82 results on '"Vitaterna MH"'
Search Results
2. 0018 DATA MINING OF MULTIPLE GENOMICS DATASETS UNCOVERS CONVERGENT GENE NETWORKS INTEGRATING CIRCADIAN TIMING AND HOMEOSTATIC DRIVE FOR SLEEP REGULATION
- Author
-
Jiang, P, primary, Scarpa, JR, additional, Gao, VD, additional, Fitzpatrick, K, additional, Gotter, A, additional, Winrow, CJ, additional, Renger, JJ, additional, Vitaterna, MH, additional, Kasarskis, A, additional, and Turek, FW, additional
- Published
- 2017
- Full Text
- View/download PDF
3. Artificial Gravity Attenuates the Transcriptomic Response to Spaceflight in the Optic Nerve and Retina.
- Author
-
Kremsky I, Pergerson R, Justinen S, Stanbouly S, Willey J, Fuller CA, Takahashi S, Vitaterna MH, Bouxsein M, and Mao X
- Subjects
- Animals, Mice, Oxidative Stress, Mice, Inbred C57BL, Male, Space Flight, Retina metabolism, Gravity, Altered adverse effects, Transcriptome, Weightlessness adverse effects, Optic Nerve metabolism
- Abstract
The development of eye pathology is a serious concern for astronauts who spend time in deep space. Microgravity is a major component of the spaceflight environment which could have adverse effects on ocular health. The use of centrifugation to exert forces that partially or fully mimic Earth-level gravity in space is a possible countermeasure to mitigate the effects of microgravity on the eye. Therefore, we subjected mice on the International Space Station (ISS) to microgravity (0 G) or artificial gravity by centrifugation at 0.33 G, 0.67 G, and 1 G, and then performed RNA sequencing (RNA-seq) on optic nerve and retinal tissue after returning them to Earth alive. We find that the microgravity environment induces transcriptomic changes in the optic nerve and retina consistent with an increased oxidative stress load, inflammation, apoptosis, and lipid metabolic stress. We also find that adding artificial gravity on board the ISS attenuates the transcriptomic response to microgravity in a dose-dependent manner. Such attenuation may effectively protect from and mitigate spaceflight-induced detrimental effects on ocular tissue.
- Published
- 2024
- Full Text
- View/download PDF
4. The impacts of sex and the 5xFAD model of Alzheimer's disease on the sleep and spatial learning responses to feeding time.
- Author
-
Campbell KJ, Jiang P, Olker C, Lin X, Kim SY, Lee CJ, Song EJ, Turek FW, and Vitaterna MH
- Abstract
Introduction: The relationships between the feeding rhythm, sleep and cognition in Alzheimer's disease (AD) are incompletely understood, but meal time could provide an easy-to-implement method of curtailing disease-associated disruptions in sleep and cognition. Furthermore, known sex differences in AD incidence could relate to sex differences in circadian rhythm/sleep/cognition interactions., Methods: The 5xFAD transgenic mouse model of AD and non-transgenic wild-type controls were studied. Both female and male mice were used. Food access was restricted each day to either the 12-h light phase (light-fed groups) or the 12-h dark phase (dark-fed groups). Sleep (electroencephalographic/electromyographic) recording and cognitive behavior measures were collected., Results: The 5xFAD genotype reduces NREM and REM as well as the number of sleep spindles. In wild-type mice, light-fed groups had disrupted vigilance state amounts, characteristics, and rhythms relative to dark-fed groups. These feeding time differences were reduced in 5xFAD mice. Sex modulates these effects. 5xFAD mice display poorer spatial memory that, in female mice, is curtailed by dark phase feeding. Similarly, female 5xFAD mice have decreased anxiety-associated behavior. These emotional and cognitive measures are correlated with REM amount., Discussion: Our study demonstrates that the timing of feeding can alter many aspects of wake, NREM and REM. Unexpectedly, 5xFAD mice are less sensitive to these feeding time effects. 5xFAD mice demonstrate deficits in cognition which are correlated with REM, suggesting that this circadian-timed aspect of sleep may link feeding time and cognition. Sex plays an important role in regulating the impact of feeding time on sleep and cognition in both wild-type and 5xFAD mice, with females showing a greater cognitive response to feeding time than males., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Campbell, Jiang, Olker, Lin, Kim, Lee, Song, Turek and Vitaterna.)
- Published
- 2024
- Full Text
- View/download PDF
5. Author Correction: Disrupted sleep-wake regulation in the MCI-Park mouse model of Parkinson's disease.
- Author
-
Summa KC, Jiang P, González-Rodríguez P, Huang X, Lin X, Vitaterna MH, Dan Y, Surmeier DJ, and Turek FW
- Published
- 2024
- Full Text
- View/download PDF
6. A Prebiotic Diet Containing Galactooligosaccharides and Polydextrose Produces Dynamic and Reproducible Changes in the Gut Microbial Ecosystem in Male Rats.
- Author
-
Thompson RS, Bowers SJ, Vargas F, Hopkins S, Kelley T, Gonzalez A, Lowry CA, Dorrestein PC, Vitaterna MH, Turek FW, Knight R, Wright KP Jr, and Fleshner M
- Subjects
- Animals, Male, Rats, Bile Acids and Salts metabolism, Feces microbiology, Bacteria classification, Bacteria metabolism, RNA, Ribosomal, 16S, Diet methods, Prebiotics, Gastrointestinal Microbiome drug effects, Rats, Sprague-Dawley, Oligosaccharides pharmacology, Oligosaccharides administration & dosage, Glucans
- Abstract
Despite substantial evidence supporting the efficacy of prebiotics for promoting host health and stress resilience, few experiments present evidence documenting the dynamic changes in microbial ecology and fecal microbially modified metabolites over time. Furthermore, the literature reports a lack of reproducible effects of prebiotics on specific bacteria and bacterial-modified metabolites. The current experiments examined whether consumption of diets enriched in prebiotics (galactooligosaccharides (GOS) and polydextrose (PDX)), compared to a control diet, would consistently impact the gut microbiome and microbially modified bile acids over time and between two research sites. Male Sprague Dawley rats were fed control or prebiotic diets for several weeks, and their gut microbiomes and metabolomes were examined using 16S rRNA gene sequencing and untargeted LC-MS/MS analysis. Dietary prebiotics altered the beta diversity, relative abundance of bacterial genera, and microbially modified bile acids over time. PICRUSt2 analyses identified four inferred functional metabolic pathways modified by the prebiotic diet. Correlational network analyses between inferred metabolic pathways and microbially modified bile acids revealed deoxycholic acid as a potential network hub. All these reported effects were consistent between the two research sites, supporting the conclusion that dietary prebiotics robustly changed the gut microbial ecosystem. Consistent with our previous work demonstrating that GOS/PDX reduces the negative impacts of stressor exposure, we propose that ingesting a diet enriched in prebiotics facilitates the development of a health-promoting gut microbial ecosystem.
- Published
- 2024
- Full Text
- View/download PDF
7. Disrupted sleep-wake regulation in the MCI-Park mouse model of Parkinson's disease.
- Author
-
Summa KC, Jiang P, González-Rodríguez P, Huang X, Lin X, Vitaterna MH, Dan Y, Surmeier DJ, and Turek FW
- Abstract
Disrupted sleep has a profound adverse impact on lives of Parkinson's disease (PD) patients and their caregivers. Sleep disturbances are exceedingly common in PD, with substantial heterogeneity in type, timing, and severity. Among the most common sleep-related symptoms reported by PD patients are insomnia, excessive daytime sleepiness, and sleep fragmentation, characterized by interruptions and decreased continuity of sleep. Alterations in brain wave activity, as measured on the electroencephalogram (EEG), also occur in PD, with changes in the pattern and relative contributions of different frequency bands of the EEG spectrum to overall EEG activity in different vigilance states consistently observed. The mechanisms underlying these PD-associated sleep-wake abnormalities are poorly understood, and they are ineffectively treated by conventional PD therapies. To help fill this gap in knowledge, a new progressive model of PD - the MCI-Park mouse - was studied. Near the transition to the parkinsonian state, these mice exhibited significantly altered sleep-wake regulation, including increased wakefulness, decreased non-rapid eye movement (NREM) sleep, increased sleep fragmentation, reduced rapid eye movement (REM) sleep, and altered EEG activity patterns. These sleep-wake abnormalities resemble those identified in PD patients. Thus, this model may help elucidate the circuit mechanisms underlying sleep disruption in PD and identify targets for novel therapeutic approaches., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
8. A Prebiotic Diet Alters the Fecal Microbiome and Improves Sleep in Response to Sleep Disruption in Rats.
- Author
-
Bowers SJ, Summa KC, Thompson RS, González A, Vargas F, Olker C, Jiang P, Lowry CA, Dorrestein PC, Knight R, Wright KP Jr, Fleshner M, Turek FW, and Vitaterna MH
- Abstract
Sleep disruption is a challenging and exceedingly common physiological state that contributes to a wide range of biochemical and molecular perturbations and has been linked to numerous adverse health outcomes. Modern society exerts significant pressure on the sleep/wake cycle via myriad factors, including exposure to electric light, psychological stressors, technological interconnection, jet travel, shift work, and widespread use of sleep-affecting compounds. Interestingly, recent research has identified a link between the microbiome and the regulation of sleep, suggesting that interventions targeting the microbiome may offer unique therapeutic approaches to challenges posed by sleep disruption. In this study, we test the hypothesis that administration of a prebiotic diet containing galactooligosaccharides (GOS) and polydextrose (PDX) in adult male rats improves sleep in response to repeated sleep disruption and during recovery sleep. We found that animals fed the GOS/PDX prebiotic diet for 4 weeks exhibit increased non-rapid eye movement (NREM) and rapid eye movement (REM) sleep during 5 days of sleep disruption and increased total sleep time during 24 h of recovery from sleep disruption compared to animals fed a control diet, despite similar baseline sleep characteristics. Further, the GOS/PDX prebiotic diet led to significant changes in the fecal microbiome. Consistent with previous reports, the prebiotic diet increased the relative abundance of the species Parabacteroides distasonis , which positively correlated with sleep parameters during recovery sleep. Taken together, these findings suggest that the GOS/PDX prebiotic diet may offer an approach to improve resilience to the physiologic challenge of sleep disruption, in part through impacts on the microbiome., Competing Interests: KW reports research support/donated materials from DuPont Nutrition & Biosciences; Grain Processing Corporation; and Friesland Campina Innovation Centre. Financial relationships: consulting with or without receiving fees and/or serving on the advisory boards for Circadian Therapeutics, Ltd., Circadian Biotherapies, Inc., Philips Respironics, and the United States Army Medical Research and Materiel Command – Walter Reed Army Institute of Research. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Bowers, Summa, Thompson, González, Vargas, Olker, Jiang, Lowry, Dorrestein, Knight, Wright, Fleshner, Turek and Vitaterna.)
- Published
- 2022
- Full Text
- View/download PDF
9. Casein kinase 1 epsilon and circadian misalignment impact affective behaviours in mice.
- Author
-
Zhou L, Fitzpatrick K, Olker C, Vitaterna MH, and Turek FW
- Subjects
- Animals, Circadian Rhythm genetics, Female, Male, Mice, Motor Activity, Photoperiod, Casein Kinase 1 epsilon genetics, Circadian Clocks
- Abstract
Affective behaviours and mental health are profoundly affected by disturbances in circadian rhythms. Casein kinase 1 epsilon (CSNK1E) is a core component of the circadian clock. Mice with tau or null mutation of this gene have shortened and lengthened circadian period respectively. Here, we examined anxiety-like, fear, and despair behaviours in both male and female mice of these two different mutants. Compared with wild-type mice, we found reductions in fear and anxiety-like behaviours in both mutant lines and in both sexes, with the tau mutants exhibiting the greatest phenotypic changes. However, the behavioural despair had distinct phenotypic patterns, with markedly less behavioural despair in female null mutants, but not in tau mutants of either sex. To determine whether abnormal light entrainment of tau mutants to 24-h light-dark cycles contributes to these phenotypic differences, we also examined these behaviours in tau mutants on a 20-h light-dark cycle close to their endogenous circadian period. The normalized entrainment restored more wild-type-like behaviours for fear and anxiety, but it induced behavioural despair in tau mutant females. These data show that both mutations of Csnk1e broadly affect fear and anxiety-like behaviours, while the effects on behavioural despair vary with genetics, photoperiod, and sex, suggesting that the mechanisms by which Csnk1e affects fear and anxiety-like behaviours may be similar, but distinct from those affecting behavioural despair. Our study also provides experimental evidence in support of the hypothesis of beneficial outcomes from properly entrained circadian rhythms in terms of the anxiety-like and fear behaviours., (© 2021 Federation of European Neuroscience Societies and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
10. Fundamental Biological Features of Spaceflight: Advancing the Field to Enable Deep-Space Exploration.
- Author
-
Afshinnekoo E, Scott RT, MacKay MJ, Pariset E, Cekanaviciute E, Barker R, Gilroy S, Hassane D, Smith SM, Zwart SR, Nelman-Gonzalez M, Crucian BE, Ponomarev SA, Orlov OI, Shiba D, Muratani M, Yamamoto M, Richards SE, Vaishampayan PA, Meydan C, Foox J, Myrrhe J, Istasse E, Singh N, Venkateswaran K, Keune JA, Ray HE, Basner M, Miller J, Vitaterna MH, Taylor DM, Wallace D, Rubins K, Bailey SM, Grabham P, Costes SV, Mason CE, and Beheshti A
- Published
- 2021
- Full Text
- View/download PDF
11. Ruminiclostridium 5, Parabacteroides distasonis, and bile acid profile are modulated by prebiotic diet and associate with facilitated sleep/clock realignment after chronic disruption of rhythms.
- Author
-
Thompson RS, Gaffney M, Hopkins S, Kelley T, Gonzalez A, Bowers SJ, Vitaterna MH, Turek FW, Foxx CL, Lowry CA, Vargas F, Dorrestein PC, Wright KP Jr, Knight R, and Fleshner M
- Subjects
- Animals, Bacteroidetes, Chromatography, Liquid, Circadian Rhythm, Diet, Male, RNA, Ribosomal, 16S genetics, Rats, Rats, Sprague-Dawley, Sleep, Tandem Mass Spectrometry, Bile Acids and Salts, Prebiotics
- Abstract
Chronic disruption of rhythms (CDR) impacts sleep and can result in circadian misalignment of physiological systems which, in turn, is associated with increased disease risk. Exposure to repeated or severe stressors also disturbs sleep and diurnal rhythms. Prebiotic nutrients produce favorable changes in gut microbial ecology, the gut metabolome, and reduce several negative impacts of acute severe stressor exposure, including disturbed sleep, core body temperature rhythmicity, and gut microbial dysbiosis. In light of previous compelling evidence that prebiotic diet broadly reduces negative impacts of acute, severe stressors, we hypothesize that prebiotic diet will also effectively mitigate the negative impacts of chronic disruption of circadian rhythms on physiology and sleep/wake behavior. Male, Sprague Dawley rats were fed diets enriched in prebiotic substrates or calorically matched control chow. After 5 weeks on diet, rats were exposed to CDR (12 h light/dark reversal, weekly for 8 weeks) or remained on undisturbed normal light/dark cycles (NLD). Sleep EEG, core body temperature, and locomotor activity were recorded via biotelemetry in freely moving rats. Fecal samples were collected on experimental days -33, 0 (day of onset of CDR), and 42. Taxonomic identification and relative abundances of gut microbes were measured in fecal samples using 16S rRNA gene sequencing and shotgun metagenomics. Fecal primary, bacterially modified secondary, and conjugated bile acids were measured using liquid chromatography with tandem mass spectrometry (LC-MS/MS). Prebiotic diet produced rapid and stable increases in the relative abundances of Parabacteroides distasonis and Ruminiclostridium 5. Shotgun metagenomics analyses confirmed reliable increases in relative abundances of Parabacteroides distasonis and Clostridium leptum, a member of the Ruminiclostridium genus. Prebiotic diet also modified fecal bile acid profiles; and based on correlational and step-wise regression analyses, Parabacteroides distasonis and Ruminiclostridium 5 were positively associated with each other and negatively associated with secondary and conjugated bile acids. Prebiotic diet, but not CDR, impacted beta diversity. Measures of alpha diversity evenness were decreased by CDR and prebiotic diet prevented that effect. Rats exposed to CDR while eating prebiotic, compared to control diet, more quickly realigned NREM sleep and core body temperature (ClockLab) diurnal rhythms to the altered light/dark cycle. Finally, both cholic acid and Ruminiclostridium 5 prior to CDR were associated with time to realign CBT rhythms to the new light/dark cycle after CDR; whereas both Ruminiclostridium 5 and taurocholic acid prior to CDR were associated with NREM sleep recovery after CDR. These results support our hypothesis and suggest that ingestion of prebiotic substrates is an effective strategy to increase the relative abundance of health promoting microbes, alter the fecal bile acid profile, and facilitate the recovery and realignment of sleep and diurnal rhythms after circadian disruption., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
12. Immunization with a heat-killed bacterium, Mycobacterium vaccae NCTC 11659, prevents the development of cortical hyperarousal and a PTSD-like sleep phenotype after sleep disruption and acute stress in mice.
- Author
-
Bowers SJ, Lambert S, He S, Lowry CA, Fleshner M, Wright KP, Turek FW, and Vitaterna MH
- Subjects
- Animals, Arousal, Electroencephalography, Hot Temperature, Immunization, Male, Mice, Mice, Inbred C57BL, Mycobacteriaceae, Phenotype, Sleep, Mycobacterium, Stress Disorders, Post-Traumatic
- Abstract
Study Objectives: Sleep deprivation induces systemic inflammation that may contribute to stress vulnerability and other pathologies. We tested the hypothesis that immunization with heat-killed Mycobacterium vaccae NCTC 11659 (MV), an environmental bacterium with immunoregulatory and anti-inflammatory properties, prevents the negative impacts of 5 days of sleep disruption on stress-induced changes in sleep, behavior, and physiology in mice., Methods: In a 2 × 2 × 2 experimental design, male C57BL/6N mice were given injections of either MV or vehicle on days -17, -10, and -3. On days 1-5, mice were exposed to intermittent sleep disruption, whereby sleep was disrupted for 20 h per day. Immediately following sleep disruption, mice were exposed to 1-h social defeat stress or novel cage (control) conditions. Object location memory (OLM) testing was conducted 24 h after social defeat, and tissues were collected 6 days later to measure inflammatory markers. Sleep was recorded using electroencephalography (EEG) and electromyography (EMG) throughout the experiment., Results: In vehicle-treated mice, only the combination of sleep disruption followed by social defeat (double hit): (1) increased brief arousals and NREM beta (15-30 Hz) EEG power in sleep immediately post-social defeat compared to baseline; (2) induced an increase in the proportion of rapid-eye-movement (REM) sleep and number of state shifts for at least 5 days post-social defeat; and (3) induced hyperlocomotion and lack of habituation in the OLM task. Immunization with MV prevented most of these sleep and behavioral changes., Conclusions: Immunization with MV ameliorates a stress-induced sleep and behavioral phenotype that shares features with human posttraumatic stress disorder., (© Sleep Research Society 2020. Published by Oxford University Press on behalf of the Sleep Research Society. All rights reserved. For permissions, please e-mail journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
13. NREM delta power and AD-relevant tauopathy are associated with shared cortical gene networks.
- Author
-
Scarpa JR, Jiang P, Gao VD, Vitaterna MH, Turek FW, and Kasarskis A
- Subjects
- Animals, Cohort Studies, Humans, Mice, Mice, Inbred C57BL, Alzheimer Disease pathology, Cerebellar Cortex metabolism, Gene Regulatory Networks, Sleep Wake Disorders pathology
- Abstract
Reduced NREM sleep in humans is associated with AD neuropathology. Recent work has demonstrated a reduction in NREM sleep in preclinical AD, pointing to its potential utility as an early marker of dementia. We test the hypothesis that reduced NREM delta power and increased tauopathy are associated with shared underlying cortical molecular networks in preclinical AD. We integrate multi-omics data from two extensive public resources, a human Alzheimer's disease cohort from the Mount Sinai Brain Bank (N = 125) reflecting AD progression and a (C57BL/6J × 129S1/SvImJ) F2 mouse population in which NREM delta power was measured (N = 98). Two cortical gene networks, including a CLOCK-dependent circadian network, are associated with NREM delta power and AD tauopathy progression. These networks were validated in independent mouse and human cohorts. Identifying gene networks related to preclinical AD elucidate possible mechanisms associated with the early disease phase and potential targets to alter the disease course.
- Published
- 2021
- Full Text
- View/download PDF
14. Effects of Immunization With the Soil-Derived Bacterium Mycobacterium vaccae on Stress Coping Behaviors and Cognitive Performance in a "Two Hit" Stressor Model.
- Author
-
Foxx CL, Heinze JD, González A, Vargas F, Baratta MV, Elsayed AI, Stewart JR, Loupy KM, Arnold MR, Flux MC, Sago SA, Siebler PH, Milton LN, Lieb MW, Hassell JE, Smith DG, Lee KAK, Appiah SA, Schaefer EJ, Panitchpakdi M, Sikora NC, Weldon KC, Stamper CE, Schmidt D, Duggan DA, Mengesha YM, Ogbaselassie M, Nguyen KT, Gates CA, Schnabel K, Tran L, Jones JD, Vitaterna MH, Turek FW, Fleshner M, Dorrestein PC, Knight R, Wright KP, and Lowry CA
- Abstract
Previous studies demonstrate that Mycobacterium vaccae NCTC 11659 ( M. vaccae ), a soil-derived bacterium with anti-inflammatory and immunoregulatory properties, is a potentially useful countermeasure against negative outcomes to stressors. Here we used male C57BL/6NCrl mice to determine if repeated immunization with M. vaccae is an effective countermeasure in a "two hit" stress exposure model of chronic disruption of rhythms (CDR) followed by acute social defeat (SD). On day -28, mice received implants of biotelemetric recording devices to monitor 24-h rhythms of locomotor activity. Mice were subsequently treated with a heat-killed preparation of M. vaccae (0.1 mg, administered subcutaneously on days -21, -14, -7, and 27) or borate-buffered saline vehicle. Mice were then exposed to 8 consecutive weeks of either stable normal 12:12 h light:dark (LD) conditions or CDR, consisting of 12-h reversals of the LD cycle every 7 days (days 0-56). Finally, mice were exposed to either a 10-min SD or a home cage control condition on day 54. All mice were exposed to object location memory testing 24 h following SD. The gut microbiome and metabolome were assessed in fecal samples collected on days -1, 48, and 62 using 16S rRNA gene sequence and LC-MS/MS spectral data, respectively; the plasma metabolome was additionally measured on day 64. Among mice exposed to normal LD conditions, immunization with M. vaccae induced a shift toward a more proactive behavioral coping response to SD as measured by increases in scouting and avoiding an approaching male CD-1 aggressor, and decreases in submissive upright defensive postures. In the object location memory test, exposure to SD increased cognitive function in CDR mice previously immunized with M. vaccae . Immunization with M. vaccae stabilized the gut microbiome, attenuating CDR-induced reductions in alpha diversity and decreasing within-group measures of beta diversity. Immunization with M. vaccae also increased the relative abundance of 1-heptadecanoyl-sn-glycero-3-phosphocholine, a lysophospholipid, in plasma. Together, these data support the hypothesis that immunization with M. vaccae stabilizes the gut microbiome, induces a shift toward a more proactive response to stress exposure, and promotes stress resilience., Competing Interests: CL serves on the Scientific Advisory Board of Immodulon Therapeutics, Ltd., is cofounder and Chief Scientific Officer of Mycobacteria Therapeutics Corporation, serves as an unpaid scientific consultant with Aurum Switzerland AG and serves at a member of the faculty of the Integrative Psychiatry Institute, Boulder, Colorado, United States. KPW has received research support from the National Institutes of Health, the Pac-12 Conference, and SomaLogic, Inc. outside of this work; consulting fees from or served as a paid member of scientific advisory boards for the Sleep Disorders Research Advisory Board – National Heart, Lung and Blood Institute and CurAegis Technologies, Circadian Therapeutics, Ltd.; and has received speaker/educational/travel consultant honorarium fees from the American Academy of Sleep Medicine, American College of Chest Physicians, American College of Sports Medicine, American Diabetes Association, Associated Professional Sleep Societies, Kellogg Company, and The European Association for the Study of Obesity. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Foxx, Heinze, González, Vargas, Baratta, Elsayed, Stewart, Loupy, Arnold, Flux, Sago, Siebler, Milton, Lieb, Hassell, Smith, Lee, Appiah, Schaefer, Panitchpakdi, Sikora, Weldon, Stamper, Schmidt, Duggan, Mengesha, Ogbaselassie, Nguyen, Gates, Schnabel, Tran, Jones, Vitaterna, Turek, Fleshner, Dorrestein, Knight, Wright and Lowry.)
- Published
- 2021
- Full Text
- View/download PDF
15. Fundamental Biological Features of Spaceflight: Advancing the Field to Enable Deep-Space Exploration.
- Author
-
Afshinnekoo E, Scott RT, MacKay MJ, Pariset E, Cekanaviciute E, Barker R, Gilroy S, Hassane D, Smith SM, Zwart SR, Nelman-Gonzalez M, Crucian BE, Ponomarev SA, Orlov OI, Shiba D, Muratani M, Yamamoto M, Richards SE, Vaishampayan PA, Meydan C, Foox J, Myrrhe J, Istasse E, Singh N, Venkateswaran K, Keune JA, Ray HE, Basner M, Miller J, Vitaterna MH, Taylor DM, Wallace D, Rubins K, Bailey SM, Grabham P, Costes SV, Mason CE, and Beheshti A
- Subjects
- Astronauts, Health, Humans, Microbiota, Risk Factors, Extraterrestrial Environment, Space Flight
- Abstract
Research on astronaut health and model organisms have revealed six features of spaceflight biology that guide our current understanding of fundamental molecular changes that occur during space travel. The features include oxidative stress, DNA damage, mitochondrial dysregulation, epigenetic changes (including gene regulation), telomere length alterations, and microbiome shifts. Here we review the known hazards of human spaceflight, how spaceflight affects living systems through these six fundamental features, and the associated health risks of space exploration. We also discuss the essential issues related to the health and safety of astronauts involved in future missions, especially planned long-duration and Martian missions., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
16. Resource competition shapes biological rhythms and promotes temporal niche differentiation in a community simulation.
- Author
-
Gao VD, Morley-Fletcher S, Maccari S, Vitaterna MH, and Turek FW
- Abstract
Competition for resources often contributes strongly to defining an organism's ecological niche. Endogenous biological rhythms are important adaptations to the temporal dimension of niches, but how other organisms influence such temporal niches has not been much studied, and the role of competition in particular has been even less examined. We investigated how interspecific competition and intraspecific competition for resources shape an organism's activity rhythms.To do this, we simulated communities of one or two species in an agent-based model. Individuals in the simulation move according to a circadian activity rhythm and compete for limited resources. Probability of reproduction is proportional to an individual's success in obtaining resources. Offspring may have variance in rhythm parameters, which allow for the population to evolve over time.We demonstrate that when organisms are arrhythmic, one species will always be competitively excluded from the environment, but the existence of activity rhythms allows niche differentiation and indefinite coexistence of the two species. Two species which are initially active at the same phase will differentiate their phase angle of entrainment over time to avoid each other. When only one species is present in an environment, competition within the species strongly selects for niche expansion through arrhythmicity, but the addition of an interspecific competitor facilitates evolution of increased rhythmic amplitude when combined with additional adaptations for temporal specialization. Finally, if individuals preferentially mate with others who are active at similar times of day, then disruptive selection by intraspecific competition can split one population into two reproductively isolated groups separated in activity time.These simulations suggest that biological rhythms are an effective method to temporally differentiate ecological niches and that competition is an important ecological pressure promoting the evolution of rhythms and sleep. This is the first study to use ecological modeling to examine biological rhythms., Competing Interests: The authors report no conflicts of interest., (© 2020 The Authors. Ecology and Evolution published by John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
17. Repeated sleep disruption in mice leads to persistent shifts in the fecal microbiome and metabolome.
- Author
-
Bowers SJ, Vargas F, González A, He S, Jiang P, Dorrestein PC, Knight R, Wright KP Jr, Lowry CA, Fleshner M, Vitaterna MH, and Turek FW
- Subjects
- Animals, Male, Mice, Bacteria classification, Bacteria genetics, Bacteria growth & development, Feces microbiology, Gastrointestinal Microbiome, RNA, Bacterial genetics, RNA, Ribosomal, 16S genetics, Sleep Deprivation microbiology
- Abstract
It has been established in recent years that the gut microbiome plays a role in health and disease, potentially via alterations in metabolites that influence host physiology. Although sleep disruption and gut dysbiosis have been associated with many of the same diseases, studies investigating the gut microbiome in the context of sleep disruption have yielded inconsistent results, and have not assessed the fecal metabolome. We exposed mice to five days of sleep disruption followed by four days of ad libitum recovery sleep, and assessed the fecal microbiome and fecal metabolome at multiple timepoints using 16S rRNA gene amplicons and untargeted LC-MS/MS mass spectrometry. We found global shifts in both the microbiome and metabolome in the sleep-disrupted group on the second day of recovery sleep, when most sleep parameters had recovered to baseline levels. We observed an increase in the Firmicutes:Bacteroidetes ratio, along with decreases in the genus Lactobacillus, phylum Actinobacteria, and genus Bifidobacterium in sleep-disrupted mice compared to control mice. The latter two taxa remained low at the fourth day post-sleep disruption. We also identified multiple classes of fecal metabolites that were differentially abundant in sleep-disrupted mice, some of which are physiologically relevant and commonly influenced by the microbiome. This included bile acids, and inference of microbial functional gene content suggested reduced levels of the microbial bile salt hydrolase gene in sleep-disrupted mice. Overall, this study adds to the evidence base linking disrupted sleep to the gut microbiome and expands it to the fecal metabolome, identifying sleep disruption-sensitive bacterial taxa and classes of metabolites that may serve as therapeutic targets to improve health after poor sleep., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
18. NMDAR activation regulates the daily rhythms of sleep and mood.
- Author
-
Burgdorf JS, Vitaterna MH, Olker CJ, Song EJ, Christian EP, Sørensen L, Turek FW, Madsen TM, Khan MA, Kroes RA, and Moskal JR
- Subjects
- Affect drug effects, Animals, Circadian Rhythm drug effects, Electroencephalography methods, Male, Rats, Rats, Sprague-Dawley, Receptors, N-Methyl-D-Aspartate agonists, Sleep drug effects, Sleep Deprivation drug therapy, Spiro Compounds therapeutic use, Wakefulness drug effects, Wakefulness physiology, Affect physiology, Circadian Rhythm physiology, Receptors, N-Methyl-D-Aspartate physiology, Sleep physiology, Sleep Deprivation physiopathology, Spiro Compounds pharmacology
- Abstract
Study Objectives: The present studies examine the effects of NMDAR activation by NYX-2925 diurnal rhythmicity of both sleep and wake as well as emotion., Methods: Twenty-four-hour sleep EEG recordings were obtained in sleep-deprived and non-sleep-deprived rats. In addition, the day-night cycle of both activity and mood was measured using home cage ultrasonic-vocalization recordings., Results: NYX-2925 significantly facilitated non-REM (NREM) sleep during the lights-on (sleep) period, and this effect persisted for 3 days following a single dose in sleep-deprived rats. Sleep-bout duration and REM latencies were increased without affecting total REM sleep, suggesting better sleep quality. In addition, delta power during wake was decreased, suggesting less drowsiness. NYX-2925 also rescued learning and memory deficits induced by sleep deprivation, measured using an NMDAR-dependent learning task. Additionally, NYX-2925 increased positive affect and decreased negative affect, primarily by facilitating the transitions from sleep to rough-and-tumble play and back to sleep. In contrast to NYX-2925, the NMDAR antagonist ketamine acutely (1-4 hours post-dosing) suppressed REM and non-REM sleep, increased delta power during wake, and blunted the amplitude of the sleep-wake activity rhythm., Discussion: These data suggest that NYX-2925 could enhance behavioral plasticity via improved sleep quality as well as vigilance during wake. As such, the facilitation of sleep by NYX-2925 has the potential to both reduce symptom burden on neurological and psychiatric disorders as well as serve as a biomarker for drug effects through restoration of sleep architecture., (© Sleep Research Society 2019. Published by Oxford University Press [on behalf of the Sleep Research Society]. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
19. Reproducible changes in the gut microbiome suggest a shift in microbial and host metabolism during spaceflight.
- Author
-
Jiang P, Green SJ, Chlipala GE, Turek FW, and Vitaterna MH
- Subjects
- Animals, Bacteria genetics, Female, Mice, Mice, Inbred C57BL, RNA, Ribosomal, 16S genetics, Bacteria isolation & purification, Feces microbiology, Gastrointestinal Microbiome genetics, Space Flight
- Abstract
Background: Space environment imposes a range of challenges to mammalian physiology and the gut microbiota, and interactions between the two are thought to be important in mammalian health in space. While previous findings have demonstrated a change in the gut microbial community structure during spaceflight, specific environmental factors that alter the gut microbiome and the functional relevance of the microbiome changes during spaceflight remain elusive., Methods: We profiled the microbiome using 16S rRNA gene amplicon sequencing in fecal samples collected from mice after a 37-day spaceflight onboard the International Space Station. We developed an analytical tool, named STARMAPs (Similarity Test for Accordant and Reproducible Microbiome Abundance Patterns), to compare microbiome changes reported here to other relevant datasets. We also integrated the gut microbiome data with the publically available transcriptomic data in the liver of the same animals for a systems-level analysis., Results: We report an elevated microbiome alpha diversity and an altered microbial community structure that were associated with spaceflight environment. Using STARMAPs, we found the observed microbiome changes shared similarity with data reported in mice flown in a previous space shuttle mission, suggesting reproducibility of the effects of spaceflight on the gut microbiome. However, such changes were not comparable with those induced by space-type radiation in Earth-based studies. We found spaceflight led to significantly altered taxon abundance in one order, one family, five genera, and six species of microbes. This was accompanied by a change in the inferred microbial gene abundance that suggests an altered capacity in energy metabolism. Finally, we identified host genes whose expression in the liver were concordantly altered with the inferred gut microbial gene content, particularly highlighting a relationship between host genes involved in protein metabolism and microbial genes involved in putrescine degradation., Conclusions: These observations shed light on the specific environmental factors that contributed to a robust effect on the gut microbiome during spaceflight with important implications for mammalian metabolism. Our findings represent a key step toward a better understanding the role of the gut microbiome in mammalian health during spaceflight and provide a basis for future efforts to develop microbiota-based countermeasures that mitigate risks to crew health during long-term human space expeditions.
- Published
- 2019
- Full Text
- View/download PDF
20. Genetics of Circadian Rhythms.
- Author
-
Vitaterna MH, Shimomura K, and Jiang P
- Subjects
- Animals, Humans, Circadian Clocks genetics, Circadian Rhythm genetics
- Abstract
In mammals, genetic influences of circadian rhythms occur at many levels. A set of core "clock genes" have been identified that form a feedback loop of gene transcription and translation. The core genetic clockwork generates circadian rhythms in cells throughout the body. Polymorphisms in both core clock genes and interacting genes contribute to individual differences in the expression and properties of circadian rhythms. The circadian clock profoundly influences the patterns of gene expression and cellular functions, providing a mechanistic basis for the impact of the genetic circadian system on normal physiological processes as well as the development of diseases., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
21. Trait-like vulnerability of higher-order cognition and ability to maintain wakefulness during combined sleep restriction and circadian misalignment.
- Author
-
Sprecher KE, Ritchie HK, Burke TM, Depner CM, Smits AN, Dorrestein PC, Fleshner M, Knight R, Lowry CA, Turek FW, Vitaterna MH, and Wright KP
- Subjects
- Adult, Attention physiology, Executive Function physiology, Female, Humans, Individuality, Male, Polysomnography, Sleep physiology, Task Performance and Analysis, Wakefulness physiology, Circadian Rhythm physiology, Cognition physiology, Psychomotor Performance physiology, Sleep Deprivation physiopathology, Sleep Disorders, Circadian Rhythm physiopathology
- Abstract
Study Objectives: Determine stability of individual differences in executive function, cognitive processing speed, selective visual attention, and maintenance of wakefulness during simulated sustained operations with combined sleep restriction and circadian misalignment., Methods: Twenty healthy adults (eight female), aged 25.7 (±4.2 SD), body mass index (BMI) 22.3 (±2.1) kg/m2 completed an 18-day protocol twice. Participants maintained habitual self-selected 8-hour sleep schedules for 2 weeks at home prior to a 4-day laboratory visit that included one sleep opportunity per day: 8 hours on night 1, 3 hours on night 2, and 3 hours on mornings 3 and 4. After 3 days of unscheduled sleep at home, participants repeated the entire protocol. Stability and task dependency of individual differences in performance were quantified by intra-class correlation coefficients (ICC) and Kendall's Tau, respectively., Results: Performance on Stroop, Visual Search, and the Maintenance of Wakefulness Test were highly consistent within individuals during combined sleep restriction and circadian misalignment. Individual differences were trait-like as indicated by ICCs (0.54-0.96) classified according to standard criteria as moderate to almost perfect. Individual differences on other performance tasks commonly reported in sleep studies showed fair to almost perfect ICCs (0.22-0.94). Kendall's rank correlations showed that individual vulnerability to sleep restriction and circadian misalignment varied by task and by metric within a task., Conclusions: Consistent vulnerability of higher-order cognition and maintenance of wakefulness to combined sleep restriction and circadian misalignment has implications for the development of precision countermeasure strategies for workers performing safety-critical tasks, e.g. military, police, health care workers and emergency responders., (© Sleep Research Society 2019. Published by Oxford University Press on behalf of the Sleep Research Society. All rights reserved. For permissions, please e-mail journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
22. The NASA Twins Study: A multidimensional analysis of a year-long human spaceflight.
- Author
-
Garrett-Bakelman FE, Darshi M, Green SJ, Gur RC, Lin L, Macias BR, McKenna MJ, Meydan C, Mishra T, Nasrini J, Piening BD, Rizzardi LF, Sharma K, Siamwala JH, Taylor L, Vitaterna MH, Afkarian M, Afshinnekoo E, Ahadi S, Ambati A, Arya M, Bezdan D, Callahan CM, Chen S, Choi AMK, Chlipala GE, Contrepois K, Covington M, Crucian BE, De Vivo I, Dinges DF, Ebert DJ, Feinberg JI, Gandara JA, George KA, Goutsias J, Grills GS, Hargens AR, Heer M, Hillary RP, Hoofnagle AN, Hook VYH, Jenkinson G, Jiang P, Keshavarzian A, Laurie SS, Lee-McMullen B, Lumpkins SB, MacKay M, Maienschein-Cline MG, Melnick AM, Moore TM, Nakahira K, Patel HH, Pietrzyk R, Rao V, Saito R, Salins DN, Schilling JM, Sears DD, Sheridan CK, Stenger MB, Tryggvadottir R, Urban AE, Vaisar T, Van Espen B, Zhang J, Ziegler MG, Zwart SR, Charles JB, Kundrot CE, Scott GBI, Bailey SM, Basner M, Feinberg AP, Lee SMC, Mason CE, Mignot E, Rana BK, Smith SM, Snyder MP, and Turek FW
- Subjects
- Adaptive Immunity, Body Weight, Carotid Arteries diagnostic imaging, Carotid Intima-Media Thickness, DNA Damage, DNA Methylation, Gastrointestinal Microbiome, Genomic Instability, Humans, Male, Telomere Homeostasis, Time Factors, United States, United States National Aeronautics and Space Administration, Adaptation, Physiological, Astronauts, Space Flight
- Abstract
To understand the health impact of long-duration spaceflight, one identical twin astronaut was monitored before, during, and after a 1-year mission onboard the International Space Station; his twin served as a genetically matched ground control. Longitudinal assessments identified spaceflight-specific changes, including decreased body mass, telomere elongation, genome instability, carotid artery distension and increased intima-media thickness, altered ocular structure, transcriptional and metabolic changes, DNA methylation changes in immune and oxidative stress-related pathways, gastrointestinal microbiota alterations, and some cognitive decline postflight. Although average telomere length, global gene expression, and microbiome changes returned to near preflight levels within 6 months after return to Earth, increased numbers of short telomeres were observed and expression of some genes was still disrupted. These multiomic, molecular, physiological, and behavioral datasets provide a valuable roadmap of the putative health risks for future human spaceflight., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2019
- Full Text
- View/download PDF
23. Parkinson's Disease is Associated with Dysregulations of a Dopamine-Modulated Gene Network Relevant to Sleep and Affective Neurobehaviors in the Striatum.
- Author
-
Jiang P, Scarpa JR, Gao VD, Vitaterna MH, Kasarskis A, and Turek FW
- Subjects
- Affective Symptoms pathology, Affective Symptoms physiopathology, Animals, CDC2 Protein Kinase metabolism, Circadian Clocks genetics, Corpus Striatum cytology, Corpus Striatum physiopathology, Datasets as Topic, Disease Models, Animal, Dopaminergic Neurons pathology, Gene Expression Profiling, Gene Expression Regulation physiology, Genetic Predisposition to Disease, Humans, Male, Mice, Parkinson Disease pathology, Parkinson Disease physiopathology, Transcription, Genetic, Affective Symptoms genetics, Corpus Striatum pathology, Dopamine deficiency, Gene Regulatory Networks physiology, Parkinson Disease genetics, Sleep genetics
- Abstract
In addition to the characteristic motor symptoms, Parkinson's disease (PD) often involves a constellation of sleep and mood symptoms. However, the mechanisms underlying these comorbidities are largely unknown. We have previously reconstructed gene networks in the striatum of a population of (C57BL/6J x A/J) F2 mice and associated the networks to sleep and affective phenotypes, providing a resource for integrated analyses to investigate perturbed sleep and affective functions at the gene network level. Combining this resource with PD-relevant transcriptomic datasets from humans and mice, we identified four networks that showed elevated gene expression in PD patients, including a circadian clock and mitotic network that was altered similarly in mouse models of PD. We then utilized multiple types of omics data from public databases and linked this gene network to postsynaptic dopamine signaling in the striatum, CDK1-modulated transcriptional regulation, and the genetic susceptibility of PD. These findings suggest that dopamine deficiency, a key aspect of PD pathology, perturbs a circadian/mitotic gene network in striatal neurons. Since the normal functions of this network were relevant to sleep and affective behaviors, these findings implicate that dysregulation of functional gene networks may be involved in the emergence of non-motor symptoms in PD. Our analyses present a framework for integrating multi-omics data from diverse sources in mice and humans to reveal insights into comorbid symptoms of complex diseases.
- Published
- 2019
- Full Text
- View/download PDF
24. Cross-species systems analysis identifies gene networks differentially altered by sleep loss and depression.
- Author
-
Scarpa JR, Jiang P, Gao VD, Fitzpatrick K, Millstein J, Olker C, Gotter A, Winrow CJ, Renger JJ, Kasarskis A, Turek FW, and Vitaterna MH
- Subjects
- Animals, Antidepressive Agents therapeutic use, Brain metabolism, Cerebral Cortex metabolism, Circadian Rhythm genetics, Depressive Disorder, Major drug therapy, Depressive Disorder, Major genetics, Disease Models, Animal, Genotype, Male, Mice, Mice, Inbred C57BL, Phenotype, Quantitative Trait Loci, Sleep Deprivation drug therapy, Sleep Deprivation genetics, Transcriptome, Depressive Disorder, Major pathology, Gene Regulatory Networks, Sleep Deprivation pathology
- Abstract
To understand the transcriptomic organization underlying sleep and affective function, we studied a population of (C57BL/6J × 129S1/SvImJ) F2 mice by measuring 283 affective and sleep phenotypes and profiling gene expression across four brain regions. We identified converging molecular bases for sleep and affective phenotypes at both the single-gene and gene-network levels. Using publicly available transcriptomic datasets collected from sleep-deprived mice and patients with major depressive disorder (MDD), we identified three cortical gene networks altered by the sleep/wake state and depression. The network-level actions of sleep loss and depression were opposite to each other, providing a mechanistic basis for the sleep disruptions commonly observed in depression, as well as the reported acute antidepressant effects of sleep deprivation. We highlight one particular network composed of circadian rhythm regulators and neuronal activity-dependent immediate-early genes. The key upstream driver of this network, Arc , may act as a nexus linking sleep and depression. Our data provide mechanistic insights into the role of sleep in affective function and MDD.
- Published
- 2018
- Full Text
- View/download PDF
25. Light/Dark Shifting Promotes Alcohol-Induced Colon Carcinogenesis: Possible Role of Intestinal Inflammatory Milieu and Microbiota.
- Author
-
Bishehsari F, Saadalla A, Khazaie K, Engen PA, Voigt RM, Shetuni BB, Forsyth C, Shaikh M, Vitaterna MH, Turek F, and Keshavarzian A
- Subjects
- Animals, Colorectal Neoplasms microbiology, Colorectal Neoplasms pathology, Dysbiosis complications, Dysbiosis microbiology, Dysbiosis pathology, Epithelial Cells pathology, Feeding Behavior, Mast Cells pathology, Mice, Alcoholism complications, Carcinogenesis pathology, Colorectal Neoplasms etiology, Inflammation pathology, Intestines microbiology, Intestines pathology, Microbiota, Photoperiod
- Abstract
Background: Colorectal cancer (CRC) is associated with the modern lifestyle. Chronic alcohol consumption-a frequent habit of majority of modern societies-increases the risk of CRC. Our group showed that chronic alcohol consumption increases polyposis in a mouse mode of CRC. Here we assess the effect of circadian disruption-another modern life style habit-in promoting alcohol-associated CRC., Method: TS4Cre × adenomatous polyposis coli (APC)
lox468 mice underwent (a) an alcohol-containing diet while maintained on a normal 12 h light:12 h dark cycle; or (b) an alcohol-containing diet in conjunction with circadian disruption by once-weekly 12 h phase reversals of the light:dark (LD) cycle. Mice were sacrificed after eight weeks of full alcohol and/or LD shift to collect intestine samples. Tumor number, size, and histologic grades were compared between animal groups. Mast cell protease 2 (MCP2) and 6 (MCP6) histology score were analyzed and compared. Stool collected at baseline and after four weeks of experimental manipulations was used for microbiota analysis., Results: The combination of alcohol and LD shifting accelerated intestinal polyposis, with a significant increase in polyp size, and caused advanced neoplasia. Consistent with a pathogenic role of stromal tryptase-positive mast cells in colon carcinogenesis, the ratio of mMCP6 (stromal)/mMCP2 (intraepithelial) mast cells increased upon LD shifting. Baseline microbiota was similar between groups, and experimental manipulations resulted in a significant difference in the microbiota composition between groups., Conclusions: Circadian disruption by Light:dark shifting exacerbates alcohol-induced polyposis and CRC. Effect of circadian disruption could, at least partly, be mediated by promoting a pro-tumorigenic inflammatory milieu via changes in microbiota., Competing Interests: The authors declare no conflict of interest.- Published
- 2016
- Full Text
- View/download PDF
26. Dopamine pathway imbalance in mice lacking Magel2, a Prader-Willi syndrome candidate gene.
- Author
-
Luck C, Vitaterna MH, and Wevrick R
- Subjects
- Animals, Binge-Eating Disorder physiopathology, Brain metabolism, Diet, High-Fat, Dopamine metabolism, Female, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Serotonin metabolism, Antigens, Neoplasm genetics, Disease Models, Animal, Dopamine genetics, Prader-Willi Syndrome genetics, Proteins genetics
- Abstract
The etiology of abnormal eating behaviors, including binge-eating disorder, is poorly understood. The neural circuits modulating the activities of the neurotransmitters dopamine and serotonin are proposed to be dysfunctional in individuals suffering from eating disorders. Prader-Willi syndrome is a neurodevelopmental disorder that causes extreme food seeking and binge-eating behaviors together with reduced satiety. One of the genes implicated in Prader-Willi syndrome, Magel2, is highly expressed in the regions of the brain that control appetite. Our objective was to examine behaviors relevant to feeding and the neural circuits controlling feeding in a mouse model of Prader-Willi syndrome that lacks expression of the Magel2 gene. We performed behavioral tests related to dopaminergic function, measuring cocaine-induced hyperlocomotion, binge eating, and saccharin-induced anhedonia in Magel2-deficient mice. Next, we analyzed dopaminergic neurons in various brain regions and compared these findings between genotypes. Finally, we examined biochemical markers in the brain under standard diet, high-fat diet, and withdrawal from a high-fat diet conditions. We identified abnormal behaviors and biomarkers reflecting dopaminergic dysfunction in mice lacking Magel2. Our results provide a biological framework for clinical studies of dopaminergic function in children with Prader-Willi syndrome, and may also provide insight into binge-eating disorders that occur in the general population. (PsycINFO Database Record, ((c) 2016 APA, all rights reserved).)
- Published
- 2016
- Full Text
- View/download PDF
27. Systems Genetic Analyses Highlight a TGFβ-FOXO3 Dependent Striatal Astrocyte Network Conserved across Species and Associated with Stress, Sleep, and Huntington's Disease.
- Author
-
Scarpa JR, Jiang P, Losic B, Readhead B, Gao VD, Dudley JT, Vitaterna MH, Turek FW, and Kasarskis A
- Subjects
- Animals, Astrocytes pathology, Corpus Striatum metabolism, Corpus Striatum physiopathology, Forkhead Box Protein O3 biosynthesis, Gene Regulatory Networks, Humans, Huntington Disease physiopathology, Mice, Nerve Net metabolism, Nerve Net pathology, Neurogenesis genetics, Signal Transduction, Sleep genetics, Stress, Psychological metabolism, Transcriptome genetics, Transforming Growth Factor beta biosynthesis, Astrocytes metabolism, Forkhead Box Protein O3 genetics, Huntington Disease genetics, Stress, Psychological genetics, Transforming Growth Factor beta genetics
- Abstract
Recent systems-based analyses have demonstrated that sleep and stress traits emerge from shared genetic and transcriptional networks, and clinical work has elucidated the emergence of sleep dysfunction and stress susceptibility as early symptoms of Huntington's disease. Understanding the biological bases of these early non-motor symptoms may reveal therapeutic targets that prevent disease onset or slow disease progression, but the molecular mechanisms underlying this complex clinical presentation remain largely unknown. In the present work, we specifically examine the relationship between these psychiatric traits and Huntington's disease (HD) by identifying striatal transcriptional networks shared by HD, stress, and sleep phenotypes. First, we utilize a systems-based approach to examine a large publicly available human transcriptomic dataset for HD (GSE3790 from GEO) in a novel way. We use weighted gene coexpression network analysis and differential connectivity analyses to identify transcriptional networks dysregulated in HD, and we use an unbiased ranking scheme that leverages both gene- and network-level information to identify a novel astrocyte-specific network as most relevant to HD caudate. We validate this result in an independent HD cohort. Next, we computationally predict FOXO3 as a regulator of this network, and use multiple publicly available in vitro and in vivo experimental datasets to validate that this astrocyte HD network is downstream of a signaling pathway important in adult neurogenesis (TGFβ-FOXO3). We also map this HD-relevant caudate subnetwork to striatal transcriptional networks in a large (n = 100) chronically stressed (B6xA/J)F2 mouse population that has been extensively phenotyped (328 stress- and sleep-related measurements), and we show that this striatal astrocyte network is correlated to sleep and stress traits, many of which are known to be altered in HD cohorts. We identify causal regulators of this network through Bayesian network analysis, and we highlight their relevance to motor, mood, and sleep traits through multiple in silico approaches, including an examination of their protein binding partners. Finally, we show that these causal regulators may be therapeutically viable for HD because their downstream network was partially modulated by deep brain stimulation of the subthalamic nucleus, a medical intervention thought to confer some therapeutic benefit to HD patients. In conclusion, we show that an astrocyte transcriptional network is primarily associated to HD in the caudate and provide evidence for its relationship to molecular mechanisms of neural stem cell homeostasis. Furthermore, we present a unified systems-based framework for identifying gene networks that are associated with complex non-motor traits that manifest in the earliest phases of HD. By analyzing and integrating multiple independent datasets, we identify a point of molecular convergence between sleep, stress, and HD that reflects their phenotypic comorbidity and reveals a molecular pathway involved in HD progression.
- Published
- 2016
- Full Text
- View/download PDF
28. Night workers with circadian misalignment are susceptible to alcohol-induced intestinal hyperpermeability with social drinking.
- Author
-
Swanson GR, Gorenz A, Shaikh M, Desai V, Kaminsky T, Van Den Berg J, Murphy T, Raeisi S, Fogg L, Vitaterna MH, Forsyth C, Turek F, Burgess HJ, and Keshavarzian A
- Subjects
- Adult, Biomarkers blood, Circadian Rhythm Signaling Peptides and Proteins blood, Circadian Rhythm Signaling Peptides and Proteins genetics, Colon metabolism, Colon physiopathology, Gene Expression Regulation, Humans, Inflammation Mediators blood, Intestine, Small metabolism, Intestine, Small physiopathology, Melatonin blood, Middle Aged, Permeability, Sleep Disorders, Circadian Rhythm blood, Sleep Disorders, Circadian Rhythm diagnosis, Sleep Disorders, Circadian Rhythm physiopathology, Time Factors, Young Adult, Alcohol Drinking adverse effects, Circadian Rhythm, Colon drug effects, Intestine, Small drug effects, Personnel Staffing and Scheduling, Sleep, Sleep Disorders, Circadian Rhythm complications, Work Schedule Tolerance
- Abstract
Alcohol-induced intestinal hyperpermeability (AIHP) is a known risk factor for alcoholic liver disease (ALD), but only 20-30% of heavy alcoholics develop AIHP and ALD. The hypothesis of this study is that circadian misalignment would promote AIHP. We studied two groups of healthy subjects on a stable work schedule for 3 mo [day workers (DW) and night workers (NW)]. Subjects underwent two circadian phase assessments with sugar challenge to access intestinal permeability between which they drank 0.5 g/kg alcohol daily for 7 days. Sleep architecture by actigraphy did not differ at baseline or after alcohol between either group. After alcohol, the dim light melatonin onset (DLMO) in the DW group did not change significantly, but in the NW group there was a significant 2-h phase delay. Both the NW and DW groups had no change in small bowel permeability with alcohol, but only in the NW group was there an increase in colonic and whole gut permeability. A lower area under the curve of melatonin inversely correlated with increased colonic permeability. Alcohol also altered peripheral clock gene amplitude of peripheral blood mononuclear cells in CLOCK, BMAL, PER1, CRY1, and CRY2 in both groups, and inflammatory markers lipopolysaccharide-binding protein, LPS, and IL-6 had an elevated mesor at baseline in NW vs. DW and became arrhythmic with alcohol consumption. Together, our data suggest that central circadian misalignment is a previously unappreciated risk factor for AIHP and that night workers may be at increased risk for developing liver injury with alcohol consumption., (Copyright © 2016 the American Physiological Society.)
- Published
- 2016
- Full Text
- View/download PDF
29. The Circadian Clock Mutation Promotes Intestinal Dysbiosis.
- Author
-
Voigt RM, Summa KC, Forsyth CB, Green SJ, Engen P, Naqib A, Vitaterna MH, Turek FW, and Keshavarzian A
- Subjects
- Animals, CLOCK Proteins genetics, CLOCK Proteins physiology, Circadian Clocks physiology, Dysbiosis physiopathology, Ethanol pharmacology, Feces microbiology, Male, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, RNA, Ribosomal, 16S, Circadian Clocks genetics, Dysbiosis genetics, Gastrointestinal Microbiome drug effects, Gastrointestinal Microbiome physiology
- Abstract
Background: Circadian rhythm disruption is a prevalent feature of modern day society that is associated with an increase in pro-inflammatory diseases, and there is a clear need for a better understanding of the mechanism(s) underlying this phenomenon. We have previously demonstrated that both environmental and genetic circadian rhythm disruption causes intestinal hyperpermeability and exacerbates alcohol-induced intestinal hyperpermeability and liver pathology. The intestinal microbiota can influence intestinal barrier integrity and impact immune system function; thus, in this study, we sought to determine whether genetic alteration of the core circadian clock gene, Clock, altered the intestinal microbiota community., Methods: Male Clock(Δ19) -mutant mice (mice homozygous for a dominant-negative-mutant allele) or littermate wild-type mice were fed 1 of 3 experimental diets: (i) a standard chow diet, (ii) an alcohol-containing diet, or (iii) an alcohol-control diet in which the alcohol calories were replaced with dextrose. Stool microbiota was assessed with 16S ribosomal RNA gene amplicon sequencing., Results: The fecal microbial community of Clock-mutant mice had lower taxonomic diversity, relative to wild-type mice, and the Clock(Δ19) mutation was associated with intestinal dysbiosis when mice were fed either the alcohol-containing or the control diet. We found that alcohol consumption significantly altered the intestinal microbiota in both wild-type and Clock-mutant mice., Conclusions: Our data support a model by which circadian rhythm disruption by the Clock(Δ19) mutation perturbs normal intestinal microbial communities, and this trend was exacerbated in the context of a secondary dietary intestinal stressor., (Copyright © 2016 by the Research Society on Alcoholism.)
- Published
- 2016
- Full Text
- View/download PDF
30. Altered Body Weight Regulation in CK1ε Null and tau Mutant Mice on Regular Chow and High Fat Diets.
- Author
-
Zhou L, Summa KC, Olker C, Vitaterna MH, and Turek FW
- Abstract
Disruption of circadian rhythms results in metabolic dysfunction. Casein kinase 1 epsilon (CK1ε) is a canonical circadian clock gene. Null and tau mutations in CK1ε show distinct effects on circadian period. To investigate the role of CK1ε in body weight regulation under both regular chow (RC) and high fat (HF) diet conditions, we examined body weight on both RC and HF diets in CK1ε (-/-) and CK1ε (tau/tau) mice on a standard 24 hr light-dark (LD) cycle. Given the abnormal entrainment of CK1ε (tau/tau) mice on a 24 hr LD cycle, a separate set of CK1ε (tau/tau) mice were tested under both diet conditions on a 20 hr LD cycle, which more closely matches their endogenous period length. On the RC diet, both CK1ε (-/-) and CK1ε (tau/tau) mutants on a 24 hr LD cycle and CK1ε (tau/tau) mice on a 20 hr LD cycle exhibited significantly lower body weights, despite similar overall food intake and activity levels. On the HF diet, CK1ε (tau/tau) mice on a 20 hr LD cycle were protected against the development of HF diet-induced excess weight gain. These results provide additional evidence supporting a link between circadian rhythms and energy regulation at the genetic level, particularly highlighting CK1ε involved in the integration of circadian biology and metabolic physiology.
- Published
- 2016
- Full Text
- View/download PDF
31. Osteoarthritis-like pathologic changes in the knee joint induced by environmental disruption of circadian rhythms is potentiated by a high-fat diet.
- Author
-
Kc R, Li X, Forsyth CB, Voigt RM, Summa KC, Vitaterna MH, Tryniszewska B, Keshavarzian A, Turek FW, Meng QJ, and Im HJ
- Subjects
- Animals, Body Weight, Disease Models, Animal, Disease Progression, Environment, Humans, Knee Joint metabolism, Male, Mice, Inbred C57BL, Obesity etiology, Obesity physiopathology, Osteoarthritis, Knee physiopathology, Proteoglycans metabolism, Circadian Rhythm, Diet, High-Fat adverse effects, Knee Joint pathology, Osteoarthritis, Knee etiology
- Abstract
A variety of environmental factors contribute to progressive development of osteoarthritis (OA). Environmental factors that upset circadian rhythms have been linked to various diseases. Our recent work establishes chronic environmental circadian disruption - analogous to rotating shiftwork-associated disruption of circadian rhythms in humans - as a novel risk factor for the development of OA. Evidence suggests shift workers are prone to obesity and also show altered eating habits (i.e., increased preference for high-fat containing food). In the present study, we investigated the impact of chronic circadian rhythm disruption in combination with a high-fat diet (HFD) on progression of OA in a mouse model. Our study demonstrates that when mice with chronically circadian rhythms were fed a HFD, there was a significant proteoglycan (PG) loss and fibrillation in knee joint as well as increased activation of the expression of the catabolic mediators involved in cartilage homeostasis. Our results, for the first time, provide the evidence that environmental disruption of circadian rhythms plus HFD potentiate OA-like pathological changes in the mouse joints. Thus, our findings may open new perspectives on the interactions of chronic circadian rhythms disruption with diet in the development of OA and may have potential clinical implications.
- Published
- 2015
- Full Text
- View/download PDF
32. Chronic Alcohol Exposure and the Circadian Clock Mutation Exert Tissue-Specific Effects on Gene Expression in Mouse Hippocampus, Liver, and Proximal Colon.
- Author
-
Summa KC, Jiang P, Fitzpatrick K, Voigt RM, Bowers SJ, Forsyth CB, Vitaterna MH, Keshavarzian A, and Turek FW
- Subjects
- Animals, Colon drug effects, Ethanol administration & dosage, Liver drug effects, Male, Mice, Mutation, Organ Specificity drug effects, CLOCK Proteins genetics, Colon metabolism, Ethanol pharmacology, Gene Expression Regulation drug effects, Hippocampus drug effects, Hippocampus metabolism, Liver metabolism
- Abstract
Background: Chronic alcohol exposure exerts numerous adverse effects, although the specific mechanisms underlying these negative effects on different tissues are not completely understood. Alcohol also affects core properties of the circadian clock system, and it has been shown that disruption of circadian rhythms confers vulnerability to alcohol-induced pathology of the gastrointestinal barrier and liver. Despite these findings, little is known of the molecular interactions between alcohol and the circadian clock system, especially regarding implications for tissue-specific susceptibility to alcohol pathologies. The aim of this study was to identify changes in expression of genes relevant to alcohol pathologies and circadian clock function in different tissues in response to chronic alcohol intake., Methods: Wild-type and circadian Clock(Δ19) mutant mice were subjected to a 10-week chronic alcohol protocol, after which hippocampal, liver, and proximal colon tissues were harvested for gene expression analysis using a custom-designed multiplex magnetic bead hybridization assay that provided quantitative assessment of 80 mRNA targets of interest, including 5 housekeeping genes and a predetermined set of 75 genes relevant for alcohol pathology and circadian clock function., Results: Significant alterations in expression levels attributable to genotype, alcohol, and/or a genotype by alcohol interaction were observed in all 3 tissues, with distinct patterns of expression changes observed in each. Of particular interest was the finding that a high proportion of genes involved in inflammation and metabolism on the array was significantly affected by alcohol and the Clock(Δ19) mutation in the hippocampus, suggesting a suite of molecular changes that may contribute to pathological change., Conclusions: These results reveal the tissue-specific nature of gene expression responses to chronic alcohol exposure and the Clock(Δ19) mutation and identify specific expression profiles that may contribute to tissue-specific vulnerability to alcohol-induced injury in the brain, colon, and liver., (Copyright © 2015 by the Research Society on Alcoholism.)
- Published
- 2015
- Full Text
- View/download PDF
33. Environmental disruption of circadian rhythm predisposes mice to osteoarthritis-like changes in knee joint.
- Author
-
Kc R, Li X, Voigt RM, Ellman MB, Summa KC, Vitaterna MH, Keshavarizian A, Turek FW, Meng QJ, Stein GS, van Wijnen AJ, Chen D, Forsyth CB, and Im HJ
- Subjects
- Animals, Cartilage, Articular pathology, Circadian Rhythm physiology, Core Binding Factor Alpha 1 Subunit biosynthesis, Disease Susceptibility, Environment, Homeostasis genetics, Humans, Knee Joint metabolism, Knee Joint physiopathology, MAP Kinase Signaling System genetics, Matrix Metalloproteinase 13 biosynthesis, Mice, Motor Activity genetics, Motor Activity physiology, Osteoarthritis, Knee physiopathology, CLOCK Proteins genetics, Cartilage, Articular metabolism, Circadian Rhythm genetics, Osteoarthritis, Knee genetics
- Abstract
Circadian rhythm dysfunction is linked to many diseases, yet pathophysiological roles in articular cartilage homeostasis and degenerative joint disease including osteoarthritis (OA) remains to be investigated in vivo. Here, we tested whether environmental or genetic disruption of circadian homeostasis predisposes to OA-like pathological changes. Male mice were examined for circadian locomotor activity upon changes in the light:dark (LD) cycle or genetic disruption of circadian rhythms. Wild-type (WT) mice were maintained on a constant 12 h:12 h LD cycle (12:12 LD) or exposed to weekly 12 h phase shifts. Alternatively, male circadian mutant mice (Clock(Δ19) or Csnk1e(tau) mutants) were compared with age-matched WT littermates that were maintained on a constant 12:12 LD cycle. Disruption of circadian rhythms promoted osteoarthritic changes by suppressing proteoglycan accumulation, upregulating matrix-degrading enzymes and downregulating anabolic mediators in the mouse knee joint. Mechanistically, these effects involved activation of the PKCδ-ERK-RUNX2/NFκB and β-catenin signaling pathways, stimulation of MMP-13 and ADAMTS-5, as well as suppression of the anabolic mediators SOX9 and TIMP-3 in articular chondrocytes of phase-shifted mice. Genetic disruption of circadian homeostasis does not predispose to OA-like pathological changes in joints. Our results, for the first time, provide compelling in vivo evidence that environmental disruption of circadian rhythms is a risk factor for the development of OA-like pathological changes in the mouse knee joint., (© 2015 Wiley Periodicals, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
34. A systems approach identifies networks and genes linking sleep and stress: implications for neuropsychiatric disorders.
- Author
-
Jiang P, Scarpa JR, Fitzpatrick K, Losic B, Gao VD, Hao K, Summa KC, Yang HS, Zhang B, Allada R, Vitaterna MH, Turek FW, and Kasarskis A
- Subjects
- Animals, Bayes Theorem, Gene Regulatory Networks, Mental Disorders genetics, Mental Disorders pathology, Mental Disorders veterinary, Mice, Inbred C57BL, Microfilament Proteins genetics, Microfilament Proteins metabolism, Phenotype, Quantitative Trait Loci, Transcriptome, Sleep, Stress, Psychological genetics
- Abstract
Sleep dysfunction and stress susceptibility are comorbid complex traits that often precede and predispose patients to a variety of neuropsychiatric diseases. Here, we demonstrate multilevel organizations of genetic landscape, candidate genes, and molecular networks associated with 328 stress and sleep traits in a chronically stressed population of 338 (C57BL/6J × A/J) F2 mice. We constructed striatal gene co-expression networks, revealing functionally and cell-type-specific gene co-regulations important for stress and sleep. Using a composite ranking system, we identified network modules most relevant for 15 independent phenotypic categories, highlighting a mitochondria/synaptic module that links sleep and stress. The key network regulators of this module are overrepresented with genes implicated in neuropsychiatric diseases. Our work suggests that the interplay among sleep, stress, and neuropathology emerges from genetic influences on gene expression and their collective organization through complex molecular networks, providing a framework for interrogating the mechanisms underlying sleep, stress susceptibility, and related neuropsychiatric disorders., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
35. Validation of video motion-detection scoring of forced swim test in mice.
- Author
-
Gao V, Vitaterna MH, and Turek FW
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Motor Activity, Software, Species Specificity, Time Factors, Depression diagnosis, Motion, Neuropsychological Tests, Pattern Recognition, Automated methods, Swimming, Video Recording methods
- Abstract
Background: The forced swim test (FST) is used to predict the effectiveness of novel antidepressant treatments. In this test, a mouse or rat is placed in a beaker of water for several minutes, and the amount of time spent passively floating is measured; antidepressants reduce the amount of such immobility. Though the FST is commonly used, manually scoring the test is time-consuming and involves considerable subjectivity., New Method: We developed a simple MATLAB-based motion-detection method to quantify mice's activity in videos of FST. FST trials are video-recorded from a side view. Each pixel of the video is compared between subsequent video frames; if the pixel's color difference surpasses a threshold, a motion count is recorded., Results: Human-scored immobility time correlates well with total motion detected by the computer (r=-0.80) and immobility time determined by the computer (r=0.83). Our computer method successfully detects group differences in activity between genotypes and different days of testing. Furthermore, we observe heterosis for this behavior, in which (C57BL/6J×A/J) F1 hybrid mice are more active in the FST than the parental strains., Comparison With Existing Methods: This computer-scoring method is much faster and more objective than human scoring. Other automatic scoring methods exist, but they require the purchase of expensive hardware and/or software., Conclusion: This computer-scoring method is an effective, fast, and low-cost method of quantifying the FST. It is validated by replicating statistical differences observed in traditional visual scoring. We also demonstrate a case of heterosis in the FST., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
36. Circadian disorganization alters intestinal microbiota.
- Author
-
Voigt RM, Forsyth CB, Green SJ, Mutlu E, Engen P, Vitaterna MH, Turek FW, and Keshavarzian A
- Subjects
- Animals, Diet, High-Fat adverse effects, Dietary Sucrose adverse effects, Dysbiosis etiology, Dysbiosis physiopathology, Intestines physiopathology, Light, Male, Mice, Mice, Inbred C57BL, Microbiota physiology, Circadian Rhythm physiology, Dysbiosis microbiology, Intestines microbiology, Photoperiod
- Abstract
Intestinal dysbiosis and circadian rhythm disruption are associated with similar diseases including obesity, metabolic syndrome, and inflammatory bowel disease. Despite the overlap, the potential relationship between circadian disorganization and dysbiosis is unknown; thus, in the present study, a model of chronic circadian disruption was used to determine the impact on the intestinal microbiome. Male C57BL/6J mice underwent once weekly phase reversals of the light:dark cycle (i.e., circadian rhythm disrupted mice) to determine the impact of circadian rhythm disruption on the intestinal microbiome and were fed either standard chow or a high-fat, high-sugar diet to determine how diet influences circadian disruption-induced effects on the microbiome. Weekly phase reversals of the light:dark (LD) cycle did not alter the microbiome in mice fed standard chow; however, mice fed a high-fat, high-sugar diet in conjunction with phase shifts in the light:dark cycle had significantly altered microbiota. While it is yet to be established if some of the adverse effects associated with circadian disorganization in humans (e.g., shift workers, travelers moving across time zones, and in individuals with social jet lag) are mediated by dysbiosis, the current study demonstrates that circadian disorganization can impact the intestinal microbiota which may have implications for inflammatory diseases.
- Published
- 2014
- Full Text
- View/download PDF
37. The circadian clock gene Csnk1e regulates rapid eye movement sleep amount, and nonrapid eye movement sleep architecture in mice.
- Author
-
Zhou L, Bryant CD, Loudon A, Palmer AA, Vitaterna MH, and Turek FW
- Subjects
- Animals, Casein Kinase 1 epsilon metabolism, Chromosomes, Mammalian genetics, Circadian Clocks physiology, Circadian Rhythm genetics, Circadian Rhythm physiology, Darkness, Electroencephalography, Electromyography, Genotype, Light, Male, Mice, Mice, Inbred C57BL, Mice, Inbred DBA, Mutation genetics, Phenotype, Quantitative Trait Loci genetics, Time Factors, Casein Kinase 1 epsilon genetics, Circadian Clocks genetics, Sleep genetics, Sleep physiology, Sleep, REM genetics, Sleep, REM physiology
- Abstract
Study Objectives: Efforts to identify the genetic basis of mammalian sleep have included quantitative trait locus (QTL) mapping and gene targeting of known core circadian clock genes. We combined three different genetic approaches to identify and test a positional candidate sleep gene - the circadian gene casein kinase 1 epsilon (Csnk1e), which is located in a QTL we identified for rapid eye movement (REM) sleep on chromosome 15., Measurements and Results: Using electroencephalographic (EEG) and electromyographic (EMG) recordings, baseline sleep was examined in a 12-h light:12-h dark (LD 12:12) cycle in mice of seven genotypes, including Csnk1e(tau/tau) and Csnk1e(-/-) mutant mice, Csnk1e (B6.D2) and Csnk1e (D2.B6) congenic mice, and their respective wild-type littermate control mice. Additionally, Csnk1e(tau/tau) and wild-type mice were examined in constant darkness (DD). Csnk1e(tau/tau) mutant mice and both Csnk1e (B6.D2) and Csnk1e (D2.B6) congenic mice showed significantly higher proportion of sleep time spent in REM sleep during the dark period than wild-type controls - the original phenotype for which the QTL on chromosome 15 was identified. This phenotype persisted in Csnk1e(tau/tau) mice while under free-running DD conditions. Other sleep phenotypes observed in Csnk1e(tau/tau) mice and congenics included a decreased number of bouts of nonrapid eye movement (NREM) sleep and an increased average NREM sleep bout duration., Conclusions: These results demonstrate a role for Csnk1e in regulating not only the timing of sleep, but also the REM sleep amount and NREM sleep architecture, and support Csnk1e as a causal gene in the sleep QTL on chromosome 15.
- Published
- 2014
- Full Text
- View/download PDF
38. C57BL/6N mutation in cytoplasmic FMRP interacting protein 2 regulates cocaine response.
- Author
-
Kumar V, Kim K, Joseph C, Kourrich S, Yoo SH, Huang HC, Vitaterna MH, de Villena FP, Churchill G, Bonci A, and Takahashi JS
- Subjects
- Adaptor Proteins, Signal Transducing, Amino Acid Substitution, Animals, Central Nervous System Stimulants administration & dosage, Methamphetamine administration & dosage, Mice, Mice, Inbred C57BL, Mice, Knockout, Motor Activity drug effects, Mutation, Nerve Tissue Proteins genetics, Phenylalanine genetics, Polymorphism, Single Nucleotide, Psychomotor Performance drug effects, Quantitative Trait Loci, Serine genetics, Cocaine administration & dosage, Cocaine-Related Disorders genetics, Cocaine-Related Disorders psychology, Drug-Seeking Behavior, Nerve Tissue Proteins physiology
- Abstract
The inbred mouse C57BL/6J is the reference strain for genome sequence and for most behavioral and physiological phenotypes. However, the International Knockout Mouse Consortium uses an embryonic stem cell line derived from a related C57BL/6N substrain. We found that C57BL/6N has a lower acute and sensitized response to cocaine and methamphetamine. We mapped a single causative locus and identified a nonsynonymous mutation of serine to phenylalanine (S968F) in Cytoplasmic FMRP interacting protein 2 (Cyfip2) as the causative variant. The S968F mutation destabilizes CYFIP2, and deletion of the C57BL/6N mutant allele leads to acute and sensitized cocaine-response phenotypes. We propose that CYFIP2 is a key regulator of cocaine response in mammals and present a framework to use mouse substrains to identify previously unknown genes and alleles regulating behavior.
- Published
- 2013
- Full Text
- View/download PDF
39. Disruption of the Circadian Clock in Mice Increases Intestinal Permeability and Promotes Alcohol-Induced Hepatic Pathology and Inflammation.
- Author
-
Summa KC, Voigt RM, Forsyth CB, Shaikh M, Cavanaugh K, Tang Y, Vitaterna MH, Song S, Turek FW, and Keshavarzian A
- Subjects
- Animals, Liver pathology, Mice, Mice, Inbred C57BL, Circadian Clocks, Ethanol pharmacology, Hepatitis, Alcoholic etiology, Liver drug effects
- Abstract
The circadian clock orchestrates temporal patterns of physiology and behavior relative to the environmental light:dark cycle by generating and organizing transcriptional and biochemical rhythms in cells and tissues throughout the body. Circadian clock genes have been shown to regulate the physiology and function of the gastrointestinal tract. Disruption of the intestinal epithelial barrier enables the translocation of proinflammatory bacterial products, such as endotoxin, across the intestinal wall and into systemic circulation; a process that has been linked to pathologic inflammatory states associated with metabolic, hepatic, cardiovascular and neurodegenerative diseases - many of which are commonly reported in shift workers. Here we report, for the first time, that circadian disorganization, using independent genetic and environmental strategies, increases permeability of the intestinal epithelial barrier (i.e., gut leakiness) in mice. Utilizing chronic alcohol consumption as a well-established model of induced intestinal hyperpermeability, we also found that both genetic and environmental circadian disruption promote alcohol-induced gut leakiness, endotoxemia and steatohepatitis, possibly through a mechanism involving the tight junction protein occludin. Circadian organization thus appears critical for the maintenance of intestinal barrier integrity, especially in the context of injurious agents, such as alcohol. Circadian disruption may therefore represent a previously unrecognized risk factor underlying the susceptibility to or development of alcoholic liver disease, as well as other conditions associated with intestinal hyperpermeability and an endotoxin-triggered inflammatory state.
- Published
- 2013
- Full Text
- View/download PDF
40. Usf1, a suppressor of the circadian Clock mutant, reveals the nature of the DNA-binding of the CLOCK:BMAL1 complex in mice.
- Author
-
Shimomura K, Kumar V, Koike N, Kim TK, Chong J, Buhr ED, Whiteley AR, Low SS, Omura C, Fenner D, Owens JR, Richards M, Yoo SH, Hong HK, Vitaterna MH, Bass J, Pletcher MT, Wiltshire T, Hogenesch J, Lowrey PL, and Takahashi JS
- Subjects
- ARNTL Transcription Factors genetics, Animals, Binding Sites, Binding, Competitive, CLOCK Proteins genetics, E-Box Elements, Gene Expression Regulation, Genotype, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Phenotype, Polymorphism, Single Nucleotide, Promoter Regions, Genetic, Protein Interaction Domains and Motifs, RNA, Messenger metabolism, Signal Transduction, Species Specificity, Time Factors, Transcription, Genetic, Transcriptional Activation, Upstream Stimulatory Factors genetics, ARNTL Transcription Factors metabolism, CLOCK Proteins metabolism, Circadian Clocks genetics, Circadian Rhythm genetics, DNA metabolism, Mutation, Upstream Stimulatory Factors metabolism
- Abstract
Genetic and molecular approaches have been critical for elucidating the mechanism of the mammalian circadian clock. Here, we demonstrate that the ClockΔ19 mutant behavioral phenotype is significantly modified by mouse strain genetic background. We map a suppressor of the ClockΔ19 mutation to a ∼900 kb interval on mouse chromosome 1 and identify the transcription factor, Usf1, as the responsible gene. A SNP in the promoter of Usf1 causes elevation of its transcript and protein in strains that suppress the Clock mutant phenotype. USF1 competes with the CLOCK:BMAL1 complex for binding to E-box sites in target genes. Saturation binding experiments demonstrate reduced affinity of the CLOCKΔ19:BMAL1 complex for E-box sites, thereby permitting increased USF1 occupancy on a genome-wide basis. We propose that USF1 is an important modulator of molecular and behavioral circadian rhythms in mammals. DOI:http://dx.doi.org/10.7554/eLife.00426.001.
- Published
- 2013
- Full Text
- View/download PDF
41. Altered sleep and affect in the neurotensin receptor 1 knockout mouse.
- Author
-
Fitzpatrick K, Winrow CJ, Gotter AL, Millstein J, Arbuzova J, Brunner J, Kasarskis A, Vitaterna MH, Renger JJ, and Turek FW
- Subjects
- Animals, Anxiety genetics, Depression genetics, Electroencephalography, Electromyography, Male, Mice, Mice, Inbred C57BL, Mice, Knockout physiology, Motor Activity physiology, Quantitative Trait Loci, Sleep Deprivation physiopathology, Affect physiology, Receptors, Neurotensin physiology, Sleep physiology
- Abstract
Study Objective: Sleep and mood disorders have long been understood to have strong genetic components, and there is considerable comorbidity of sleep abnormalities and mood disorders, suggesting the involvement of common genetic pathways. Here, we examine a candidate gene implicated in the regulation of both sleep and affective behavior using a knockout mouse model., Design: Previously, we identified a quantitative trait locus (QTL) for REM sleep amount, REM sleep bout number, and wake amount in a genetically segregating population of mice. Here, we show that traits mapping to this QTL correlated with an expression QTL for neurotensin receptor 1 (Ntsr1), a receptor for neurotensin, a ligand known to be involved in several psychiatric disorders. We examined sleep as well as behaviors indicative of anxiety and depression in the NTSR1 knockout mouse., Measurements and Results: NTSR1 knockouts had a lower percentage of sleep time spent in REM sleep in the dark phase and a larger diurnal variation in REM sleep duration than wild types under baseline conditions. Following sleep deprivation, NTSR1 knockouts exhibited more wake and less NREM rebound sleep. NTSR1 knockouts also showed increased anxious and despair behaviors., Conclusions: Here we illustrate a link between expression of the Ntsr1 gene and sleep traits previously associated with a particular QTL. We also demonstrate a relationship between Ntsr1 and anxiety and despair behaviors. Given the considerable evidence that anxiety and depression are closely linked with abnormalities in sleep, the data presented here provide further evidence that neurotensin and Ntsr1 may be a component of a pathway involved in both sleep and mood disorders.
- Published
- 2012
- Full Text
- View/download PDF
42. Csnk1e is a genetic regulator of sensitivity to psychostimulants and opioids.
- Author
-
Bryant CD, Parker CC, Zhou L, Olker C, Chandrasekaran RY, Wager TT, Bolivar VJ, Loudon AS, Vitaterna MH, Turek FW, and Palmer AA
- Subjects
- Animals, Casein Kinase 1 epsilon deficiency, Female, Genome-Wide Association Study methods, Male, Mice, Mice, Inbred C57BL, Mice, Inbred DBA, Mice, Inbred Strains, Mice, Knockout, Motor Activity drug effects, Motor Activity genetics, Analgesics, Opioid pharmacology, Casein Kinase 1 epsilon genetics, Central Nervous System Stimulants pharmacology, Drug Resistance genetics, Quantitative Trait Loci genetics
- Abstract
Csnk1e, the gene encoding casein kinase 1-epsilon, has been implicated in sensitivity to amphetamines. Additionally, a polymorphism in CSNK1E was associated with heroin addiction, suggesting that this gene may also affect opioid sensitivity. In this study, we first conducted genome-wide quantitative trait locus (QTL) mapping of methamphetamine (MA)-induced locomotor activity in C57BL/6J (B6) × DBA/2J (D2)-F(2) mice and a more highly recombinant F(8) advanced intercross line. We identified a QTL on chromosome 15 that contained Csnk1e (63-86 Mb; Csnk1e=79.25 Mb). We replicated this result and further narrowed the locus using B6.D2(Csnk1e) and D2.B6(Csnk1e) reciprocal congenic lines (78-86.8 and 78.7-81.6 Mb, respectively). This locus also affected sensitivity to the μ-opioid receptor agonist fentanyl. Next, we directly tested the hypothesis that Csnk1e is a genetic regulator of sensitivity to psychostimulants and opioids. Mice harboring a null allele of Csnk1e showed an increase in locomotor activity following MA administration. Consistent with this result, coadministration of a selective pharmacological inhibitor of Csnk1e (PF-4800567) increased the locomotor stimulant response to both MA and fentanyl. These results show that a narrow genetic locus that contains Csnk1e is associated with differences in sensitivity to MA and fentanyl. Furthermore, gene knockout and selective pharmacological inhibition of Csnk1e define its role as a negative regulator of sensitivity to psychostimulants and opioids.
- Published
- 2012
- Full Text
- View/download PDF
43. High-resolution mapping of a novel genetic locus regulating voluntary physical activity in mice.
- Author
-
Yang HS, Shimomura K, Vitaterna MH, and Turek FW
- Subjects
- Animals, Behavior, Animal drug effects, Central Nervous System Stimulants pharmacology, Cocaine pharmacology, Crosses, Genetic, DNA, Recombinant, Mice, Mice, Inbred C57BL, Motor Activity drug effects, Physical Exertion drug effects, Quantitative Trait Loci genetics, Behavior, Animal physiology, Chromosome Mapping, Chromosomes, Mammalian genetics, Motor Activity genetics, Physical Exertion genetics
- Abstract
Both human beings and animals exhibit substantial inter-individual variation in voluntary physical activity, and evidence indicates that a significant component of this variation is because of genetic factors. However, little is known of the genetic basis underlying central regulation of voluntary physical activity in mammals. In this study, using an F(2) intercross population and interval-specific congenic strains (ISCS) derived from the C57BL/6J strain and a chromosome 13 substitution strain, C57BL/6J-Chr13A/J/NA/J, we identified a 3.76-Mb interval on chromosome 13 containing 25 genes with a significant impact on daily voluntary wheel running activity in mice. Brain expression and polymorphisms between the C57BL/6J and A/J strains were examined to prioritize candidate genes. As the dopaminergic pathway regulates motor movement and motivational behaviors, we tested its function by examining cocaine-induced locomotor responses in ISCS with different levels of activity. The low-activity ISCS exhibited a significantly higher response to acute cocaine administration than the high-activity ISCS. Expression analysis of key dopamine-related genes (dopamine transporter and D1, D2, D3, D4 and D5 receptors) revealed that expression of D1 receptor was higher in the low-activity ISCS than in the high-activity ISCS in both the dorsal striatum and nucleus accumbens. Pathway analysis implicated Tcfap2a, a gene found within the 3.76-Mb interval, involved in the D1 receptor pathway. Using a luciferase reporter assay, we confirmed that the transcriptional factor, Tcfap2a, regulates the promoter activity of the D1 receptor gene. Thus, Tcfap2a is proposed as a candidate genetic regulator of the level of voluntary physical activity through its influence on a dopaminergic pathway., (© 2011 The Authors. Genes, Brain and Behavior © 2011 Blackwell Publishing Ltd and International Behavioural and Neural Genetics Society.)
- Published
- 2012
- Full Text
- View/download PDF
44. Environmental perturbation of the circadian clock disrupts pregnancy in the mouse.
- Author
-
Summa KC, Vitaterna MH, and Turek FW
- Subjects
- Animals, Female, Locomotion physiology, Mice, Mice, Inbred C57BL, Photoperiod, Pregnancy, Pregnancy Outcome, Circadian Rhythm physiology, Environment, Reproduction physiology
- Abstract
Background: The circadian clock has been linked to reproduction at many levels in mammals. Epidemiological studies of female shift workers have reported increased rates of reproductive abnormalities and adverse pregnancy outcomes, although whether the cause is circadian disruption or another factor associated with shift work is unknown. Here we test whether environmental disruption of circadian rhythms, using repeated shifts of the light:dark (LD) cycle, adversely affects reproductive success in mice., Methodology/principal Findings: Young adult female C57BL/6J (B6) mice were paired with B6 males until copulation was verified by visual identification of vaginal plug formation. Females were then randomly assigned to one of three groups: control, phase-delay or phase-advance. Controls remained on a constant 12-hr light:12-hr dark cycle, whereas phase-delayed and phase-advanced mice were subjected to 6-hr delays or advances in the LD cycle every 5-6 days, respectively. The number of copulations resulting in term pregnancies was determined. Control females had a full-term pregnancy success rate of 90% (11/12), which fell to 50% (9/18; p<0.1) in the phase-delay group and 22% (4/18; p<0.01) in the phase-advance group., Conclusions/significance: Repeated shifting of the LD cycle, which disrupts endogenous circadian timekeeping, dramatically reduces pregnancy success in mice. Advances of the LD cycle have a greater negative impact on pregnancy outcomes and, in non-pregnant female mice, require longer for circadian re-entrainment, suggesting that the magnitude or duration of circadian misalignment may be related to the severity of the adverse impact on pregnancy. These results explicitly link disruptions of circadian entrainment to adverse pregnancy outcomes in mammals, which may have important implications for the reproductive health of female shift workers, women with circadian rhythm sleep disorders and/or women with disturbed circadian rhythms for other reasons.
- Published
- 2012
- Full Text
- View/download PDF
45. Pharmacological validation of candidate causal sleep genes identified in an N2 cross.
- Author
-
Brunner JI, Gotter AL, Millstein J, Garson S, Binns J, Fox SV, Savitz AT, Yang HS, Fitzpatrick K, Zhou L, Owens JR, Webber AL, Vitaterna MH, Kasarskis A, Uebele VN, Turek F, Renger JJ, and Winrow CJ
- Subjects
- Animals, Calcium Channels, N-Type, Calcium Channels, P-Type genetics, Calcium Channels, Q-Type genetics, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Rats, Rats, Sprague-Dawley, Receptor, Muscarinic M3 genetics, Receptors, Dopamine D5 genetics, Receptors, Nicotinic genetics, Sleep Wake Disorders metabolism, Crosses, Genetic, Sleep drug effects, Sleep genetics, Sleep Wake Disorders drug therapy, Sleep Wake Disorders genetics
- Abstract
Despite the substantial impact of sleep disturbances on human health and the many years of study dedicated to understanding sleep pathologies, the underlying genetic mechanisms that govern sleep and wake largely remain unknown. Recently, the authors completed large-scale genetic and gene expression analyses in a segregating inbred mouse cross and identified candidate causal genes that regulate the mammalian sleep-wake cycle, across multiple traits including total sleep time, amounts of rapid eye movement (REM), non-REM, sleep bout duration, and sleep fragmentation. Here the authors describe a novel approach toward validating candidate causal genes, while also identifying potential targets for sleep-related indications. Select small-molecule antagonists and agonists were used to interrogate candidate causal gene function in rodent sleep polysomnography assays to determine impact on overall sleep architecture and to evaluate alignment with associated sleep-wake traits. Significant effects on sleep architecture were observed in validation studies using compounds targeting the muscarinic acetylcholine receptor M3 subunit (Chrm3) (wake promotion), nicotinic acetylcholine receptor alpha4 subunit (Chrna4) (wake promotion), dopamine receptor D5 subunit (Drd5) (sleep induction), serotonin 1D receptor (Htr1d) (altered REM fragmentation), glucagon-like peptide-1 receptor (Glp1r) (light sleep promotion and reduction of deep sleep), and calcium channel, voltage-dependent, T type, alpha 1I subunit (Cacna1i) (increased bout duration of slow wave sleep). Taken together, these results show the complexity of genetic components that regulate sleep-wake traits and highlight the importance of evaluating this complex behavior at a systems level. Pharmacological validation of genetically identified putative targets provides a rapid alternative to generating knock out or transgenic animal models, and may ultimately lead towards new therapeutic opportunities.
- Published
- 2011
- Full Text
- View/download PDF
46. Generation of N-ethyl-N-nitrosourea (ENU) diabetes models in mice demonstrates genotype-specific action of glucokinase activators.
- Author
-
Fenner D, Odili S, Hong HK, Kobayashi Y, Kohsaka A, Siepka SM, Vitaterna MH, Chen P, Zelent B, Grimsby J, Takahashi JS, Matschinsky FM, and Bass J
- Subjects
- Alkylating Agents pharmacology, Amino Acid Substitution, Animals, Binding Sites genetics, Blood Glucose genetics, Blood Glucose metabolism, Ethylnitrosourea pharmacology, Gene Expression Regulation, Enzymologic drug effects, Gene Expression Regulation, Enzymologic genetics, Humans, Hyperglycemia chemically induced, Hyperglycemia enzymology, Hyperglycemia genetics, Liver pathology, Male, Mice, Mice, Mutant Strains, Organ Specificity, Protein Folding, Recombinant Proteins genetics, Recombinant Proteins metabolism, Alkylating Agents adverse effects, Diabetes Mellitus, Experimental enzymology, Diabetes Mellitus, Experimental genetics, Diabetes Mellitus, Experimental pathology, Enzyme Activators metabolism, Ethylnitrosourea adverse effects, Glucokinase antagonists & inhibitors, Glucokinase biosynthesis, Glucokinase genetics, Liver enzymology, Mutation, Missense
- Abstract
We performed genome-wide mutagenesis in C57BL/6J mice using N-ethyl-N-nitrosourea to identify mutations causing high blood glucose early in life and to produce new animal models of diabetes. Of a total of 13 new lines confirmed by heritability testing, we identified two semi-dominant pedigrees with novel missense mutations (Gck(K140E) and Gck(P417R)) in the gene encoding glucokinase (Gck), the mammalian glucose sensor that is mutated in human maturity onset diabetes of the young type 2 and the target of emerging anti-hyperglycemic agents that function as glucokinase activators (GKAs). Diabetes phenotype corresponded with genotype (mild-to-severe: Gck(+/+) < Gck(P417R/+), Gck(K140E)(/+) < Gck(P417R/P417R), Gck(P417R/K140E), and Gck(K140E/K140E)) and with the level of expression of GCK in liver. Each mutant was produced as the recombinant enzyme in Escherichia coli, and analysis of k(cat) and tryptophan fluorescence (I(320/360)) during thermal shift unfolding revealed a correlation between thermostability and the severity of hyperglycemia in the whole animal. Disruption of the glucokinase regulatory protein-binding site (GCK(K140E)), but not the ATP binding cassette (GCK(P417R)), prevented inhibition of enzyme activity by glucokinase regulatory protein and corresponded with reduced responsiveness to the GKA drug. Surprisingly, extracts from liver of diabetic GCK mutants inhibited activity of the recombinant enzyme, a property that was also observed in liver extracts from mice with streptozotocin-induced diabetes. These results indicate a relationship between genotype, phenotype, and GKA efficacy. The integration of forward genetic screening and biochemical profiling opens a pathway for preclinical development of mechanism-based diabetes therapies.
- Published
- 2011
- Full Text
- View/download PDF
47. Identification of causal genes, networks, and transcriptional regulators of REM sleep and wake.
- Author
-
Millstein J, Winrow CJ, Kasarskis A, Owens JR, Zhou L, Summa KC, Fitzpatrick K, Zhang B, Vitaterna MH, Schadt EE, Renger JJ, and Turek FW
- Subjects
- Animals, Cerebral Cortex metabolism, Gene Expression Profiling, Gene Expression Regulation genetics, Genotype, Hypothalamus metabolism, Male, Mesencephalon metabolism, Mice, Mice, Inbred BALB C genetics, Mice, Inbred C57BL genetics, Quantitative Trait Loci genetics, Thalamus metabolism, Regulatory Elements, Transcriptional genetics, Sleep, REM genetics, Wakefulness genetics
- Abstract
Study Objective: Sleep-wake traits are well-known to be under substantial genetic control, but the specific genes and gene networks underlying primary sleep-wake traits have largely eluded identification using conventional approaches, especially in mammals. Thus, the aim of this study was to use systems genetics and statistical approaches to uncover the genetic networks underlying 2 primary sleep traits in the mouse: 24-h duration of REM sleep and wake., Design: Genome-wide RNA expression data from 3 tissues (anterior cortex, hypothalamus, thalamus/midbrain) were used in conjunction with high-density genotyping to identify candidate causal genes and networks mediating the effects of 2 QTL regulating the 24-h duration of REM sleep and one regulating the 24-h duration of wake., Setting: Basic sleep research laboratory., Patients or Participants: Male [C57BL/6J × (BALB/cByJ × C57BL/6J*) F1] N(2) mice (n = 283)., Interventions: None., Measurements and Results: The genetic variation of a mouse N2 mapping cross was leveraged against sleep-state phenotypic variation as well as quantitative gene expression measurement in key brain regions using integrative genomics approaches to uncover multiple causal sleep-state regulatory genes, including several surprising novel candidates, which interact as components of networks that modulate REM sleep and wake. In particular, it was discovered that a core network module, consisting of 20 genes, involved in the regulation of REM sleep duration is conserved across the cortex, hypothalamus, and thalamus. A novel application of a formal causal inference test was also used to identify those genes directly regulating sleep via control of expression., Conclusion: Systems genetics approaches reveal novel candidate genes, complex networks and specific transcriptional regulators of REM sleep and wake duration in mammals.
- Published
- 2011
- Full Text
- View/download PDF
48. Rhythmic leptin is required for weight gain from circadian desynchronized feeding in the mouse.
- Author
-
Arble DM, Vitaterna MH, and Turek FW
- Subjects
- Animals, Feeding Behavior, Male, Mice, Mice, Obese, Body Weight drug effects, Circadian Rhythm drug effects, Leptin pharmacology, Obesity etiology, Periodicity, Weight Gain drug effects
- Abstract
The neuroendocrine and metabolic effects of leptin have been extensively researched since the discovery, and the later identification, of the leptin gene mutated within the ob/ob mouse. Leptin is required for optimal health in a number of physiological systems (e.g. fertility, bone density, body weight regulation). Despite the extensive leptin literature and many observations of leptin's cyclical pattern over the 24-hour day, few studies have specifically examined how the circadian rhythm of leptin may be essential to leptin signaling and health. Here we present data indicating that a rhythmic leptin profile (e.g. 1 peak every 24 hours) leads to excessive weight gain during desynchronized feeding whereas non-rhythmic leptin provided in a continuous manner does not lead to excessive body weight gain under similar feeding conditions. This study suggests that feeding time can interact with leptin's endogenous rhythm to influence metabolic signals, specifically leading to excessive body weight gains during 'wrongly' timed feeding.
- Published
- 2011
- Full Text
- View/download PDF
49. Molecular neurobiology of circadian rhythms.
- Author
-
Turek FW and Vitaterna MH
- Subjects
- Animals, Circadian Rhythm physiology, Circadian Rhythm Signaling Peptides and Proteins metabolism, Gene Expression Regulation, Humans, Models, Molecular, Circadian Rhythm genetics, Circadian Rhythm Signaling Peptides and Proteins genetics, Neurobiology
- Published
- 2011
- Full Text
- View/download PDF
50. Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes.
- Author
-
Marcheva B, Ramsey KM, Buhr ED, Kobayashi Y, Su H, Ko CH, Ivanova G, Omura C, Mo S, Vitaterna MH, Lopez JP, Philipson LH, Bradfield CA, Crosby SD, JeBailey L, Wang X, Takahashi JS, and Bass J
- Subjects
- ARNTL Transcription Factors deficiency, ARNTL Transcription Factors metabolism, Aging genetics, Aging pathology, Animals, Blood Glucose analysis, Blood Glucose metabolism, CLOCK Proteins deficiency, CLOCK Proteins metabolism, Cell Proliferation, Cell Size, Cell Survival, Circadian Rhythm genetics, Diabetes Mellitus genetics, Gene Expression Profiling, Glucose Intolerance genetics, Glucose Tolerance Test, In Vitro Techniques, Insulin metabolism, Insulin Secretion, Islets of Langerhans pathology, Mice, Period Circadian Proteins genetics, Period Circadian Proteins metabolism, Phenotype, Sleep genetics, Sleep physiology, Synaptic Vesicles metabolism, Wakefulness genetics, Wakefulness physiology, ARNTL Transcription Factors genetics, CLOCK Proteins genetics, Circadian Rhythm physiology, Diabetes Mellitus metabolism, Insulin blood, Islets of Langerhans metabolism
- Abstract
The molecular clock maintains energy constancy by producing circadian oscillations of rate-limiting enzymes involved in tissue metabolism across the day and night. During periods of feeding, pancreatic islets secrete insulin to maintain glucose homeostasis, and although rhythmic control of insulin release is recognized to be dysregulated in humans with diabetes, it is not known how the circadian clock may affect this process. Here we show that pancreatic islets possess self-sustained circadian gene and protein oscillations of the transcription factors CLOCK and BMAL1. The phase of oscillation of islet genes involved in growth, glucose metabolism and insulin signalling is delayed in circadian mutant mice, and both Clock and Bmal1 (also called Arntl) mutants show impaired glucose tolerance, reduced insulin secretion and defects in size and proliferation of pancreatic islets that worsen with age. Clock disruption leads to transcriptome-wide alterations in the expression of islet genes involved in growth, survival and synaptic vesicle assembly. Notably, conditional ablation of the pancreatic clock causes diabetes mellitus due to defective beta-cell function at the very latest stage of stimulus-secretion coupling. These results demonstrate a role for the beta-cell clock in coordinating insulin secretion with the sleep-wake cycle, and reveal that ablation of the pancreatic clock can trigger the onset of diabetes mellitus.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.