption.


Subject(s)
Aging/metabolism , Chronobiology Disorders/prevention & control , Galactose/toxicity , Sapogenins/pharmacology , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Aging/genetics , Animals , Chronobiology Disorders/chemically induced , Chronobiology Disorders/genetics , Chronobiology Disorders/metabolism , Mice , Mice, Transgenic , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism
12.
J Alzheimers Dis ; 69(2): 521-528, 2019.
Article in English | MEDLINE | ID: mdl-30958375

ABSTRACT

BACKGROUND: Sundown syndrome (SS) in patients with Alzheimer's disease (AD) is characterized by aggravation of behavioral problems at sunset. Disturbance of the circadian rhythm, a possible cause of SS, also facilitates amyloidopathy and reduces sleep quality. However, the associations of SS with amyloidopathy and sleep quality remain unclear. OBJECTIVE: To investigate the prevalence of SS in patients with AD, the association between SS and APOEɛ4 carrier, representing an enhanced amyloid pathology, and the relationship between SS and sleep quality in AD. METHODS: We included 104 patients with late-onset AD and known APOE genotype. All participants underwent a structured interview via informant-based questionnaires to assess sleep quality and the presence of SS. Binary logistic regression analysis was performed to determine odds ratios (ORs) of APOEɛ4 carrier and parameters of sleep quality for SS. RESULTS: The prevalence of SS in AD was 27.8% (n = 29). Patients with SS were significantly more likely to be APOEɛ4 carriers and to have rapid eye movement sleep behavior disorder (RBD) and a higher Clinical Dementia Rating (CDR) score compared to those without SS. In the multivariate regression analysis, APOEɛ4 carrier (OR 3.158, CI 1.022-9.758), RBD (OR 2.166, CI 1.073-4.371), and higher CDR score (OR 2.453, CI 1.084-5.550) were associated with an increased risk of SS. CONCLUSION: The prevalence of SS in patients with AD was 27.8%. The presence of the APOEɛ4 allele, RBD, and more severe dementia are associated with an increased risk of SS in AD.


Subject(s)
Alzheimer Disease/etiology , Apolipoprotein E4/genetics , Chronobiology Disorders/genetics , Circadian Rhythm/genetics , REM Sleep Behavior Disorder/genetics , Aged , Aged, 80 and over , Alzheimer Disease/diagnosis , Alzheimer Disease/epidemiology , Chronobiology Disorders/diagnosis , Chronobiology Disorders/epidemiology , Cross-Sectional Studies , Female , Humans , Male , REM Sleep Behavior Disorder/diagnosis , REM Sleep Behavior Disorder/epidemiology , Risk Factors
13.
J Biol Rhythms ; 34(2): 167-177, 2019 04.
Article in English | MEDLINE | ID: mdl-30712475

ABSTRACT

Circadian misalignment between sleep and behavioral/feeding rhythms is thought to lead to various health impairments in shift workers. Therefore, we investigated how shift work leads to genome-wide circadian dysregulation in hospital nurses. Female nurses from the University of Alabama at Birmingham (UAB) Hospital working night shift ( n = 9; 29.6 ± 11.4 y) and day shift ( n = 8; 34.9 ± 9.4 y) participated in a 9-day study measuring locomotor activity and core body temperature (CBT) continuously. Additionally, cortisol and melatonin were assayed and peripheral blood mononuclear cells (PBMCs) were harvested for RNA extraction every 3 h on a day off from work. We saw phase desynchrony of core body temperature, peak cortisol, and dim light melatonin onset in individual night-shift subjects compared with day-shift subjects. This variability was evident even though day- and night-shift nurses had similar sleep timing and scheduled meal times on days off. Surprisingly, the phase and rhythmicity of the expression of the clock gene, PER1, in PBMCs were similar for day-shift and night-shift subjects. Genome-wide microarray analysis of PBMCs from a subset of nurses revealed distinct gene expression patterns between night-shift and day-shift subjects. Enrichment analysis showed that day-shift subjects expressed pathways involved in generic transcription and regulation of signal transduction, whereas night-shift subjects expressed pathways such as RNA polymerase I promoter opening, the matrisome, and endocytosis. In addition, there was large variability in the number of rhythmic transcripts among subjects, regardless of shift type. Interestingly, the amplitude of the CBT rhythm appeared to be more consistent with the number of cycling transcripts for each of the 6 subjects than was melatonin rhythm. In summary, we show that shift-work patterns affect circadian alignment and gene expression in PBMCs.


Subject(s)
Chronobiology Disorders/etiology , Chronobiology Disorders/genetics , Circadian Rhythm , Nurses/statistics & numerical data , Shift Work Schedule/adverse effects , Transcriptome , Adult , Body Temperature , Female , Gene Expression Regulation , Hospitals , Humans , Hydrocortisone/blood , Leukocytes, Mononuclear , Locomotion , Melatonin/blood , Microarray Analysis , Sleep Disorders, Circadian Rhythm/etiology , Young Adult
14.
Psychiatr Genet ; 29(2): 29-36, 2019 04.
Article in English | MEDLINE | ID: mdl-30516584

ABSTRACT

Circadian rhythm abnormalities have been recognized as a central feature of bipolar disorder (BD) but a coherent biological explanation for them remains lacking. Using genetic mutation of 'clock genes', robust animal models of mania and depression have been developed that elucidate key aspects of circadian rhythms and the circadian clock-mood connection. However, translation of this knowledge into humans remains incomplete. In recent years, very large genome-wide association studies (GWAS) have been conducted and the genetic underpinnings of BD are beginning to emerge. However, these genetic studies in BD do not match well with the evidence from animal studies that implicate the circadian clock in mood regulation. Even larger GWAS have been conducted for circadian phenotypes including chronotype, rhythm amplitude, sleep duration, and insomnia. These studies have identified a diverse set of associated genes, including a minority with previously well-characterized functions in the circadian clock. Taken together, the data from recent GWAS of BD and circadian phenotypes indicate that the genetic organization of the circadian clock, both in health and in BD is complex. The findings from GWAS elucidate potentially novel circadian mechanism that may be partly distinct from those identified in animal models. Pleiotropy, epistasis and nongenetic factors may play important roles in regulating circadian rhythms, some of which may underlie circadian rhythm disturbances in BD.


Subject(s)
Bipolar Disorder/metabolism , Bipolar Disorder/physiopathology , Chronobiology Disorders/physiopathology , Animals , Bipolar Disorder/genetics , Chronobiology Disorders/genetics , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Disease Models, Animal , Genome-Wide Association Study , Genomics , Humans , Lithium/metabolism , Sleep/genetics , Sleep/physiology
15.
Proc Natl Acad Sci U S A ; 115(52): E12388-E12397, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30487216

ABSTRACT

The suprachiasmatic nucleus (SCN) is the principal circadian clock of mammals, coordinating daily rhythms of physiology and behavior. Circadian timing pivots around self-sustaining transcriptional-translational negative feedback loops (TTFLs), whereby CLOCK and BMAL1 drive the expression of the negative regulators Period and Cryptochrome (Cry). Global deletion of Cry1 and Cry2 disables the TTFL, resulting in arrhythmicity in downstream behaviors. We used this highly tractable biology to further develop genetic code expansion (GCE) as a translational switch to achieve reversible control of a biologically relevant protein, Cry1, in the SCN. This employed an orthogonal aminoacyl-tRNA synthetase/tRNACUA pair delivered to the SCN by adeno-associated virus (AAV) vectors, allowing incorporation of a noncanonical amino acid (ncAA) into AAV-encoded Cry1 protein carrying an ectopic amber stop codon. Thus, translational readthrough and Cry1 expression were conditional on the supply of ncAA via culture medium or drinking water and were restricted to neurons by synapsin-dependent expression of aminoacyl tRNA-synthetase. Activation of Cry1 translation by ncAA in neurons of arrhythmic Cry-null SCN slices immediately and dose-dependently initiated TTFL circadian rhythms, which dissipated rapidly after ncAA withdrawal. Moreover, genetic activation of the TTFL in SCN neurons rapidly and reversibly initiated circadian behavior in otherwise arrhythmic Cry-null mice, with rhythm amplitude being determined by the number of transduced SCN neurons. Thus, Cry1 does not specify the development of circadian circuitry and competence but is essential for its labile and rapidly reversible activation. This demonstrates reversible control of mammalian behavior using GCE-based translational switching, a method of potentially broad neurobiological interest.


Subject(s)
Chronobiology Disorders/genetics , Cryptochromes/genetics , Cryptochromes/metabolism , Animals , Chronobiology Disorders/physiopathology , Circadian Clocks/genetics , Circadian Clocks/physiology , Circadian Rhythm/physiology , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Period Circadian Proteins/metabolism , Protein Biosynthesis/physiology , Protein Processing, Post-Translational , Suprachiasmatic Nucleus/metabolism , Transcription Factors/metabolism
16.
Neurobiol Aging ; 72: 159-170, 2018 12.
Article in English | MEDLINE | ID: mdl-30273830

ABSTRACT

Mutations in fused in sarcoma (Fus) cause familial amyotrophic lateral sclerosis (ALS) and occasionally frontotemporal dementia. Here we report the establishment and characterization of a novel knockin (KI) rat model expressing a Fus point mutation (R521C) via CRISPR/Cas9. The mutant animals developed adult-onset learning and memory behavioral deficits, with reduced spine density in hippocampal neurons. Remarkably, sleep-wake cycle and circadian abnormalities preceded the onset of cognitive deficit. RNA-seq study further demonstrated altered expression of some key sleep and circadian regulators, such as orexin/hypocretin receptor type 2 and casein kinase 1 epsilon, in the mutant rats. Therefore, we have established a rodent model expressing physiological level of a pathogenic mutant FUS, and we found cognitive impairment as a main behavioral deficit at mid age. Furthermore, we have revealed a new role of FUS in sleep and circadian regulation and demonstrated that functional change in FUS could cause sleep-wake and circadian disturbance as early symptoms.


Subject(s)
Behavior, Animal , Chronobiology Disorders/genetics , Cognitive Dysfunction/genetics , RNA-Binding Protein FUS/metabolism , Sleep/genetics , Wakefulness/genetics , Age Factors , Animals , Cells, Cultured , Disease Models, Animal , Electroencephalography , Electromyography , Embryo, Mammalian , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Sleep Disorders, Circadian Rhythm/genetics
17.
Article in English | MEDLINE | ID: mdl-30228028

ABSTRACT

Circadian rhythms, endogenous and entrainable adaptations to 24-hour cycles of light and dark, influence almost all physiologic functions. Emerging evidence suggests that the disruption of normal circadian rhythms, termed chronodisruption, could affect a wide range of disease-related processes. In this review, we describe the molecular generation of circadian rhythms, the effects of chronodisruption on human health, the circadian timing of birth in multiple species, the possible effects of chronodisruption on preterm birth, and some of the open questions in this field.


Subject(s)
Chronobiology Disorders/complications , Circadian Rhythm , Premature Birth/etiology , Animals , Chronobiology Disorders/genetics , Female , Humans , Labor Onset , Pregnancy
18.
Horm Behav ; 105: 177-184, 2018 09.
Article in English | MEDLINE | ID: mdl-30031683

ABSTRACT

Circadian disruptions impair reproductive health in human populations and in animal models. We tested the hypothesis that mistimed food, a common disruptive feature of shift work, impairs reproductive success in mice. Male and female mPer2Luc mice on a C57BL/6 background were fed during the light or dark phase in two experiments. Food-induced internal misalignment of the liver clock was verified by in vivo bioluminescence in anesthetized mice in both experiments. In Experiment 1, food-restricted pairs were monitored for litters for 18 weeks. In the light-fed group, birth of the first litter was significantly delayed, and total reproductive output was significantly reduced by 38%. In Experiment 2, estrous cycling was monitored for 3 weeks, and then after pairing, copulatory plugs, pregnancy, litter sizes, and uterine implantation sites were measured. Fewer light-fed females birthed litters (25% versus 73%). This was attributable to a difference in behavior as mating success was significantly reduced in light-fed mice: 42% were observed with a copulatory plug compared to 82% for dark-fed mice. The proportion of mice displaying uterine implantation sites was the same as the proportion observed with copulatory plugs, suggesting no deficit in initiating pregnancy after mating. Estrous cycling and pregnancy maintenance did not differ between the groups. We conclude that mistimed feeding inhibits reproduction in mice by reducing successful mating behavior.


Subject(s)
Chronobiology Disorders/genetics , Feeding Behavior/physiology , Period Circadian Proteins/genetics , Reproduction/physiology , Animal Nutritional Physiological Phenomena/genetics , Animals , Chronobiology Disorders/complications , Chronobiology Disorders/physiopathology , Circadian Rhythm/genetics , Estrous Cycle/genetics , Female , Food , Infertility, Female/etiology , Litter Size/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Reproduction/genetics , Time Factors
19.
Angew Chem Int Ed Engl ; 57(36): 11532-11539, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30003624

ABSTRACT

Living organisms have a biological clock that helps to prepare our physiology for the fluctuations of the day. Key research to elucidate the biological mechanisms of this regular adaptation, referred to as the circadian rhythm, is described by M. W. Young in his Nobel lecture.


Subject(s)
Chronobiology Disorders/etiology , Circadian Rhythm , Animals , Chronobiology Disorders/genetics , Chronobiology Disorders/metabolism , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Drosophila/genetics , Drosophila/physiology , Gene Expression Regulation , Humans , Mutation
20.
Toxicol Sci ; 165(1): 118-130, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29788408

ABSTRACT

Polychlorinated biphenyls (PCBs) and their hydroxylated metabolites (OH-PCBs) have been detected in tissues of both wild animals and humans. Several previous studies have suggested adverse effects of OH-PCBs on the endocrine and nervous systems in mammals. However, there have been no studies on transcriptome analysis of the effects of OH-PCBs, and thus, the whole picture and mechanisms underlying the adverse effects induced by OH-PCBs are still poorly understood. We therefore investigated the mRNA expression profile in the liver of adult male Wistar rats treated with 4-hydroxy-2,3,3',4',5-pentachlorobiphenyl (4-OH-CB107) to explore the genes responsive to OH-PCBs and to understand the potential effects of the chemical. Next-generation RNA sequencing analysis revealed changes in the expression of genes involved in the circadian rhythm and fatty acid metabolism, such as nuclear receptor subfamily 1, group D, member 1, aryl hydrocarbon receptor nuclear translocator-like protein 1, cryptochrome circadian clock 1, and enoyl-CoA hydratase and 3-hydroxyacyl-CoA dehydrogenase, in 4-OH-CB107-treated rats. In addition, biochemical analysis of the plasma revealed a dose-dependent increase in the leucine aminopeptidase, indicating the onset of liver damage. These results suggest that OH-PCB exposure may induce liver injury as well as disrupt the circadian rhythm and peroxisome proliferator-activated receptor-related fatty acid metabolism.


Subject(s)
Chronobiology Disorders/chemically induced , Environmental Pollutants/toxicity , Fatty Acids/metabolism , Liver/drug effects , Polychlorinated Biphenyls/toxicity , Transcriptome/drug effects , Animals , Chronobiology Disorders/genetics , Chronobiology Disorders/metabolism , Dose-Response Relationship, Drug , Gene Expression Profiling , Leucyl Aminopeptidase/blood , Liver/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...