Category: Moms

Circadian rhythm metabolism

Circadian rhythm metabolism

Circaxian Collections In-Press Preview Recovery resources for families Research Letters Letters to the Editor Editorials Circadiann JCI Berry Cheesecake Recipes Month Top read articles Metaboolism Medicine. The circadian clock rgythm the heart: Recovery resources for families influence Vegan omega- sources myocardial gene expression, metabolism, and function. Circadian rhythms in myocardial metabolism and contractile function: influence of workload and oleate. Studies have shown that when a circadian misalignment happens in humansthe result is decreased glucose tolerance; it is believed that ongoing misalignment could lead to insulin resistance and increase the risk of metabolic disorders like Type 2 diabetes and cardiovascular disease. Circadian rhythm metabolism

Metabloism Series Free access Address correspondence to: Joseph Bass, Northwestern University, East Superior St. Phone: Find articles ehythm Huang, Ehythm. in: Metabolsm PubMed Google Scholar.

Iodine for thyroid hormone production articles by Mmetabolism, K. Find articles by Marcheva, B. Find articles by Bass, Mteabolism. Published June 1, - Circadiah info. The discovery metabolismm the genetic basis for circadian rhythms has expanded our knowledge of the temporal organization of behavior and physiology.

The observations that the circadian gene network is present in metabooism living organisms from rhythmm to humans, that most ehythm and tissues express autonomous rhytum, and mftabolism disruption of clock genes results in metabolic dysregulation have revealed interactions between Circadin and Carpal tunnel and hand cramps rhythms at Weight control meal plans, molecular, and cellular levels.

Metabloism major challenge remains in Anti-fatigue energy formula the interplay between brain rhythmm peripheral Antioxidant foods for digestive health and in determining how these interactions promote energy homeostasis CCircadian the metabolim cycle.

In this Thermogenic fat loss supplements, we evaluate how metabokism of molecular timing may create new opportunities to understand and develop therapies for obesity and diabetes.

Studies in meatbolism dating from the 18th century originally established that hour periodic phenomena Nutrition and wound healing from rhyrhm oscillators that internally track the Circadkan of rnythm Earth. Discovery of this mechanism stemmed from deliberate Recovery resources for families experiments in flies 2and forward genetics in mice emtabolism led to the discovery of the first mwtabolism Clock gene 3 — 5.

Subsequent work has revealed that the mammalian molecular clock Recovery resources for families not only expressed metaboism the master suprachiasmatic nucleus SCN pacemaker neurons, but Berry Cheesecake Recipes within nearly ketabolism cells 67.

The purpose of this Review is to Hyperglycemia and fertility Berry Cheesecake Recipes overview of the genetic and neurobiological evidence linking circadian and Diabetic-friendly holiday meals systems and to highlight gaps in our understanding of the molecular pathways that couple these processes.

Metabolismm control Cricadian energy metabolism. B Metabolisn master clock in the SCN sends signals to the extra-SCN metzbolism, which metsbolism turn entrain peripheral tissues via hormonal, autonomic nervous Circaeian ANSmetabollsm behavioral pathways in order to regulate peripheral Circadoan control of fuel utilization and energy harvesting.

Extra-SCN regions also regulate energy homeostasis by controlling cyclic energy intake and locomotor activity. Through regulation of ryhthm intake, physical activity, and metabolic processes, both Antioxidant-Rich Wellbeing and peripheral clocks contribute to long-term weight stability by maintaining a precise balance between Circadin intake and energy expenditure.

A positive energy balance deposits stored energy mostly into adipose tissue, leading to obesity, while a rhthm energy balance results in leanness.

BMR, basal metabolic rate. In vertebrates, circadian systems are organized hierarchically with master pacemaker neurons within the hypothalamic SCN rhythj over a distributed metabbolism of extra-SCN Circadia peripheral clocks 9. The environmental light-dark Circcadian provides the principal entraining signal to the SCN, which in turn produces synchronized rhythms of behavior and physiology through alignment of circadian gene rhuthm within both extra-SCN metabolims and rhytym tissues Figure metabolisn B.

Circxdian of the neural circuit generating circadian rhythms involves projections of Post-workout muscle recovery massage SCN to a wide range of cell netabolism within metabolim the hypothalamus including the arcuate nucleus [ARC], paraventricular nucleus [PVN], lateral hypothalamic area [LHA], and dorsomedial hypothalamus rhythk refs.

For instance, clock genes are expressed cell-autonomously within cell bodies metabolis receive projections from the SCN; thus it is Detoxification and natural energy boost that oscillation of meetabolism transcripts important in the Metabolsim to metabolksm and orexigenic African mango extract for blood sugar may be subject to circadian control.

Interestingly, feeding an rhyhtm high-fat Circaduan at the incorrect circadian time has Cifcadian shown to result in increased weight Circxdian in mice 25suggesting that circadian alignment of feeding and activity is critical in the Preventing diabetes during pregnancy control of body Alleviating arthritis symptoms. It is also intriguing that Dark chocolate sophistication originating in the Rhytum directly synapse at the orexin-producing ORX-producing, also referred to as hypocretin-producing neurons within Body fat percentage evaluation LHA 1426 ORX neurons in the LHA are important in peripheral glucose metabolism 28 and also participate in Regulating healthy sugar absorption weight homeostasis, since Metaboliem receptor knockout mice rhyhtm susceptible to diet-induced obesity metaboliwm Finally, since dopaminergic signaling in the VTA neurons is implicated in reward systems associated Circadisn feeding 3031an intriguing question is Circadian rhythm metabolism metagolism drive might similarly display circadian variation Further studies are needed to define the repertoire of connections Circarian SCN and energetic neurons, to determine the impact Circaian feeding rhytthm on function and entrainment of Holistic Recovery resources for families, and to mmetabolism how clocks participate in melanocortinergic and dopaminergic signaling.

Map Cidcadian neural circuits linking SCN and extra-SCN mwtabolism important in circadian and energetic control. Non-allergenic personal care products centers receive dual input of light Sports nutrition for basketball players metabolic signals.

Light Herbal remedies for hair growth the SCN via the RHT, which in turn sends neural projections to various extra-SCN regions in the hypothalamus and brainstem that are critical for energy homeostasis and sleep, including the ARC, PVN, and ventrolateral preoptic nucleus VLPO.

The hypothalamus also receives metabolic inputs, including peptidergic hormones and nutrient metabolites, which modulate the CNS signaling. Thus signals from the exogenous environment i. IML, intermediolateral nucleus; NTS, nucleus tractus solitarius.

dSPZ, dorsal subparaventricular zone; RHT, retinohypothalamic tract; vSPZ, ventral subparaventricular zone; MCH, melanocyte concentrating hormone. In contrast, the SCN is entrained only to light but not to feeding; thus shifts in feeding time uncouple the phase of peripheral tissue clocks from that of the SCN 37 — Interactions between the molecular clock and downstream metabolic genes.

CLOCK and BMAL1 heterodimerize to drive rhythmic expression of downstream target genes shown in redwhich in turn regulate diverse metabolic processes, including glucose metabolism, lipid homeostasis, and thermogenesis.

Dashed lines represent metabolic inputs; solid lines depict interactions among core clock genes, clock-controlled genes, and nutrient sensors. Recent studies have demonstrated that various nutrient sensors are able to relay information regarding the cellular nutrient status to the circadian clock.

Interestingly, the connection between nutrient sensors and molecular clocks is bidirectional. As such, SIRT1 activity is indirectly regulated by the clock network. Together, these data demonstrate on a molecular level that there is a complex crosstalk between highly conserved nutrient sensors and the molecular clock networks, at least at the level of peripheral tissues see below for discussion regarding potential roles of CNS nutrient sensors in circadian control of metabolism.

Metabolic transcription factor and transcriptional co-activator feedback on the core clock in peripheral tissues REV-ERB, ROR, PPAR, DBP, PGC1α. NHRs are ligand-activated transcription factors, a majority of which, including REV-ERBα, RORs and PPARs, demonstrate circadian expression in peripheral tissues REV-ERBα regulates hepatic gluconeogenesis, adipocyte differentiation, and lipid metabolism, and also represses Bmal1 transcription 52 RORα competes with REV-ERBα in binding to the Bmal1 promoter and induces Bmal1 expression 54 in addition to regulating lipid metabolism PPARα, when activated by endogenous fatty acids FAsstimulates FA oxidation, regulates genes controlling lipid homeostasis, and prevents atherosclerosis In addition, PPARα positively regulates Bmal1 expression, while BMAL1 likewise activates PPARα, generating a positive feedback loop Another subtype, PPARγ, plays an important role in adipocyte differentiation and triglyceride synthesis PPARγ induces Bmal1 expression in the blood vessels, and vasculature-specific PPARγ knockout leads to marked reduction in the circadian variation in blood pressure and heart rate In addition to NHRs, Dbpa known clock target gene, regulates expression of key metabolic genes involved in gluconeogenesis and lipogenesis Additionally, the transcriptional co-activator PPARγ co-activator 1α PGC-1α 61 also displays circadian oscillations and regulates Bmal1 and Rev-erba expression.

Knockout of Pgc1a leads to abnormal diurnal rhythms of activity, body temperature, and metabolic rate, in addition to aberrant expression of clock and metabolic genes Interestingly, SIRT1 suppresses PPARγ 63 but activates PGC-1α 64and thus affects the clock network through multiple mechanisms.

Possible molecular integrators of circadian and metabolic systems in the CNS. Molecular analyses of interplay between circadian and metabolic pathways have primarily emerged from studies in liver and other peripheral tissues, yet it remains uncertain whether similar mechanisms might also couple these processes within the brain.

One question is whether specific metabolites that vary according to time of day and nutrient state fasting vs. feedingmay also affect circadian function of energy-sensing neurons.

For instance, hour oscillation in levels of glucose and FAs may in turn influence expression of circadian genes and rhythmic transcriptional outputs within hypothalamic neurons involved in glucose homeostasis 65food intake 66and energy expenditure 67 — Perturbation of metabolic homeostasis with a high-fat diet is sufficient to alter both period length and amplitude of locomotor activity Additional nutrient factors that may participate in circadian oscillations of SCN and extra-SCN neurons include AMPK, SIRT1, and the mammalian target of rapamycin mTOR.

Hypothalamic AMPK is regulated by nutrient state and hormones such as leptin and insulin 72and manipulation of its expression alters food intake and body weight 73 ; however the impact of AMPK on CNS control of locomotor behavior and physiological rhythms is still not known. NPY and POMC neuron-specific knockout of the α2 subunit of AMPK results in lean and obese phenotypes, respectively, so it will be interesting to learn whether AMPK also participates in synchronizing activity and feeding through actions within these cell groups A final potential mediator involved in both energy sensing and circadian function in the CNS is mTOR, a regulator of protein synthesis present in ARC neurons that has been shown to modulate food intake mTOR is also expressed within pacemaker neurons of the SCN, where its expression is activated by light Moreover, activation of mTOR causes phase resetting of SCN explants 79while inhibition of mTOR alters light induction of the Period gene within the SCN of the intact animal Thus amino acid metabolism may participate in both entrainment of the master clock and in the temporal organization of feeding.

Indeed, different zeitgebers may exert differing effects on circadian entrainment in the brain and peripheral tissues, leading to distinct effects on phase and amplitude of gene oscillation within these locales.

For instance, glucocorticoid secretion from adrenal glands in response to adrenocorticotropin is dependent on time of day In addition to metabolite and hormonal input into the clock, recent studies by Buhr et al. suggest that temperature and heat shock signaling pathways play a central role in entrainment of peripheral tissues A major objective in future research will thus be to delineate the role of nutrient signaling in entrainment of CNS and peripheral clocks, and to determine how these signals interact with synchronizing signals such as feeding and neuroendocrine hormones e.

Evidence for circadian integration of energetics, metabolism, and sleep in humans. In humans, many aspects of metabolism display circadian cycles, including hour variation of glucose, insulin, and leptin levels 82 Genome-wide association studies have also suggested connections between clock gene variation and fasting glucose levels 8485obesity, and metabolic syndrome 86raising interest in understanding the impact of circadian systems on human disease.

One common clinical condition suggestive of interactions between circadian rhythms and metabolism in humans is that of shift work. Numerous reports have indicated that shift workers have a higher incidence of diabetes, obesity, and cardiovascular events 187although the mechanism underlying this association is uncertain.

Scheer et al. recently tested the impact of forced circadian misalignment a simulation of shift work on neuroendocrine control of glucose metabolism and energetics In participants subjected to circadian misalignment, the investigators observed hypoleptinemia, insulin resistance, inverted cortisol rhythms, and increased blood pressure It is also interesting to note that patients with diabetes exhibit dampened amplitude of rhythms of glucose tolerance and insulin secretion 89 ; thus the relationship between circadian disruption and metabolic pathologies appears to be bidirectional in humans, suggesting that circadian disruption may lead to a vicious cycle and contribute to augmentation and progression of metabolic disease.

Direct genetic evidence in humans has linked the molecular clock with sleep 9091 through the positional cloning of mutations causing familial advanced sleep phase syndrome, which is characterized by early sleep onset and awakening In the general population, observational studies have also found that short sleep, sleep deprivation, and poor sleep quality are associated with diabetes, metabolic syndrome 8293hypoleptinemia, increased appetite, and obesity 94 A recent study showed that sleep duration correlates with the magnitude of weight loss as fat in response to caloric restriction; short sleepers appear to have more difficulty losing fat compared to long sleepers despite similar amount of weight loss Narcolepsy, a sleep disorder in which patients present with extreme daytime sleepiness due to loss of hypocretin-producing neurons 9798has been associated with elevated BMI 99 and increased incidence of obesity Night eating syndrome NES is another instance in which disrupted rhythmic patterns of sleep and eating correlate with altered metabolism and obesity Patients with NES consume significantly more of their daily energy intake at night, although their total daily food intake is similar to that of control subjects They also have abnormal rhythms of metabolic hormones, including decreased nocturnal rise in leptin, a phase shift in insulin, cortisol, and ghrelin, and inverted hour rhythms of blood glucose Interestingly, the nocturnal pattern of eating observed in NES patients is reminiscent of feeding alterations in the Clock mutant mouse.

These animals exhibit increased feeding during the normal sleep period together with increased susceptibility to diet-induced obesity A major goal is to determine whether the adverse metabolic consequences of sleep loss and accompanying feeding alterations are due to the disrupted circadian rhythms per se, to the altered sleep, or to some combination of the two.

Integration of circadian rhythms and energy homeostasis in animal models. Studies in rodents have also attempted to experimentally simulate shift work in order to further probe mechanisms linking circadian disruption with metabolic disorders.

When rats were exposed to a daily eight-hour activity schedule during their normal resting phase, they succumbed to diminished rhythms of glucose and locomotor activity, as well as obesity, which correlated with increased food intake during their resting phase

: Circadian rhythm metabolism

Access options

The results confirmed that energy expenditure was not significantly affected by the feeding time This daily change is small, but over time it could be clinically significant.

Metabolic flexibility, that is the ability to switch between carbohydrate and fat oxidation, was also increased in the TRF group, who more effectively burned fat during their fasting period. In a final trial using the same eTRF schedule, this eating pattern was shown to decrease mean h glucose and glycemic excursions Intriguingly, many of the clock genes appeared to be upregulated in the eTRF group, although the interpretation of rhythm was not possible because only two timepoints were sampled.

Taken together, the results of these trials supported that eTRF can cause weight loss primarily through reducing hunger and partly through minor increases in the thermic effect of food, while also independently improving metabolic parameters.

Human clinical trials aimed at better defining the contribution of endogenous circadian physiology to these effects are currently underway as part of the Big Breakfast Study In addition to weight loss, a highly promising application of TRF is to maintain the cardiometabolic health of shift workers.

The predominant effect of properly timed feeding in the presence of the factors of light exposure, sleep disturbance, and activity during the rest phase was clearly demonstrated by Salgado-Delgado et al.

Shift work was simulated by keeping rats on slow rotating wheels that forced wakefulness and a low level of activity from to ZT2-ZT10 , which is the inactive phase in nocturnal rodents, on weekdays for 5 weeks. On weekends, all rats were left undisturbed in their home cages with AL food access.

During the work week, shift workers and controls were assigned to AL feeding, active phase TRF, or inactive phase TRF. Unsurprisingly, the AL-fed shift workers ate most of their food during the inactive phase when they were kept awake.

They and the inactive TRF groups gained more weight and had greater visceral adiposity compared with the AL controls, active phase TRF controls, or active phase TRF shift workers, despite similar food intake across all groups.

Even though the work schedule disrupted sleep and activity patterns, only mistimed feeding explained the differences in weight and fat gain. The corticosterone rhythms remained unchanged, corroborating that SCN clock outputs are resilient to behavioral feedback, which contributes to internal desynchrony during shiftwork.

However, disturbances in glucose and TAG rhythms and the accumulation of abdominal fat were all prevented when shift work was combined with active phase TRF. Similar protection by active phase TRF is also observed in simulated jetlag. In addition to the timing of food intake, the composition of the diet alters peripheral circadian alignment and acutely induces phase shifts.

An important mechanism by which this occurs is through feedback from altered behavioral rhythms in feeding and activity. Other mechanisms include impaired or enhanced inter-tissue communication as well as interactions of the core clock with particular nutrient-sensing pathways.

The following section focuses on the effects of the well-studied high-fat diet and the increasingly studied ketogenic diet on circadian metabolism. The modern obesogenic food environment is most commonly modeled in animals with the high-fat diet HFD. Mice given AL access to the HFD reliably develop obesity and a phenotype similar to the metabolic syndrome in humans, characterized by insulin resistance, hepatic steatosis, hypercholesterolemia, and dyslipidemia As also occurs in humans, this metabolic disruption is preceded by circadian disarrangement.

In animals, an immediate alteration in rhythmic behavior is observed upon the start of the HFD. The rapid onset of behavioral arrhythmicity indicates that it is likely to occur by a mechanism independent of clock gene regulation, which would take longer to adapt.

Highly palatable foods or particular nutrient compositions can directly and rapidly signal to orexigenic centers It appears that the homeostatic feeding circuits acutely responsive to the HFD are independent of clock regulation. One week of HFD did not alter clock gene expression in the SCN, arcuate nucleus, or pituitary This is despite reciprocal connections between the arcuate and SCN Moreover, 6 weeks of HFD did not alter clock gene expression in the mediobasal hypothalamus 49 , a region containing AgRP neurons known to possess an autonomous clock controlling the rhythms of hunger and satiety The HFD therefore appears to acutely alter feeding behavior by clock-independent central mechanisms.

Although the rapid shift in feeding behavior upon beginning high-fat feeding is likely to be clock-independent, the HFD does also interfere with the master clock in the SCN.

Interestingly, hypocaloric feeding also alters photic entrainment , , but in contrast to the HFD, enables a more rapid phase shift response to light In summary, diet composition can alter photic entrainment of the SCN, but the most important effects of the HFD on the circadian system are likely to occur indirectly through phase shifts in peripheral clocks in response to altered feeding behavior.

Shortly following the altered feeding rhythm, the clock genes of peripheral tissues are altered in amplitude and phase. The liver clock is the most widely studied and, as previously mentioned, perhaps the most sensitive to feeding rhythms.

Long-term HFD feeding was shown to dampen hepatic clock gene expression , After just 1 week on HFD, the hepatic clock was found to phase advance by 5 h A similarly sized phase advance of 4 h was confirmed by Branecky et al.

Interestingly, they found that similar to the liver's rapid response at the beginning of HFD, it rapidly reverted to its normal phase within 7 days of the return to normal chow. This response lagged behind the feeding rhythms, which were restored within 2 days.

As the changes in amplitude and phase are consistently found to occur after the changes in feeding rhythm, they are likely to be the result behavioral feedback. Diet composition may act as a zeitgeber in the liver by altering food intake timing through initially clock-independent mechanisms.

The phase advance in the liver clock is comparatively mild compared with the large-scale changes to downstream clock-controlled genes CCG observed with the HFD.

In a comprehensive study of the effects of HFD compared with normal chow NC on the circadian transcriptome and metabolome in mouse liver, Eckel-Mahan et al. Moreover, most of those that were oscillatory under both diet conditions exhibited phase shifts and overall advance under high-fat feeding.

Another common effect of the HFD was to reduce the amplitude of oscillating metabolites and transcripts, and the transcripts that lost oscillations on the HFD had a robust peak at ZT8, coinciding with the greatest activity of CLOCK:BMAL1 at target genes. In other cases, the HFD produced de novo oscillation of the metabolites and transcripts encoding their regulatory enzymes, such as in the methionine cycle.

These de novo oscillations appeared to be downstream of the oscillations in the nuclear accumulation of transcription factors outside the core clock, especially PPARγ and SREBP Because of the relation between the methionine cycle and epigenetic methylation reactions , its newly oscillatory status may be one mechanism in the large-scale reprogramming observed under an HFD.

Importantly, many of these changes were observable after an acute HFD exposure of just 3 days and were reversed with 2 weeks on NC, confirming these effects were the result of the HFD rather than diet-induced obesity Corroborating and expanding on these results, the HFD was again recently shown to induce the rhythmic binding of non-core clock transcription factors to metabolic target genes in the liver, including SREBP Guan et al.

As is usual under high-fat feeding, the mice developed insulin resistance, hyperlipidemia, and fatty liver. The latter condition is targeted in humans by PPAR agonist drugs. Impressively, treatment with the PPARα agonist in the HFD fed mice at the peak of its activity ZT8 resulted in a greater reduction of hepatic lipid accumulation and serum triglyceride levels than when it was given at the nadir of PPARα activity ZT20 This strategic administration of drugs in coordination with circadian rhythms is an example chronotherapy, which is a promising direction for translational work.

It is known that lipid profiles in humans have a robust circadian rhythm In this study, however, the samples were taken only at three timepoints within an 8-h period, which was insufficient to extrapolate h oscillations.

The effects of dietary fat on clock gene expression and circadian regulation in humans will require further investigation. In addition to altering the liver clock and causing widespread alterations in rhythmic metabolism, the HFD impairs inter-tissue communication, thereby further exacerbating the misalignment of circadian rhythms between key metabolic tissues.

For instance, adiponectin from mature adipocytes regulates hepatic lipid metabolism by activating AMPK, leading to the phosphorylation and inactivation of the fatty acid synthesis enzyme ACC, the activation of PPARα to increase FA oxidation, and the potentiation of insulin signaling to inhibit gluconeogenesis In the livers of mice fed the HFD, rhythms in AMPK and ACC transcripts were abolished, and a 3-h phase delay and dramatic dampening were observed in key components of the adiponectin signaling pathway AdipoR1, Pepck, and PPARα as well as the core clock gene Per1 A similar phenotype during HFD feeding was observed in skeletal muscle and adipose In addition to adiponectin, normal daily rhythms in plasma insulin, ghrelin, and leptin have been shown to be disrupted in rats fed an HFD Along with endocrine system interference, the HFD also directly misaligns tissues by differentially regulating their clocks.

The HFD causes rapid phase shifts in the liver within 1 week, whereas the clocks of the lung, spleen, aorta, gonadal white adipose were unchanged in phase It is possible that these tissues rely less on food intake as a zeitgeber, or that they adapt more gradually to HFD exposure.

Whether the liver responds more strongly or just more rapidly to the HFD, this discrepancy in responsiveness is important because the liver then quickly becomes misaligned with other tissue clocks. The induction of peripheral misalignment by HFD was strikingly demonstrated in recent circadian metabolomics studies.

Abbondante et al. Dyar et al. They found tissue-specific alterations in circadian metabolism that caused the large-scale disruption of the temporal cohesion between the tissues.

Positive and negative correlations in time of tissue metabolites are indicative of shared metabolic networks and the temporal gating of incompatible metabolic processes, respectively.

The major source of metabolite correlations on NC were through those circulating in serum. Lipids in particular were highly correlated in time under NC and they lost this inter-tissue alignment under the HFD. Unfortunately, however, samples were taken at a.

in the fasted state and at p. after dinner, making it impossible to separate the effect of time of day from acute effects of the evening meal; indeed, among the metabolites heightened in the evening after dinner on the HFD were fatty acids and ketone bodies, while xenobiotics that are known food additives were higher in the evening on both diets.

The effect of diet composition on the human circadian metabolome requires further investigation. Hence, the term HFD is somewhat of a misnomer that perhaps more accurately designates a high-fat, moderate-carbohydrate diet. This is to be distinguished from the even higher fat and very low carbohydrate ketogenic diet KD.

In humans, a KD typically restricts daily carbohydrates to under 50 g per day Whereas, the HFD is used experimentally to induce obesity and metabolic disease, the KD is under investigation because of its therapeutic potential to ameliorate these states Whereas, the HFD induces hyperphagia, appetite inhibition is thought to be an important part of KD efficacy Similarly, the interaction between the KD and the circadian clock is quite distinct from the HFD case.

The KD drives metabolism toward the pathways induced under fasting or caloric restriction without the need to restrict energy intake. Gluconeogenesis, fatty acid oxidation, and ketogenesis are upregulated, while glycolysis and de novo lipogenesis are inhibited The circulating levels of ketone bodies in healthy humans have long been recognized to follow daily rhythms, with overall higher levels occur during carbohydrate restriction as in the KD or in fasting The induction of ketogenic genes in the liver during fasting is controlled by the mTOR—PPARα axis , and the PPAR family is known to be circadian-regulated 17 , Moreover, the hepatic expression of the clock component Per2 was shown to be necessary in ketogenesis.

Per2 is a direct regulator of Cpt1a expression, a rate-limiting enzyme that transfers long-chain fatty acids to the inner mitochondrial membrane for ß-oxidation, and is an indirect regulator of Hmgcs2 , the rate-limiting enzyme for ketogenesis from the resulting acetyl-CoA The KD may also increase transcriptional activation of CCG's by the CLOCK:BMAL1 complex peaking at ZT ; the KD diet has been shown to upregulate the clock output gene Dbp in the liver, heart, kidney, and adipose tissue The circadian transcriptome was recently analyzed in the liver and intestine ileal epithelia of mice fed the KD or isocaloric quantities of NC, revealing large-scale alterations in oscillating transcripts In contrast to the HFD, which mildly dampened clock gene expression , the amplitude of clock genes were either unaltered or slightly increased under the KD.

The rhythm in the respiratory metabolism was abolished by KD as it was in the HFD The rhythm in respiratory metabolism was abolished by KD as in the HFD. An arrhythmic respiratory exchange ratio RER is considered evidence of metabolic impairment , as the body is unable to efficiently switch from burning carbohydrate to burning fatty acids during the overnight fast.

However, in the context of the KD, this flattened RER is an expected outcome given that fatty acids are the predominant fuel available. Indeed, the RER remained at ~0. Similar to the case of the HFD, the greatest alteration in the liver was not at the level of the clock genes themselves but in the chromatin recruitment of CLOCK:BMAL1 to clock-controlled genes CCGs.

However, the overall effects were opposite Figure 4. In the HFD, the occupancy of CLOCK:BMAL1 at target gene promoters was attenuated, causing blunted oscillations of CCGs In contrast, the KD increased the amplitude of clock target genes, including Dbp and Nampt , following greater Bmal1 recruitment to these sites at the critical timepoints.

This KD-induced amplitude increase was not seen in Clock- mutant mice, confirming it is the output of the clock Figure 4. The effects of a high-fat diet HFD and ketogenic diet KD on clock-controlled gene CCG expression in mouse liver.

The HFD abolishes rhythms in CCGs by reducing CLOCK:BMAL1 chromatin binding , whereas the KD increases rhythmic CLOCK:BMAL1 binding and thereby CCG expression amplitude Both diets induce de novo rhythms in other metabolic genes because of the rhythmic nuclear accumulation of non-clock transcription regulators PPARg and SREBP-1 in liver under HFD; PPARα in intestine and rhythmic histone deacetylation by serum BHB under KD.

In the intestine of KD fed mice, the dietary intake of fatty acids induced rhythmic nuclear accumulation of PPARα and the expression of its target genes Corresponding to the HMGCS2 induction were oscillations of the ketone body ß-hydroxybutyrate ßOHB in the gut and serum.

ßOHB has received recent attention as a signaling molecule with histone deacetylase activity , and it appears to also be a cofactor in a recently defined epigenetic marker, histone b-hydroxyl-butyrylation, which is associated with active gene expression Therefore, transcriptional reprogramming under the KD may partly be through chromatin remodeling by ßOHB.

Whether the overall effect of this large-scale remodeling is beneficial is not yet clear; however, a positive effect of intestinal ßOHB in maintaining the stemness and regenerative capability of intestinal stem cells was recently identified Experiments using circadian mutant mice fed the HFD illustrate the interaction between circadian clock function and the response to different diet compositions.

The RAR-related orphan receptor alpha RORα is a nuclear receptor that functions as a ligand-dependent transcriptional factor in lipid metabolism and in circadian regulation These mice exhibit a lean and dyslipidemic phenotype when fed NC.

When fed the HFD, they are resistant to weight gain and hepatic steatosis, although they also develop particularly severe atherosclerosis This finding attracted attention to RORα and other circadian nuclear receptors Reverb's as drug targets for metabolic disease Recent work has suggested that RORα agonists may be effective in preventing hepatic steatosis , and liver-specific Ror α deletion correspondingly worsens hepatic steatosis Thus, the particular phenotype of circadian mutant mice is dependent on diet composition.

Further illustrating this interaction between the circadian clock and diet composition, some circadian mutants have strikingly different phenotypes on NC compared to the HFD.

It is interesting that in both cases, the mutant mice exhibited disrupted feeding rhythms even on NC, but a disrupted core clock and feeding rhythm were not sufficient to produce weight gain.

Instead, they strongly predisposed the mice to diet-induced obesity. Both eating at the wrong time and the HFD independently lead to internal misalignment between metabolic organs 84 , The HFD also acutely alters food intake timing, and this appears to be at the root of some of its adverse effects on circadian metabolism.

The time-restricted feeding TRF of HFD within 8 h during the active phase, despite isocaloric intake compared with AL-fed controls, protected mice against diet-induced obesity DIO and related metabolic dysfunction Similarly, mice on a h active phase TRF were able to maintain normal metabolic parameters on both low- and high-fat diets The combination of this obesogenic diet with active phase TRF also restored the rhythms in liver clock gene expression and reduced plasma insulin, leptin, and proinflammatory cytokines, suggesting protection against systemic inflammation Active phase TRF acts as a protective buffer against an array of nutritional challenges.

Although the HFD is the most common diet used to induce obesity in rodent studies, it is one among multiple experimental diets that model the energy-dense and processed foods eaten by modern humans and produce characteristic metabolic disease.

For instance, a high-fructose diet causes insulin resistance, cardiac damage, and hepatic steatosis , They found that restricting food access to 8—9 h during the active phase effectively prevented or even reversed the already established obesity and metabolic dysfunction caused by any of these diets when fed AL, and this was accompanied by the restoration of temporal dynamics in several key metabolic pathways Relevant to humans, this active phase TRF provided effective protection against diet-induced metabolic disease even when mice fed freely on weekends.

To get the protective benefits of TRF it is imperative that the feeding window occur during the active phase. Timing the food intake with the active phase appears to confer a metabolic advantage because physiological systems are more prepared for the nutritional challenge.

For example, in the response to a high-fat meal at the beginning of the active period fatty acid oxidation was shown to be increased, but this metabolic flexibility was lacking in the response to the same high-fat meal at the end of the active phase Over time, improperly timed feeding can promote metabolic disease independently of the daily total number of fat-derived calories consumed In addition, inactive phase TRF of either the HFD or NC for 6 days almost completely inverted the liver clock's phase , causing misalignment with other peripheral clocks that were not as responsive to this zeitgeber.

The same peripheral misalignment was observed in mice on inactive phase TRF of a high-fat high-sucrose HFHS diet, along with hyperphagia due to central leptin resistance Collectively, the inactive phase feeding of an array of diets NC, HFD, or HFHS impairs nutrient handling, induces peripheral clock misalignment, and disrupts signaling between metabolic tissues.

The HFD and other obesogenic diets interfere with normal feeding rhythms, but mice consuming isocaloric amounts of obesogenic diets stay slim and healthy as long as consumption is confined to the active phase.

Mutations of clock genes also result in disrupted feeding and activity rhythms in gene-specific metabolic phenotypes. As the results of previous work have shown, the susceptibility of circadian mutants to obesity and various metabolic diseases depends on the composition of the diet they are fed in interaction with their particular genotype.

Such an interpretation is further supported by a recent study in mice with SCN-targeted ablation of Bmal1. In constant darkness, these mice develop arrhythmic activity, feeding, and peripheral clock gene expression in the liver, pancreas, and adipose.

With this arrhythmicity comes increased adiposity and impaired glucose tolerance. However, TRF was sufficient to restore peripheral tissue rhythms, body weight, and glucose utilization Disrupted peripheral clock rhythms, whether from circadian mutations or environmental perturbation e.

TRF is a promising intervention in this context. Periods of fasting are likely to restore metabolic function in an otherwise obesogenic environment independent of h calorie intake partly by permitting the expression of metabolic pathways inhibited by food intake.

For instance, liver-specific AMPK overexpression mimicking the fasting state inhibited de novo lipogenesis and was sufficient to prevent hepatic steatosis in mice fed a high-fructose diet The takeaway is therefore 2-fold: optimal nutrient intake is confined to a restricted time period during the animal's active phase and leaves a sufficiently long fasting window.

A key question that remains is how long of a fasting window is necessary to see the protective benefits of TRF in humans.

Is it necessary to fast as long as 18 h or are 11—12 h sufficient 68? The answer almost certainly depends on baseline metabolic health and the extent of the desired change in weight and metabolic parameters. The circadian expression of the core clock and the genes under its regulation is found not only in the master clock i.

Peripheral clocks respond not only to the synchronizing cues emanating from the light-entrained master clock but also to rhythms in feeding and fasting. Furthermore, different peripheral tissues have varying degrees of responses to food intake during the inactive phase, thus potentiating peripheral misalignment.

Obesogenic diets also disrupt feeding rhythms and thereby circadian metabolism. In modern humans, the discordance between behavioral and endogenous clock rhythms is prevalent, and this temporal misalignment leads to systemic metabolic dysregulation.

While evening light exposure will likely continue to be a reality, food intake is a powerful zeitgeber around which behaviors are more plastic. Active phase TRF may have remarkable potential to prevent the deleterious metabolic effects of both obesogenic diets and night shift work.

The relative importance of the core molecular clock as a mediator of food intake signals remains to be delineated. The answers to these questions could be important in developing potential pharmacological interventions e.

Other lifestyle interventions that could influence circadian metabolism and prevent its misalignment are also being examined. Exercise in particular may influence peripheral clocks in skeletal muscles and adipose tissue as it activates many of the same pathways as fasting does, and these feed back into the core clock Indeed, acute exercise was found to alter the human subcutaneous adipose tissue transcriptome Recently, a human phase response curve for exercise was created Phase response curves illustrate the relationship between the time of zeitgeber exposure and the resultant phase shifts advances or delays of the clock.

A phase response curve for food intake will likewise be an essential tool for understanding how to prevent or alleviate circadian misalignment by TRF.

Accessing the nutrient-responsive peripheral circadian system in humans remains challenging, and innovative methods are required to translate the large body of mechanistic work in animals. Serial sampling is necessary to observe h rhythms, which presents the particular challenge for invasive studies e.

Nevertheless, because animal studies have shown that the circadian metabolome and its response to nutrition are highly tissue-specific , , this work is highly important. Circadian transcriptomics and metabolomics studies in humans have been carried out using serial samples of plasma, muscle biopsies, and subcutaneous adipose tissue.

However, further studies are needed to synthesize these large datasets toward identification of biomarkers of circadian metabolic function.

Indeed, preliminary efforts are underway to identify human circadian biomarkers If they are sufficiently well-defined, these biomarkers could be screened at their peaks and nadirs to determine interpretable signatures of circadian alignment and identify early markers of circadian disruption in metabolic pathophysiology.

Alternatives to plasma for non-invasive serial sampling include urine and breath. The latter was used in circadian metabolomics in a proof-of-principle study , and it has recently been highlighted as an ideal method for continuous sampling and rapid untargeted metabolomic analysis , making this an exciting avenue for translational work.

A key challenge in the translational application of circadian biology is the large interindividual variations in metabolite rhythms 92 , , What causes these variations, and how are they related to health outcomes?

In some cases, variability may be associated with chronotype The interindividual differences in the circadian metabolic response to misaligned zeitgebers, especially in the context of shift work, warrant further attention.

Moreover, the response of the human circadian metabolome under various diet conditions deserves further exploration given the largescale alterations observed in mice At the same time, future research in animal models may elucidate the circadian effects of specific macronutrient ratios, micronutrients and supplements [e.

Together this will inform novel therapeutic approaches to combat metabolic disease in the modern environment. This work was funded by grants from the Natural Sciences and Engineering Research Council NSERC, RGPIN of Canada, the New Investigator Grant of Banting and Best Diabetes Centre BBDC , and the Canadian Institutes of Health Research CIHR, PJT to H-KS.

LP was a recipient of the Charles Hollenberg Summer Studentship awarded by the Banting and Best Diabetes Centre BBDC at the University of Toronto.

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. Patel VR, Ceglia N, Zeller M, Eckel-Mahan K, Sassone-Corsi P, Baldi P.

The pervasiveness and plasticity of circadian oscillations: the coupled circadian-oscillators framework.

doi: PubMed Abstract CrossRef Full Text Google Scholar. Schibler U. Circadian time keeping: the daily ups and downs of genes, cells, and organisms. Prog Brain Res. Rosbash M. The implications of multiple circadian clock origins. PLoS Biol. Roenneberg T, Merrow M. The circadian clock and human health.

Curr Biol. Spoelstra K, Wikelski M, Daan S, Loudon AS, Hau M. Natural selection against a circadian clock gene mutation in mice. Proc Natl Acad Sci. Bass J, Takahashi JS.

Circadian integration of metabolism and energetics. Eckel-Mahan K, Sassone-Corsi P. Metabolism and the circadian clock converge. Physiol Rev. de Goede P, Wefers J, Brombacher EC, Schrauwen P, Kalsbeek A. Circadian rhythms in mitochondrial respiration.

J Mol Endocrinol. Panda S. Circadian physiology of metabolism. Koronowski KB, Kinouchi K, Welz PS, Smith JG, Zinna VM, Shi J, et al. Defining the Independence of the liver circadian clock. Partch CL, Green CB, Takahashi JS.

Molecular architecture of the mammalian circadian clock. Trends Cell Biol. Gaucher J, Montellier E, Sassone-Corsi P. Molecular cogs: interplay between circadian clock and cell cycle.

Takahashi JS. Transcriptional architecture of the mammalian circadian clock. Nat Rev Genet. Papazyan R, Zhang Y, Lazar MA. Genetic and epigenomic mechanisms of mammalian circadian transcription. Nat Struct Mol Biol. Ribas-Latre A, Eckel-Mahan K. Interdependence of nutrient metabolism and the circadian clock system: importance for metabolic health.

Mol Metab. Atger F, Mauvoisin D, Weger B, Gobet C, Gachon F. Regulation of mammalian physiology by interconnected circadian and feeding rhythms.

Front Endocrinol. Chen L, Yang G. PPARs integrate the mammalian clock and energy metabolism. PPAR Res. Asher G, Schibler U. Crosstalk between components of circadian and metabolic cycles in mammals. Cell Metab.

Gachon F. Physiological function of PARbZip circadian clock-controlled transcription factors. Ann Med. Sato S, Parr EB, Devlin BL, Hawley JA, Sassone-Corsi P.

Human metabolomics reveal daily variations under nutritional challenges specific to serum and skeletal muscle. Dibner C, Schibler U, Albrecht U. The mammalian circadian timing system: organization and coordination of central and peripheral clocks. Ann Rev Physiol. Bhadra U, Thakkar N, Das P, Bhadra MP.

Evolution of circadian rhythms: from bacteria to human. Sleep Med. Daan S, Pittendrigh CS. A functional analysis of circadian pacemakers in nocturnal rodents. J Comp Physiol. CrossRef Full Text Google Scholar.

Jones JR, Simon T, Lones L, Herzog ED. SCN VIP neurons are essential for normal light-mediated resetting of the circadian system. J Neurosci. Schibler U, Gotic I, Saini C, Gos P, Curie T, Emmenegger Y, et al.

Clock-talk: interactions between central and peripheral circadian oscillators in mammals. In: Grodzicker T, Stewart D, Stillman B, editors. Cold Spring Harbor Symposia on Quantitative Biology. Woodbury, NY: Cold Spring Harbor Laboratory Press Mohawk JA, Green CB, Takahashi JS. Central and peripheral circadian clocks in mammals.

Ann Rev Neurosci. Challet E. Circadian aspects of adipokine regulation in rodents. Best Pract Res Clin Endocrinol Metab. Sujino M, Masumoto KH, Yamaguchi S, van der Horst GT, Okamura H, Inouye SIT. Suprachiasmatic nucleus grafts restore circadian behavioral rhythms of genetically arrhythmic mice.

Nagai K, Nishio T, Nakagawa H, Nakamura S, Fukuda Y. Effect of bilateral lesions of the suprachiasmatic nuclei on the circadian rhythm of food-intake. Brain Res. Husse J, Leliavski A, Tsang AH, Oster H, Eichele G.

The light-dark cycle controls peripheral rhythmicity in mice with a genetically ablated suprachiasmatic nucleus clock. FASEB J. Weaver DR, van der Vinne V, Giannaris EL, Vajtay TJ, Holloway KL, Anaclet C.

Functionally complete excision of conditional alleles in the mouse suprachiasmatic nucleus by Vgat-ires-Cre. J Biol Rhythms. Coomans CP, van den Berg SA, Lucassen EA, Houben T, Pronk AC, van der Spek RD, et al.

The suprachiasmatic nucleus controls circadian energy metabolism and hepatic insulin sensitivity. Su Y, Cailotto C, Foppen E, Jansen R, Zhang Z, Buijs R, et al. The role of feeding rhythm, adrenal hormones and neuronal inputs in synchronizing daily clock gene rhythms in the liver.

Mol Cell Endocrinol. Su Y, Foppen E, Zhang Z, Fliers E, Kalsbeek A. Effects of 6-meals-a-day feeding and 6-meals-a-day feeding combined with adrenalectomy on daily gene expression rhythms in rat epididymal white adipose tissue. Genes Cells. Damiola F, Le Minh N, Preitner N, Kornmann B, Fleury-Olela F, Schibler U.

Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes Dev. Stokkan KA, Yamazaki S, Tei H, Sakaki Y, Menaker M. Entrainment of the circadian clock in the liver by feeding.

Hara R, Wan K, Wakamatsu H, Aida R, Moriya T, Akiyama M, et al. Restricted feeding entrains liver clock without participation of the suprachiasmatic nucleus.

Schibler U, Ripperger J, Brown SA. Peripheral circadian oscillators in mammals: time and food. Bray MS, Ratcliffe WF, Grenett MH, Brewer RA, Gamble KL, Young ME. Quantitative analysis of light-phase restricted feeding reveals metabolic dyssynchrony in mice. Int J Obes. Crosby P, Hamnett R, Putker M, Hoyle NP, Reed M, Karam CJ, et al.

Le Minh N, Damiola F, Tronche F, Schütz G, Schibler U. Glucocorticoid hormones inhibit food-induced phase-shifting of peripheral circadian oscillators. EMBO J. Saini C, Liani A, Curie T, Gos P, Kreppel F, Emmenegger Y, et al. Real-time recording of circadian liver gene expression in freely moving mice reveals the phase-setting behavior of hepatocyte clocks.

Sen S, Raingard H, Dumont S, Kalsbeek A, Vuillez P, Challet E. Ultradian feeding in mice not only affects the peripheral clock in the liver, but also the master clock in the brain. Chronobiol Int. Challet E, Caldelas I, Graff C, Pévet P.

Synchronization of the molecular clockwork by light-and food-related cues in mammals. Biol Chem. Rudic RD, McNamara P, Curtis AM, Boston RC, Panda S, Hogenesch JB, et al. BMAL1 and CLOCK, two essential components of the circadian clock, are involved in glucose homeostasis.

Zarrinpar A, Chaix A, Panda S. Daily eating patterns and their impact on health and disease. Trends Endocrinol Metab. Ando H, Kumazaki M, Motosugi Y, Ushijima K, Maekawa T, Ishikawa E, et al.

Ho A, Chin A. Circadian feeding and drinking patterns of genetically obese mice fed solid chow diet. Physiol Behav. Kohsaka A, Laposky AD, Ramsey KM, Estrada C, Joshu C, Kobayashi Y, et al. High-fat diet disrupts behavioral and molecular circadian rhythms in mice.

Mendoza J, Pévet P, Challet E. High-fat feeding alters the clock synchronization to light. J Physiol. James SM, Honn KA, Gaddameedhi S, Van Dongen HP. Shift work: disrupted circadian rhythms and sleep—implications for health and well-being.

Curr Sleep Med Rep. Wang F, Zhang L, Zhang Y, Zhang B, He Y, Xie S, et al. Meta-analysis on night shift work and risk of metabolic syndrome.

Obes Rev. Brum MCB, Dantas Filho FF, Schnorr CC, Bottega GB, Rodrigues TC. Shift work and its association with metabolic disorders.

Diabetol Metab Syndr. Kervezee L, Kosmadopoulos A, Boivin DB. Metabolic and cardiovascular consequences of shift work: the role of circadian disruption and sleep disturbances. Eur J Neurosci.

Opperhuizen AL, van Kerkhof LW, Proper KI, Rodenburg W, Kalsbeek A. Rodent models to study the metabolic effects of shift work in humans. Front Pharmacol.

Scheer FA, Hilton MF, Mantzoros CS, Shea SA. Adverse metabolic and cardiovascular consequences of circadian misalignment. Proc Natl Acad Sci USA. Javeed N, Matveyenko AV. Circadian etiology of type 2 diabetes mellitus. Stenvers DJ, Scheer FA, Schrauwen P, la Fleur SE, Kalsbeek A. Circadian clocks and insulin resistance.

Nat Rev Endocrinol. Morris CJ, Yang JN, Garcia JI, Myers S, Bozzi I, Wang W, et al. Endogenous circadian system and circadian misalignment impact glucose tolerance via separate mechanisms in humans.

Depner CM, Melanson EL, McHill AW, Wright KP. Mistimed food intake and sleep alters hour time-of-day patterns of the human plasma proteome. Lee YH, Wang MY, Yu XX, Unger RH. Glucagon is the key factor in the development of diabetes.

Peschke E, Bähr I, Mühlbauer E. Melatonin and pancreatic islets: interrelationships between melatonin, insulin and glucagon. Int J Mol Sci. Gan Y, Yang C, Tong X, Sun H, Cong Y, Yin X, et al. Shift work and diabetes mellitus: a meta-analysis of observational studies.

Occup Environ Med. Shan Z, Li Y, Zong G, Guo Y, Li J, Manson JE, et al. Rotating night shift work and adherence to unhealthy lifestyle in predicting risk of type 2 diabetes: results from two large US cohorts of female nurses.

Shaw E, Dorrian J, Coates AM, Leung GK, Davis R, Rosbotham E, et al. Temporal pattern of eating in night shift workers. Joo J, Cox CC, Kindred ED, Lashinger LM, Young ME, Bray MS.

The acute effects of time-of-day-dependent high fat feeding on whole body metabolic flexibility in mice. Lennernäs M, Hambraeus L, Åkerstedt T. Shift related dietary intake in day and shift workers.

Gill S, Panda S. A smartphone app reveals erratic diurnal eating patterns in humans that can be modulated for health benefits. Kolbe I, Oster H. Focus: clocks and cycles: chronodisruption, metabolic homeostasis, and the regulation of inflammation in adipose tissues.

Yale J Biol Med. Google Scholar. Roenneberg T, Allebrandt KV, Merrow M, Vetter C. Social jetlag and obesity. Vetter C, Devore EE, Ramin CA, Speizer FE, Willett WC, Schernhammer ES. Mismatch of sleep and work timing and risk of type 2 diabetes. Diabetes Care. Parsons MJ, Moffitt TE, Gregory AM, Goldman-Mellor S, Nolan PM, Poulton R, et al.

Social jetlag, obesity and metabolic disorder: investigation in a cohort study. Korsiak J, Tranmer J, Day A, Aronson KJ. Sleep duration as a mediator between an alternating day and night shift work schedule and metabolic syndrome among female hospital employees.

Eckel RH, Depner CM, Perreault L, Markwald RR, Smith MR, McHill AW, et al. Morning circadian misalignment during short sleep duration impacts insulin sensitivity. Nedeltcheva AV, Scheer FA. Metabolic effects of sleep disruption, links to obesity and diabetes. Curr Opin Endocrinol Diabetes Obes.

Spaeth AM, Dinges DF, Goel N. Effects of experimental sleep restriction on weight gain, caloric intake, and meal timing in healthy adults. Chattu VK, Chattu SK, Burman D, Spence DW, Pandi-Perumal SR. The interlinked rising epidemic of insufficient sleep and diabetes mellitus.

Moore RY. Suprachiasmatic nucleus in sleep—wake regulation. Burgess HJ. Partial sleep deprivation reduces phase advances to light in humans. Leproult R, Holmbäck U, Van Cauter E. Circadian misalignment augments markers of insulin resistance and inflammation, independently of sleep loss.

Stone JE, Sletten TL, Magee M, Ganesan S, Mulhall MD, Collins A, et al. Temporal dynamics of circadian phase shifting response to consecutive night shifts in healthcare workers: role of light—dark exposure. Ding G, Gong Y, Eckel-Mahan KL, Sun Z. Central circadian clock regulates energy metabolism.

In: Wu Q, Zheng R, editors. Neural Regulation of Metabolism. Advances in Experimental Medicine and Biology , Vol. Singapore: Springer McFadden E, Jones ME, Schoemaker MJ, Ashworth A, Swerdlow AJ.

The relationship between obesity and exposure to light at night: cross-sectional analyses of over , women in the Breakthrough Generations Study. Am J Epidemiol.

Opperhuizen AL, Stenvers DJ, Jansen RD, Foppen E, Fliers E, Kalsbeek A. Light at night acutely impairs glucose tolerance in a time-, intensity-and wavelength-dependent manner in rats. Fonken LK, Workman JL, Walton JC, Weil ZM, Morris JS, Haim A, et al. Light at night increases body mass by shifting the time of food intake.

Christie S, Vincent AD, Li H, Frisby CL, Kentish SJ, O'Rielly R, et al. A rotating light cycle promotes weight gain and hepatic lipid storage in mice. Am J Physiol. Dallmann R, Viola AU, Tarokh L, Cajochen C, Brown SA. The human circadian metabolome. Davies SK, Ang JE, Revell VL, Holmes B, Mann A, Robertson FP, et al.

Effect of sleep deprivation on the human metabolome. Brandauer J, Vienberg SG, Andersen MA, Ringholm S, Risis S, Larsen PS, et al.

AMP-activated protein kinase regulates nicotinamide phosphoribosyl transferase expression in skeletal muscle. Aschoff J. Circadian rhythms: influences of internal and external factors on the period measured in constant conditions 1. Zeitschrift für Tierpsychologie.

Skene DJ, Skornyakov E, Chowdhury NR, Gajula RP, Middleton B, Satterfield BC, et al. Separation of circadian-and behavior-driven metabolite rhythms in humans provides a window on peripheral oscillators and metabolism.

Kervezee L, Cermakian N, Boivin DB. Individual metabolomic signatures of circadian misalignment during simulated night shifts in humans. Rotter M, Brandmaier S, Covic M, Burek K, Hertel J, Troll M, et al. Night shift work affects urine metabolite profiles of nurses with early chronotype.

McHill AW, Melanson EL, Higgins J, Connick E, Moehlman TM, Stothard ER, et al. Impact of circadian misalignment on energy metabolism during simulated nightshift work. Folkard S. Do permanent night workers show circadian adjustment? A review based on the endogenous melatonin rhythm.

Stenvers DJ, Jonkers CF, Fliers E, Bisschop PH, Kalsbeek A. Knockout of Pgc1a leads to abnormal diurnal rhythms of activity, body temperature, and metabolic rate, in addition to aberrant expression of clock and metabolic genes Interestingly, SIRT1 suppresses PPARγ 63 but activates PGC-1α 64 , and thus affects the clock network through multiple mechanisms.

Possible molecular integrators of circadian and metabolic systems in the CNS. Molecular analyses of interplay between circadian and metabolic pathways have primarily emerged from studies in liver and other peripheral tissues, yet it remains uncertain whether similar mechanisms might also couple these processes within the brain.

One question is whether specific metabolites that vary according to time of day and nutrient state fasting vs. feeding , may also affect circadian function of energy-sensing neurons.

For instance, hour oscillation in levels of glucose and FAs may in turn influence expression of circadian genes and rhythmic transcriptional outputs within hypothalamic neurons involved in glucose homeostasis 65 , food intake 66 , and energy expenditure 67 — Perturbation of metabolic homeostasis with a high-fat diet is sufficient to alter both period length and amplitude of locomotor activity Additional nutrient factors that may participate in circadian oscillations of SCN and extra-SCN neurons include AMPK, SIRT1, and the mammalian target of rapamycin mTOR.

Hypothalamic AMPK is regulated by nutrient state and hormones such as leptin and insulin 72 , and manipulation of its expression alters food intake and body weight 73 ; however the impact of AMPK on CNS control of locomotor behavior and physiological rhythms is still not known. NPY and POMC neuron-specific knockout of the α2 subunit of AMPK results in lean and obese phenotypes, respectively, so it will be interesting to learn whether AMPK also participates in synchronizing activity and feeding through actions within these cell groups A final potential mediator involved in both energy sensing and circadian function in the CNS is mTOR, a regulator of protein synthesis present in ARC neurons that has been shown to modulate food intake mTOR is also expressed within pacemaker neurons of the SCN, where its expression is activated by light Moreover, activation of mTOR causes phase resetting of SCN explants 79 , while inhibition of mTOR alters light induction of the Period gene within the SCN of the intact animal Thus amino acid metabolism may participate in both entrainment of the master clock and in the temporal organization of feeding.

Indeed, different zeitgebers may exert differing effects on circadian entrainment in the brain and peripheral tissues, leading to distinct effects on phase and amplitude of gene oscillation within these locales.

For instance, glucocorticoid secretion from adrenal glands in response to adrenocorticotropin is dependent on time of day In addition to metabolite and hormonal input into the clock, recent studies by Buhr et al. suggest that temperature and heat shock signaling pathways play a central role in entrainment of peripheral tissues A major objective in future research will thus be to delineate the role of nutrient signaling in entrainment of CNS and peripheral clocks, and to determine how these signals interact with synchronizing signals such as feeding and neuroendocrine hormones e.

Evidence for circadian integration of energetics, metabolism, and sleep in humans. In humans, many aspects of metabolism display circadian cycles, including hour variation of glucose, insulin, and leptin levels 82 , Genome-wide association studies have also suggested connections between clock gene variation and fasting glucose levels 84 , 85 , obesity, and metabolic syndrome 86 , raising interest in understanding the impact of circadian systems on human disease.

One common clinical condition suggestive of interactions between circadian rhythms and metabolism in humans is that of shift work.

Numerous reports have indicated that shift workers have a higher incidence of diabetes, obesity, and cardiovascular events 1 , 87 , although the mechanism underlying this association is uncertain. Scheer et al.

recently tested the impact of forced circadian misalignment a simulation of shift work on neuroendocrine control of glucose metabolism and energetics In participants subjected to circadian misalignment, the investigators observed hypoleptinemia, insulin resistance, inverted cortisol rhythms, and increased blood pressure It is also interesting to note that patients with diabetes exhibit dampened amplitude of rhythms of glucose tolerance and insulin secretion 89 ; thus the relationship between circadian disruption and metabolic pathologies appears to be bidirectional in humans, suggesting that circadian disruption may lead to a vicious cycle and contribute to augmentation and progression of metabolic disease.

Direct genetic evidence in humans has linked the molecular clock with sleep 90 , 91 through the positional cloning of mutations causing familial advanced sleep phase syndrome, which is characterized by early sleep onset and awakening In the general population, observational studies have also found that short sleep, sleep deprivation, and poor sleep quality are associated with diabetes, metabolic syndrome 82 , 93 , hypoleptinemia, increased appetite, and obesity 94 , A recent study showed that sleep duration correlates with the magnitude of weight loss as fat in response to caloric restriction; short sleepers appear to have more difficulty losing fat compared to long sleepers despite similar amount of weight loss Narcolepsy, a sleep disorder in which patients present with extreme daytime sleepiness due to loss of hypocretin-producing neurons 97 , 98 , has been associated with elevated BMI 99 and increased incidence of obesity , Night eating syndrome NES is another instance in which disrupted rhythmic patterns of sleep and eating correlate with altered metabolism and obesity Patients with NES consume significantly more of their daily energy intake at night, although their total daily food intake is similar to that of control subjects , They also have abnormal rhythms of metabolic hormones, including decreased nocturnal rise in leptin, a phase shift in insulin, cortisol, and ghrelin, and inverted hour rhythms of blood glucose , Interestingly, the nocturnal pattern of eating observed in NES patients is reminiscent of feeding alterations in the Clock mutant mouse.

These animals exhibit increased feeding during the normal sleep period together with increased susceptibility to diet-induced obesity A major goal is to determine whether the adverse metabolic consequences of sleep loss and accompanying feeding alterations are due to the disrupted circadian rhythms per se, to the altered sleep, or to some combination of the two.

Integration of circadian rhythms and energy homeostasis in animal models. Studies in rodents have also attempted to experimentally simulate shift work in order to further probe mechanisms linking circadian disruption with metabolic disorders.

When rats were exposed to a daily eight-hour activity schedule during their normal resting phase, they succumbed to diminished rhythms of glucose and locomotor activity, as well as obesity, which correlated with increased food intake during their resting phase However, shifting food intake back to the active phase restored their metabolic rhythms and prevented obesity in these same animals , suggesting that the normal alignment of feeding and activity with the environment light cycle is critical for the maintenance of energy homeostasis.

Furthermore, feeding wild-type mice ad libitum exclusively during the daytime resulted in greater weight gain than in animals fed exclusively at night Similarly, when genetically obese mice with disrupted diurnal feeding patterns were fed exclusively at night, their obesity and metabolic disorders improved Genetic animal models of clock gene disruption have provided an additional approach to specifically dissect the effects of molecular clock genes on energy homeostasis.

For example, Clock mutant mice are obese and hyperphagic , although this phenotype may be modified by genetic background In contrast, Clock mutants have increased locomotor activity and food intake during the period when wild-type mice are at rest and fasting , suggesting non-redundant functions between the Clock and Npas2 paralogs with regard to energy balance.

Global Bmal1 knockout mice are arrhythmic and display increased adiposity at early ages ; however, these animals also develop arthropathy and myopathy, resulting in decreased activity and weight Interestingly, brain rescue of Bmal1 -knockout mice only restores behavioral circadian rhythms, whereas muscle rescue restores activity levels and body weight, but not behavioral rhythms , , suggesting that BMAL1 function within the brain controls period length and activity rhythms, whereas its expression in muscle affects mitochondrial function and exercise tolerance Curiously, mice with mutations in the Period genes display increased adiposity Future application of neuron-specific targeting approaches will be important to identify selective effects of circadian gene disruption on the interrelation between sleep and energy homeostasis.

Integration of circadian rhythms and glucose homeostasis in animal models. Glucose homeostasis is under circadian control at the level of both peripheral tissues and the SCN.

Ad libitum glucose levels peak and glucose tolerance is enhanced at the beginning of the active phase compared with the rest phase , The morning peak of glucose is postulated to result from increased hepatic gluconeogenesis as well as low insulin secretion , whereas the improved glucose tolerance early in the active period is likely due to elevated glucose uptake at skeletal muscle and adipose tissues , Genetic disruption of components of the clock network has identified a role of core clock genes in glucose homeostasis.

For example, multi-tissue Clock mutant mice develop age-dependent hyperglycemia and hypoinsulinemia , , in part due to impaired insulin secretion and defects in proliferation of pancreatic islets. Cry -knockout animals have increased hepatic gluconeogenesis, in part due to upregulation of cAMP signaling A major advance toward understanding the tissue-specific roles of peripheral clocks in glucose homeostasis came with the development of conditional gene-targeting approaches.

While deletion of Bmal1 within liver causes hypoglycemia , deletion of Bmal1 within pancreas causes hyperglycemia and hypoinsulinemia ; thus at the physiological level, the actions of clock activator genes in the liver oppose those in the pancreas.

Interestingly, both Clock mutant and Bmal1 -knockout mice are hypersensitive to insulin, although the mechanism for this remains to be defined While the above genetic models demonstrate a role for peripheral clocks in regulation of glucose metabolism, the SCN also plays a critical role in controlling the circadian variation of glucose and glucose tolerance , in part through rhythmic modulation of autonomic nervous system efferents Integration of circadian rhythms and lipid homeostasis in animal models.

In addition to glucoregulatory pathways, the circadian system also regulates lipid homeostasis and adipose tissue metabolism.

Both intestinal lipid transport and de novo lipid synthesis exhibit circadian variation , as do levels of adipose tissue hormones such as adiponectin and leptin Hypertriglyceridemia is evident in Clock mutant mice , likely due to both intestinal overabsorption and hepatic overproduction One node of coupling circadian and lipogenic pathways involves REV-ERBα REV-ERBα controls SREBP signaling and bile acid homeostasis, both of which are essential for lipid metabolism Furthermore, BMAL1 is necessary for adipogenesis, as embryonic fibroblasts from Bmal1 knockout mice fail to differentiate into adipocytes Clock genes may also indirectly regulate adipogenesis via PPARs.

Finally, the SCN has also been shown to be critical for regulation of the diurnality of leptin release , Further studies are necessary to assess the potential impact of clock genes on additional functions of the adipocytes, including thermogenesis and lipokine secretion.

Reciprocal effects of nutrient signaling on circadian rhythms and sleep. The relationship between circadian rhythms and metabolism is bidirectional; as noted above, a high-fat diet lengthens the intrinsic period length of locomotor activity, alters the circadian rhythms of feeding, reduces the amplitude, and shifts the phase of metabolic gene expression cycles in liver, fat, and hypothalamus In addition, glucocorticoid, a metabolic hormone involved in numerous biological processes such as gluconeogenesis, also entrains peripheral clocks 40 and increases expression of multiple clock genes, including Per1 and Per2 Restricted feeding studies have further suggested that nutrient conditions influence rhythmic locomotor activity and peripheral clock gene entrainment, although the role of clock genes per se in activity rhythms remains controversial.

Whereas the period of locomotor activity programmed by the SCN remains constant even when food is restricted each day to the light period, food-restricted animals display an anticipatory increase in locomotor activity , temperature, and glucocorticoid secretion that persists even when the food is removed Moreover, food availability can entrain rhythms in peripheral tissues such as liver and kidney, but not in the SCN 37 , 38 , suggesting the existence of a food-entrainable oscillator FEO The anatomic location of the FEO has been a topic of intensive investigation, although efforts to understand the mechanism for this process have produced inconclusive results.

While several studies have suggested that the DMH is necessary and sufficient to induce food-entrainable circadian rhythms , , other studies have questioned the role of the DMH as the FEO An important remaining question concerns the identity of the synchronizing signals that adjust circadian oscillations according to feeding time.

Potential signals involved in resetting peripheral clocks are numerous, including glucose, lipids, sterols, peptidergic molecules, and catecholamines.

Elucidating mechanisms involved in circadian entrainment of feeding may lead to improved interventions for clinical pathologies associated with shift work and jet lag. Advances in genetic studies of circadian rhythms have led to the recognition that the circadian system is tightly coupled with processes controlling both sleep and metabolism.

These dynamic interactions ensure that energy metabolism is coordinated in a proper temporal pattern and that circadian control is also subject to modulation by the energy status of the organism. Disruption of either the circadian clock or metabolism can lead to derangement of the other, thus predisposing to metabolic disorders such as obesity and type 2 diabetes.

Future research will continue to focus on expanding our understanding of how brain and peripheral clocks coordinately regulate metabolic processes at both the cell-autonomous and non-autonomous level, how nutrient flux translates information regarding environmental milieu to the clock, and the impact of circadian rhythms in human health and disease.

We thank Ravi Allada, Shin-ichiro Imai, Joseph S.

What is our circadian system, and how does it relate to glucose metabolism? In this study, however, the samples were taken only at three timepoints within an 8-h period, which was insufficient to extrapolate h oscillations. Importantly, while sleep deprivation alone induces insulin resistance in controlled laboratory experiments, circadian misalignment causes additive impairment of insulin sensitivity even with sleep time kept constant at a meager 5 h Circadian variation of the human metabolome captured by real-time breath analysis. Download references. Sookoian S , Gemma C , Gianotti TF , Burgueño A , Castaño G , Pirola CJ. Published in Volume , Issue 15 on August 2, J Clin Invest.
Circadian Rhythms and Metabolism Aviram and G. View this article via: Caffeine and cholesterol levels PubMed CrossRef Ckrcadian Scholar. Whitmore DCircadian rhythm metabolism NMegabolism CFoulkes NSPando MPTravnickova ZSassone-Corsi P A clockwork organ. Asher GSchibler U. One of the most important factors affecting the basal metabolic rate is the sleep pattern.

Circadian rhythm metabolism -

Gooley JJ , Lu J , Chou TC , Scammell TE , Saper CB Melanopsin in cells of origin of the retinohypothalamic tract. Nat Neurosci 4 : Lucas RJ , Freedman MS , Lupi D , Munoz M , David-Gray ZK , Foster RG Identifying the photoreceptive inputs to the mammalian circadian system using transgenic and retinally degenerate mice.

Behav Brain Res : 97 — Harmar AJ , Marston HM , Shen S , Spratt C , West KM , Sheward WJ , Morrison CF , Dorin JR , Piggins HD , Reubi JC , Kelly JS , Maywood ES , Hastings MH The VPAC 2 receptor is essential for circadian function in the mouse suprachiasmatic nuclei.

Maywood ES , Reddy AB , Wong GK , O'Neill JS , O'Brien JA , McMahon DG , Harmar AJ , Okamura H , Hastings MH Synchronization and maintenance of timekeeping in suprachiasmatic circadian clock cells by neuropeptidergic signaling.

Curr Biol 16 : — Le Minh N , Damiola F , Tronche F , Schütz G , Schibler U Glucocorticoid hormones inhibit food-induced phase-shifting of peripheral circadian oscillators.

EMBO J 20 : — Kramer A , Yang FC , Snodgrass P , Li X , Scammell TE , Davis FC , Weitz CJ Regulation of daily locomotor activity and sleep by hypothalamic EGF receptor signaling.

Science : — Cheng MY , Bullock CM , Li C , Lee AG , Bermak JC , Belluzzi J , Weaver DR , Leslie FM , Zhou QY Prokineticin 2 transmits the behavioural circadian rhythm of the suprachiasmatic nucleus.

Kraves S , Weitz CJ A role for cardiotrophin-like cytokine in the circadian control of mammalian locomotor activity. Nat Neurosci 9 : — Saeb-Parsy K , Lombardelli S , Khan FZ , McDowall K , Au-Yong IT , Dyball RE Neural connections of hypothalamic neuroendocrine nuclei in the rat.

J Neuroendocrinol 12 : — Proc Natl Acad Sci USA : — Welsh DK , Yoo SH , Liu AC , Takahashi JS , Kay SA Bioluminescence imaging of individual fibroblasts reveals persistent, independently phased circadian rhythms of clock gene expression.

Curr Biol 14 : — Matsumoto S , Basil J , Jetton AE , Lehman MN , Bittman EL Regulation of the phase and period of circadian rhythms restored by suprachiasmatic transplants. J Biol Rhythms 11 : — Froy O , Chapnik N , Miskin R Long-lived αMUPA transgenic mice exhibit pronounced circadian rhythms.

Am J Physiol Endocrinol Metab : E — E Schibler U , Ripperger J , Brown SA Peripheral circadian oscillators in mammals: time and food. J Biol Rhythms 18 : — Dunlap JC Molecular bases for circadian clocks. Cell 96 : — Cardone L , Hirayama J , Giordano F , Tamaru T , Palvimo JJ , Sassone-Corsi P Circadian clock control by SUMOylation of BMAL1.

Vitaterna MH , King DP , Chang AM , Kornhauser JM , Lowrey PL , McDonald JD , Dove WF , Pinto LH , Turek FW , Takahashi JS Mutagenesis and mapping of a mouse gene, Clock , essential for circadian behavior.

Asher G , Schibler U A CLOCK-less clock. Trends Cell Biol 16 : — Debruyne JP , Noton E , Lambert CM , Maywood ES , Weaver DR , Reppert SM A clock shock: mouse CLOCK is not required for circadian oscillator function.

Neuron 50 : — Zylka MJ , Shearman LP , Weaver DR , Reppert SM Three Period homologs in mammals: differential light responses in the suprachiasmatic circadian clock and oscillating transcripts outside of brain.

Neuron 20 : — Froy O , Chang DC , Reppert SM Redox potential: differential roles in dCRY and mCRY1 functions. Curr Biol 12 : — Doi M , Hirayama J , Sassone-Corsi P Circadian regulator CLOCK is a histone acetyltransferase. Nakahata Y , Grimaldi B , Sahar S , Hirayama J , Sassone-Corsi P Signaling to the circadian clock: plasticity by chromatin remodeling.

Curr Opin Cell Biol 19 : — Etchegaray JP , Lee C , Wade PA , Reppert SM Rhythmic histone acetylation underlies transcription in the mammalian circadian clock.

Curtis AM , Seo SB , Westgate EJ , Rudic RD , Smyth EM , Chakravarti D , FitzGerald GA , McNamara P Histone acetyltransferase-dependent chromatin remodeling and the vascular clock. J Biol Chem : — Naruse Y , Oh-hashi K , Iijima N , Naruse M , Yoshioka H , Tanaka M Circadian and light-induced transcription of clock gene Per1 depends on histone acetylation and deacetylation.

Mol Cell Biol 24 : — Ripperger JA , Schibler U Rhythmic CLOCK-BMAL1 binding to multiple E-box motifs drives circadian Dbp transcription and chromatin transitions. Nat Genet 38 : — Etchegaray JP , Yang X , DeBruyne JP , Peters AH , Weaver DR , Jenuwein T , Reppert SM The polycomb group protein EZH2 is required for mammalian circadian clock function.

Whitmore D , Cermakian N , Crosio C , Foulkes NS , Pando MP , Travnickova Z , Sassone-Corsi P A clockwork organ. Biol Chem : — Eide EJ , Virshup DM Casein kinase I: another cog in the circadian clockworks.

Chronobiol Int 18 : — Eide EJ , Woolf MF , Kang H , Woolf P , Hurst W , Camacho F , Vielhaber EL , Giovanni A , Virshup DM Control of mammalian circadian rhythm by CKIε-regulated proteasome-mediated PER2 degradation.

Mol Cell Biol 25 : — Eide EJ , Kang H , Crapo S , Gallego M , Virshup DM Casein kinase I in the mammalian circadian clock. Methods Enzymol : — Preitner N , Damiola F , Lopez-Molina L , Zakany J , Duboule D , Albrecht U , Schibler U The orphan nuclear receptor REV-ERBα controls circadian transcription within the positive limb of the mammalian circadian oscillator.

Mol Endocrinol 19 : — Sato TK , Panda S , Miraglia LJ , Reyes TM , Rudic RD , McNamara P , Naik KA , FitzGerald GA , Kay SA , Hogenesch JB A functional genomics strategy reveals Rora as a component of the mammalian circadian clock. Neuron 43 : — Ueda HR , Hayashi S , Chen W , Sano M , Machida M , Shigeyoshi Y , Iino M , Hashimoto S System-level identification of transcriptional circuits underlying mammalian circadian clocks.

Nat Genet 37 : — Kornmann B , Preitner N , Rifat D , Fleury-Olela F , Schibler U Analysis of circadian liver gene expression by ADDER, a highly sensitive method for the display of differentially expressed mRNAs. Nucleic Acids Res 29 : E51 — 1. Akhtar RA , Reddy AB , Maywood ES , Clayton JD , King VM , Smith AG , Gant TW , Hastings MH , Kyriacou CP Circadian cycling of the mouse liver transcriptome, as revealed by cDNA microarray, is driven by the suprachiasmatic nucleus.

Duffield GE , Best JD , Meurers BH , Bittner A , Loros JJ , Dunlap JC Circadian programs of transcriptional activation, signaling, and protein turnover revealed by microarray analysis of mammalian cells. Storch KF , Lipan O , Leykin I , Viswanathan N , Davis FC , Wong WH , Weitz CJ Extensive and divergent circadian gene expression in liver and heart.

Nature : 78 — Kita Y , Shiozawa M , Jin W , Majewski RR , Besharse JC , Greene AS , Jacob HJ Implications of circadian gene expression in kidney, liver and the effects of fasting on pharmacogenomic studies.

Pharmacogenetics 12 : 55 — Zvonic S , Ptitsyn AA , Conrad SA , Scott LK , Floyd ZE , Kilroy G , Wu X , Goh BC , Mynatt RL , Gimble JM Characterization of peripheral circadian clocks in adipose tissues.

Diabetes 55 : — Reddy AB , Karp NA , Maywood ES , Sage EA , Deery M , O'Neill JS , Wong GK , Chesham J , Odell M , Lilley KS , Kyriacou CP , Hastings MH Circadian orchestration of the hepatic proteome. McCarthy JJ , Andrews JL , McDearmon EL , Campbell KS , Barber BK , Miller BH , Walker JR , Hogenesch JB , Takahashi JS , Esser KA Identification of the circadian transcriptome in adult mouse skeletal muscle.

Physiol Genomics 31 : 86 — Kornmann B , Schaad O , Bujard H , Takahashi JS , Schibler U System-driven and oscillator-dependent circadian transcription in mice with a conditionally active liver clock. PLoS Biol 5 : e Froy O , Chapnik N , Miskin R The suprachiasmatic nuclei are involved in determining circadian rhythms during restricted feeding.

Neuroscience : — Saper CB , Lu J , Chou TC , Gooley J The hypothalamic integrator for circadian rhythms. Trends Neurosci 28 : — Saper CB , Scammell TE , Lu J Hypothalamic regulation of sleep and circadian rhythms.

Yi CX , van der Vliet J , Dai J , Yin G , Ru L , Buijs RM Ventromedial arcuate nucleus communicates peripheral metabolic information to the suprachiasmatic nucleus. Endocrinology : — Chou TC , Scammell TE , Gooley JJ , Gaus SE , Saper CB , Lu J Critical role of dorsomedial hypothalamic nucleus in a wide range of behavioral circadian rhythms.

Lu J , Zhang YH , Chou TC , Gaus SE , Elmquist JK , Shiromani P , Saper CB Contrasting effects of ibotenate lesions of the paraventricular nucleus and subparaventricular zone on sleep-wake cycle and temperature regulation.

J Neurosci 21 : — Ramsey KM , Marcheva B , Kohsaka A , Bass J The clockwork of metabolism. Annu Rev Nutr 27 : — Buijs RM , Wortel J , Van Heerikhuize JJ , Feenstra MG , Ter Horst GJ , Romijn HJ , Kalsbeek A Anatomical and functional demonstration of a multisynaptic suprachiasmatic nucleus adrenal cortex pathway.

Eur J Neurosci 11 : — Buijs RM , Scheer FA , Kreier F , Yi C , Bos N , Goncharuk VD , Kalsbeek A Organization of circadian functions: interaction with the body. Prog Brain Res : — Roland BL , Sawchenko PE Local origins of some GABAergic projections to the paraventricular and supraoptic nuclei of the hypothalamus in the rat.

J Comp Neurol : — Shekhar A , Katner JS Dorsomedial hypothalamic GABA regulates anxiety in the social interaction test. Pharmacol Biochem Behav 50 : — Gamble KL , Allen GC , Zhou T , McMahon DG Gastrin-releasing peptide mediates light-like resetting of the suprachiasmatic nucleus circadian pacemaker through cAMP response element-binding protein and Per1 activation.

J Neurosci 27 : — Akanmu MA , Ukponmwan OE , Katayama Y , Honda K Neuropeptide-Y Y2-receptor agonist, PYY3—36 promotes non-rapid eye movement sleep in rat.

Neurosci Res 54 : — Proc Soc Exp Biol Med 77 : — Fan W , Boston BA , Kesterson RA , Hruby VJ , Cone RD Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Huszar D , Lynch CA , Fairchild-Huntress V , Dunmore JH , Fang Q , Berkemeier LR , Gu W , Kesterson RA , Boston BA , Cone RD , Smith FJ , Campfield LA , Burn P , Lee F Targeted disruption of the melanocortin-4 receptor results in obesity in mice.

Cell 88 : — Adan RA , Cone RD , Burbach JP , Gispen WH Differential effects of melanocortin peptides on neural melanocortin receptors.

Mol Pharmacol 46 : — Cone RD Anatomy and regulation of the central melanocortin system. Nat Neurosci 8 : — Krude H , Biebermann H , Luck W , Horn R , Brabant G , Grüters A Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans.

Nat Genet 19 : — Yeo GS , Farooqi IS , Aminian S , Halsall DJ , Stanhope RG , O'Rahilly S A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nat Genet 20 : — Vaisse C , Clement K , Guy-Grand B , Froguel P A frameshift mutation in human MC4R is associated with a dominant form of obesity.

Hinney A , Schmidt A , Nottebom K , Heibült O , Becker I , Ziegler A , Gerber G , Sina M , Görg T , Mayer H , Siegfried W , Fichter M , Remschmidt H , Hebebrand J Several mutations in the melanocortin-4 receptor gene including a nonsense and a frameshift mutation associated with dominantly inherited obesity in humans.

J Clin Endocrinol Metab 84 : — Farooqi IS , Yeo GS , Keogh JM , Aminian S , Jebb SA , Butler G , Cheetham T , O'Rahilly S Dominant and recessive inheritance of morbid obesity associated with melanocortin 4 receptor deficiency. J Clin Invest : — Vaisse C , Clement K , Durand E , Hercberg S , Guy-Grand B , Froguel P Melanocortin-4 receptor mutations are a frequent and heterogeneous cause of morbid obesity.

Ollmann MM , Wilson BD , Yang YK , Kerns JA , Chen Y , Gantz I , Barsh GS Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Quillan JM , Sadée W , Wei ET , Jimenez C , Ji L , Chang JK A synthetic human Agouti-related protein- 83— -NH2 fragment is a potent inhibitor of melanocortin receptor function.

FEBS Lett : 59 — Rossi M , Kim MS , Morgan DG , Small CJ , Edwards CM , Sunter D , Abusnana S , Goldstone AP , Russell SH , Stanley SA , Smith DM , Yagaloff K , Ghatei MA , Bloom SR A C-terminal fragment of Agouti-related protein increases feeding and antagonizes the effect of α-melanocyte stimulating hormone in vivo.

Baskin DG , Breininger JF , Schwartz MW Leptin receptor mRNA identifies a subpopulation of neuropeptide Y neurons activated by fasting in rat hypothalamus.

Diabetes 48 : — Elias CF , Aschkenasi C , Lee C , Kelly J , Ahima RS , Bjorbaek C , Flier JS , Saper CB , Elmquist JK Leptin differentially regulates NPY and POMC neurons projecting to the lateral hypothalamic area.

Neuron 23 : — Elias CF , Kelly JF , Lee CE , Ahima RS , Drucker DJ , Saper CB , Elmquist JK Chemical characterization of leptin-activated neurons in the rat brain. Marsh DJ , Miura GI , Yagaloff KA , Schwartz MW , Barsh GS , Palmiter RD Effects of neuropeptide Y deficiency on hypothalamic agouti-related protein expression and responsiveness to melanocortin analogues.

Brain Res : 66 — Stephens TW , Basinski M , Bristow PK , Bue-Valleskey JM , Burgett SG , Craft L , Hale J , Hoffmann J , Hsiung HM , Kriauciunas A , MacKellar W , Rosteck Jr PR , Schoner B , Smith D , Tinsley FC , Zhang XY , Heiman M The role of neuropeptide Y in the antiobesity action of the obese gene product.

Guan XM , Hess JF , Yu H , Hey PJ , van der Ploeg LH Differential expression of mRNA for leptin receptor isoforms in the rat brain. Mol Cell Endocrinol : 1 — 7. Zigman JM , Jones JE , Lee CE , Saper CB , Elmquist JK Expression of ghrelin receptor mRNA in the rat and the mouse brain.

Yi CX , Challet E , Pévet P , Kalsbeek A , Escobar C , Buijs RM A circulating ghrelin mimetic attenuates light-induced phase delay of mice and light-induced Fos expression in the suprachiasmatic nucleus of rats.

Eur J Neurosci 27 : — Yannielli PC , Molyneux PC , Harrington ME , Golombek DA Ghrelin effects on the circadian system of mice. Kalra SP , Dube MG , Pu S , Xu B , Horvath TL , Kalra PS Interacting appetite-regulating pathways in the hypothalamic regulation of body weight.

Endocr Rev 20 : 68 — Prog Neurobiol 74 : 59 — Challet E Minireview: entrainment of the suprachiasmatic clockwork in diurnal and nocturnal mammals. Sakkou M , Wiedmer P , Anlag K , Hamm A , Seuntjens E , Ettwiller L , Tschöp MH , Treier M A role for brain-specific homeobox factor Bsx in the control of hyperphagia and locomotory behavior.

Cell Metab 5 : — Cowley MA , Smart JL , Rubinstein M , Cerdán MG , Diano S , Horvath TL , Cone RD , Low MJ Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus.

Elmquist JK , Ahima RS , Elias CF , Flier JS , Saper CB Leptin activates distinct projections from the dorsomedial and ventromedial hypothalamic nuclei.

Proc Natl Acad Sci USA 95 : — Elmquist JK , Elias CF , Saper CB From lesions to leptin: hypothalamic control of food intake and body weight. Neuron 22 : — Flier JS , Maratos-Flier E Obesity and the hypothalamus: novel peptides for new pathways. Cell 92 : — Friedman JM , Halaas JL Leptin and the regulation of body weight in mammals.

Schwartz MW , Woods SC , Porte Jr D , Seeley RJ , Baskin DG Central nervous system control of food intake. Shimada M , Tritos NA , Lowell BB , Flier JS , Maratos-Flier E Mice lacking melanin-concentrating hormone are hypophagic and lean. Sutcliffe JG , de Lecea L The hypocretins: excitatory neuromodulatory peptides for multiple homeostatic systems, including sleep and feeding.

J Neurosci Res 62 : — Willie JT , Chemelli RM , Sinton CM , Yanagisawa M To eat or to sleep? Orexin in the regulation of feeding and wakefulness. Annu Rev Neurosci 24 : — Turek FW , Joshu C , Kohsaka A , Lin E , Ivanova G , McDearmon E , Laposky A , Losee-Olson S , Easton A , Jensen DR , Eckel RH , Takahashi JS , Bass J Obesity and metabolic syndrome in circadian Clock mutant mice.

Trends Endocrinol Metab 11 : — Lin L , Faraco J , Li R , Kadotani H , Rogers W , Lin X , Qiu X , de Jong PJ , Nishino S , Mignot E The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin orexin receptor 2 gene. Cell 98 : — Hara J , Beuckmann CT , Nambu T , Willie JT , Chemelli RM , Sinton CM , Sugiyama F , Yagami K , Goto K , Yanagisawa M , Sakurai T Genetic ablation of orexin neurons in mice results in narcolepsy, hypophagia, and obesity.

Neuron 30 : — Nishino S , Mignot E Article reviewed: Plasma orexin-A is lower in patients with narcolepsy. Sleep Med 3 : — Peptides 27 : — La Fleur SE , Kalsbeek A , Wortel J , Buijs RM A suprachiasmatic nucleus generated rhythm in basal glucose concentrations.

J Neuroendocrinol 11 : — Ruiter M , La Fleur SE , van Heijningen C , van der Vliet J , Kalsbeek A , Buijs RM The daily rhythm in plasma glucagon concentrations in the rat is modulated by the biological clock and by feeding behavior.

Diabetes 52 : — Ando H , Yanagihara H , Hayashi Y , Obi Y , Tsuruoka S , Takamura T , Kaneko S , Fujimura A Rhythmic messenger ribonucleic acid expression of clock genes and adipocytokines in mouse visceral adipose tissue. De Boer SF , Van der Gugten J Daily variations in plasma noradrenaline, adrenaline and corticosterone concentrations in rats.

Physiol Behav 40 : — Ahima RS , Prabakaran D , Flier JS Postnatal leptin surge and regulation of circadian rhythm of leptin by feeding. Implications for energy homeostasis and neuroendocrine function. Bodosi B , Gardi J , Hajdu I , Szentirmai E , Obal Jr F , Krueger JM Rhythms of ghrelin, leptin, and sleep in rats: effects of the normal diurnal cycle, restricted feeding, and sleep deprivation.

Am J Physiol Regul Integr Comp Physiol : R — R Kalra SP , Bagnasco M , Otukonyong EE , Dube MG , Kalra PS Rhythmic, reciprocal ghrelin and leptin signaling: new insight in the development of obesity. Regul Pept : 1 — Kalsbeek A , Fliers E , Romijn JA , La Fleur SE , Wortel J , Bakker O , Endert E , Buijs RM The suprachiasmatic nucleus generates the diurnal changes in plasma leptin levels.

Shen J , Tanida M , Niijima A , Nagai K In vivo effects of leptin on autonomic nerve activity and lipolysis in rats. Neurosci Lett : — Froy O The relationship between nutrition and circadian rhythms in mammals. Front Neuroendocrinol 28 : 61 — Green CB , Takahashi JS , Bass J The meter of metabolism.

Hirota T , Fukada Y Resetting mechanism of central and peripheral circadian clocks in mammals. Zoolog Sci 21 : — Kohsaka A , Bass J A sense of time: how molecular clocks organize metabolism.

Trends Endocrinol Metab 18 : 4 — La Fleur SE Daily rhythms in glucose metabolism: suprachiasmatic nucleus output to peripheral tissue. J Neuroendocrinol 15 : — Davidson AJ , Castañón-Cervantes O , Stephan FK Daily oscillations in liver function: diurnal vs circadian rhythmicity.

Liver Int 24 : — Froy O Cytochrome p and the biological clock in mammals. Curr Drug Metab 10 : — Frederiks WM , Marx F , Bosch KS Diurnal variation in glycogen phosphorylase activity in rat liver.

A quantitative histochemical study. Eur J Cell Biol 43 : — Ximenes da Silva A , Gendrot G , Servière J , Lavialle M Daily changes of cytochrome oxidase activity within the suprachiasmatic nucleus of the Syrian hamster.

Rivera-Coll A , Fuentes-Arderiu X , Díez-Noguera A Circadian rhythms of serum concentrations of 12 enzymes of clinical interest. Chronobiol Int 10 : — Fukuda H , Iritani N Diurnal variations of lipogenic enzyme mRNA quantities in rat liver.

Biochim Biophys Acta : — Davies SP , Carling D , Munday MR , Hardie DG Diurnal rhythm of phosphorylation of rat liver acetyl-CoA carboxylase by the AMP-activated protein kinase, demonstrated using freeze-clamping.

Effects of high-fat diets. Eur J Biochem : — Cailotto C , La Fleur SE , Van Heijningen C , Wortel J , Kalsbeek A , Feenstra M , Pévet P , Buijs RM The suprachiasmatic nucleus controls the daily variation of plasma glucose via the autonomic output to the liver: are the clock genes involved?

Eur J Neurosci 22 : — Kalsbeek A , Ruiter M , La Fleur SE , Cailotto C , Kreier F , Buijs RM The hypothalamic clock and its control of glucose homeostasis. Yamazaki S , Ishida Y , Inouye S Circadian rhythms of adenosine triphosphate contents in the suprachiasmatic nucleus, anterior hypothalamic area and caudate putamen of the rat—negative correlation with electrical activity.

Brain Res : — Yang X , Downes M , Yu RT , Bookout AL , He W , Straume M , Mangelsdorf DJ , Evans RM Nuclear receptor expression links the circadian clock to metabolism.

Stephan FK , Davidson AJ Glucose, but not fat, phase shifts the feeding-entrained circadian clock. Physiol Behav 65 : — Young ME , Wilson CR , Razeghi P , Guthrie PH , Taegtmeyer H Alterations of the circadian clock in the heart by streptozotocin-induced diabetes. J Mol Cell Cardiol 34 : — Hirota T , Okano T , Kokame K , Shirotani-Ikejima H , Miyata T , Fukada Y Glucose down-regulates Per1 and Per2 mRNA levels and induces circadian gene expression in cultured Rat-1 fibroblasts.

Iwanaga H , Yano M , Miki H , Okada K , Azama T , Takiguchi S , Fujiwara Y , Yasuda T , Nakayama M , Kobayashi M , Oishi K , Ishida N , Nagai K , Monden M Per2 gene expressions in the suprachiasmatic nucleus and liver differentially respond to nutrition factors in rats.

JPEN J Parenter Enteral Nutr 29 : — Mohri T , Emoto N , Nonaka H , Fukuya H , Yagita K , Okamura H , Yokoyama M Alterations of circadian expressions of clock genes in Dahl salt-sensitive rats fed a high-salt diet.

Hypertension 42 : — Waddington Lamont E , Harbour VL , Barry-Shaw J , Renteria Diaz L , Robinson B , Stewart J , Amir S Restricted access to food, but not sucrose, saccharine, or salt, synchronizes the expression of Period2 protein in the limbic forebrain. Chen CP , Kuhn P , Advis JP , Sarkar DK Chronic ethanol consumption impairs the circadian rhythm of pro-opiomelanocortin and period genes mRNA expression in the hypothalamus of the male rat.

J Neurochem 88 : — Alcohol Clin Exp Res 29 : — Antle MC , Steen NM , Mistlberger RE Adenosine and caffeine modulate circadian rhythms in the Syrian hamster.

Neuroreport 12 : — Langlais PJ , Hall T Thiamine deficiency-induced disruptions in the diurnal rhythm and regulation of body temperature in the rat. Metab Brain Dis 13 : — Bennett MR , Schwartz WJ Altered circadian rhythmicity is an early sign of murine dietary thiamine deficiency.

J Neurol Sci : 6 — McNamara P , Seo SB , Rudic RD , Sehgal A , Chakravarti D , FitzGerald GA Regulation of CLOCK and MOP4 by nuclear hormone receptors in the vasculature: a humoral mechanism to reset a peripheral clock.

Biochem Biophys Res Commun : — Balsalobre A , Marcacci L , Schibler U Multiple signaling pathways elicit circadian gene expression in cultured Rat-1 fibroblasts.

Curr Biol 10 : — Balsalobre A , Brown SA , Marcacci L , Tronche F , Kellendonk C , Reichardt HM , Schütz G , Schibler U Resetting of circadian time in peripheral tissues by glucocorticoid signaling.

Reddy AB , Maywood ES , Karp NA , King VM , Inoue Y , Gonzalez FJ , Lilley KS , Kyriacou CP , Hastings MH Glucocorticoid signaling synchronizes the liver circadian transcriptome.

Hepatology 45 : — Fu L , Patel MS , Bradley A , Wagner EF , Karsenty G The molecular clock mediates leptin-regulated bone formation.

Rutter J , Reick M , Wu LC , McKnight SL Regulation of clock and NPAS2 DNA binding by the redox state of NAD cofactors. Rutter J , Reick M , McKnight SL Metabolism and the control of circadian rhythms. Annu Rev Biochem 71 : — Vieira E , Nilsson EC , Nerstedt A , Ormestad M , Long YC , Garcia-Roves PM , Zierath JR , Mahlapuu M Relationship between AMPK and the transcriptional balance of clock-related genes in skeletal muscle.

Martin TL , Alquier T , Asakura K , Furukawa N , Preitner F , Kahn BB Diet-induced obesity alters AMP kinase activity in hypothalamus and skeletal muscle. Blander G , Guarente L The Sir2 family of protein deacetylases.

Annu Rev Biochem 73 : — Ann Med 39 : — Asher G , Gatfield D , Stratmann M , Reinke H , Dibner C , Kreppel F , Mostoslavsky R , Alt FW , Schibler U SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Hirayama J , Sahar S , Grimaldi B , Tamaru T , Takamatsu K , Nakahata Y , Sassone-Corsi P CLOCK-mediated acetylation of BMAL1 controls circadian function.

Belden WJ , Dunlap JC SIRT1 is a circadian deacetylase for core clock components. J Biol Rhythms 17 : — Cassone VM , Stephan FK Central and peripheral regulation of feeding and nutrition by the mammalian circadian clock: implications for nutrition during manned space flight.

Nutrition 18 : — Honma KI , Honma S , Hiroshige T Critical role of food amount for prefeeding corticosterone peak in rats. Grasl-Kraupp B , Bursch W , Ruttkay-Nedecky B , Wagner A , Lauer B , Schulte-Hermann R Food restriction eliminates preneoplastic cells through apoptosis and antagonizes carcinogenesis in rat liver.

Proc Natl Acad Sci USA 91 : — Saito M , Murakami E , Suda M Circadian rhythms in disaccharidases of rat small intestine and its relation to food intake. Comperatore CA , Stephan FK Entrainment of duodenal activity to periodic feeding.

J Biol Rhythms 2 : — Stephan FK , Swann JM , Sisk CL Anticipation of hr feeding schedules in rats with lesions of the suprachiasmatic nucleus. Behav Neural Biol 25 : — Mistlberger RE Circadian food-anticipatory activity: formal models and physiological mechanisms.

Neurosci Biobehav Rev 18 : — Hara R , Wan K , Wakamatsu H , Aida R , Moriya T , Akiyama M , Shibata S Restricted feeding entrains liver clock without participation of the suprachiasmatic nucleus. Genes Cells 6 : — Oishi K , Miyazaki K , Ishida N Functional CLOCK is not involved in the entrainment of peripheral clocks to the restricted feeding: entrainable expression of mPer2 and Bmal1 mRNAs in the heart of Clock mutant mice on Jcl:ICR background.

Horikawa K , Minami Y , Iijima M , Akiyama M , Shibata S Rapid damping of food-entrained circadian rhythm of clock gene expression in clock-defective peripheral tissues under fasting conditions.

Damiola F , Le Minh N , Preitner N , Kornmann B , Fleury-Olela F , Schibler U Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus.

Genes Dev 14 : — Stokkan KA , Yamazaki S , Tei H , Sakaki Y , Menaker M Entrainment of the circadian clock in the liver by feeding.

Lin JD , Liu C , Li S Integration of energy metabolism and the mammalian clock. Cell Cycle 7 : — Boulamery-Velly A , Simon N , Vidal J , Mouchet J , Bruguerolle B Effects of three-hour restricted food access during the light period on circadian rhythms of temperature, locomotor activity, and heart rate in rats.

Chronobiol Int 22 : — Hirao J , Arakawa S , Watanabe K , Ito K , Furukawa T Effects of restricted feeding on daily fluctuations of hepatic functions including p monooxygenase activities in rats.

Mieda M , Williams SC , Richardson JA , Tanaka K , Yanagisawa M The dorsomedial hypothalamic nucleus as a putative food-entrainable circadian pacemaker. Gooley JJ , Schomer A , Saper CB The dorsomedial hypothalamic nucleus is critical for the expression of food-entrainable circadian rhythms.

Landry GJ , Simon MM , Webb IC , Mistlberger RE Persistence of a behavioral food-anticipatory circadian rhythm following dorsomedial hypothalamic ablation in rats. Landry GJ , Yamakawa GR , Webb IC , Mear RJ , Mistlberger RE The dorsomedial hypothalamic nucleus is not necessary for the expression of circadian food-anticipatory activity in rats.

J Biol Rhythms 22 : — Davidson AJ , Cappendijk SL , Stephan FK Feeding-entrained circadian rhythms are attenuated by lesions of the parabrachial region in rats.

Mistlberger RE , Mumby DG The limbic system and food-anticipatory circadian rhythms in the rat: ablation and dopamine blocking studies. Behav Brain Res 47 : — Mendoza J , Angeles-Castellanos M , Escobar C Differential role of the accumbens Shell and Core subterritories in food-entrained rhythms of rats.

Behav Brain Res : — Davidson AJ Search for the feeding-entrainable circadian oscillator: a complex proposition. Comperatore CA , Stephan FK Effects of vagotomy on entrainment of activity rhythms to food access. Physiol Behav 47 : — Mistlberger RE , Marchant EG Enhanced food-anticipatory circadian rhythms in the genetically obese Zucker rat.

Physiol Behav 66 : — Am J Physiol Regul Integr Comp Physiol : R57 — R Pendergast JS , Nakamura W , Friday RC , Hatanaka F , Takumi T , Yamazaki S Robust food anticipatory activity in BMAL1-deficient mice. PLoS ONE 4 : e Storch KF , Weitz CJ Daily rhythms of food-anticipatory behavioral activity do not require the known circadian clock.

Feillet CA , Ripperger JA , Magnone MC , Dulloo A , Albrecht U , Challet E Lack of food anticipation in Per2 mutant mice. Mistlberger RE Circadian rhythms: perturbing a food-entrained clock.

Curr Biol 16 : R — R Masoro EJ , Shimokawa I , Higami Y , McMahan CA , Yu BP Temporal pattern food intake not a factor in the retardation of aging processes by dietary restriction.

J Gerontol A Biol Sci Med Sci 50A : B48 — B Koubova J , Guarente L How does calorie restriction work? Genes Dev 17 : — Masoro EJ Overview of caloric restriction and ageing. Mech Ageing Dev : — Weindruch R , Sohal RS Seminars in medicine of the Beth Israel Deaconess Medical Center.

Caloric intake and aging. N Engl J Med : — Roth GS , Lane MA , Ingram DK , Mattison JA , Elahi D , Tobin JD , Muller D , Metter EJ Biomarkers of caloric restriction may predict longevity in humans.

Science : Roth GS , Mattison JA , Ottinger MA , Chachich ME , Lane MA , Ingram DK Aging in rhesus monkeys: relevance to human health interventions.

Challet E , Solberg LC , Turek FW Entrainment in calorie-restricted mice: conflicting zeitgebers and free-running conditions. Challet E , Caldelas I , Graff C , Pévet P Synchronization of the molecular clockwork by light- and food-related cues in mammals.

J Neurosci 25 : — Resuehr D , Olcese J Caloric restriction and melatonin substitution: effects on murine circadian parameters. Froy O , Miskin R The interrelations among feeding, circadian rhythms and ageing. Prog Neurobiol 82 : — Anson RM , Guo Z , de Cabo R , Iyun T , Rios M , Hagepanos A , Ingram DK , Lane MA , Mattson MP Intermittent fasting dissociates beneficial effects of dietary restriction on glucose metabolism and neuronal resistance to injury from calorie intake.

Descamps O , Riondel J , Ducros V , Roussel AM Mitochondrial production of reactive oxygen species and incidence of age-associated lymphoma in OF1 mice: effect of alternate-day fasting. Goodrick CL , Ingram DK , Reynolds MA , Freeman JR , Cider N Effects of intermittent feeding upon body weight and lifespan in inbred mice: interaction of genotype and age.

Mech Ageing Dev 55 : 69 — Contestabile A , Ciani E , Contestabile A Dietary restriction differentially protects from neurodegeneration in animal models of excitotoxicity. Mattson MP Energy intake, meal frequency, and health: a neurobiological perspective.

Annu Rev Nutr 25 : — Sharma S , Kaur G Neuroprotective potential of dietary restriction against kainate-induced excitotoxicity in adult male Wistar rats. Brain Res Bull 67 : — Ahmet I , Wan R , Mattson MP , Lakatta EG , Talan M Cardioprotection by intermittent fasting in rats. Circulation : — Mager DE , Wan R , Brown M , Cheng A , Wareski P , Abernethy DR , Mattson MP Caloric restriction and intermittent fasting alter spectral measures of heart rate and blood pressure variability in rats.

FASEB J 20 : — Mattson MP , Duan W , Wan R , Guo Z Prophylactic activation of neuroprotective stress response pathways by dietary and behavioral manipulations. NeuroRx 1 : — Mattson MP Dietary factors, hormesis and health.

Ageing Res Rev 7 : 43 — Froy O , Chapnik N , Miskin R Effect of intermittent fasting on circadian rhythms in mice depends on feeding time. Bamshad M , Aoki VT , Adkison MG , Warren WS , Bartness TJ Central nervous system origins of the sympathetic nervous system outflow to white adipose tissue.

Bartness TJ , Bamshad M Innervation of mammalian white adipose tissue: implications for the regulation of total body fat. Cantu RC , Goodman HM Effects of denervation and fasting on white adipose tissue.

Am J Physiol : — Youngstrom TG , Bartness TJ White adipose tissue sympathetic nervous system denervation increases fat pad mass and fat cell number. Kreier F , Fliers E , Voshol PJ , Van Eden CG , Havekes LM , Kalsbeek A , Van Heijningen CL , Sluiter AA , Mettenleiter TC , Romijn JA , Sauerwein HP , Buijs RM Selective parasympathetic innervation of subcutaneous and intra-abdominal fat—functional implications.

Zvonic S , Floyd ZE , Mynatt RL , Gimble JM Circadian rhythms and the regulation of metabolic tissue function and energy homeostasis. Obesity Silver Spring 15 : — Loboda A , Kraft WK , Fine B , Joseph J , Nebozhyn M , Zhang C , He Y , Yang X , Wright C , Morris M , Chalikonda I , Ferguson M , Emilsson V , Leonardson A , Lamb J , Dai H , Schadt E , Greenberg HE , Lum PY Diurnal variation of the human adipose transcriptome and the link to metabolic disease.

BMC Med Genomics 2 : 7. Suzuki M , Shimomura Y , Satoh Y Diurnal changes in lipolytic activity of isolated fat cells and their increased responsiveness to epinephrine and theophylline with meal feeding in rats. J Nutr Sci Vitaminol Tokyo 29 : — Saad MF , Riad-Gabriel MG , Khan A , Sharma A , Michael R , Jinagouda SD , Boyadjian R , Steil GM Diurnal and ultradian rhythmicity of plasma leptin: effects of gender and adiposity.

J Clin Endocrinol Metab 83 : — Gavrila A , Peng CK , Chan JL , Mietus JE , Goldberger AL , Mantzoros CS Diurnal and ultradian dynamics of serum adiponectin in healthy men: comparison with leptin, circulating soluble leptin receptor, and cortisol patterns.

J Clin Endocrinol Metab 88 : — Bray MS , Young ME Circadian rhythms in the development of obesity: potential role for the circadian clock within the adipocyte. Obes Rev 8 : — Shimba S , Ishii N , Ohta Y , Ohno T , Watabe Y , Hayashi M , Wada T , Aoyagi T , Tezuka M Brain and muscle Arnt-like protein-1 BMAL1 , a component of the molecular clock, regulates adipogenesis.

Oishi K , Shirai H , Ishida N CLOCK is involved in the circadian transactivation of peroxisome-proliferator-activated receptor α PPARα in mice. Biochem J : — J Atheroscler Thromb 12 : — Canaple L , Rambaud J , Dkhissi-Benyahya O , Rayet B , Tan NS , Michalik L , Delaunay F , Wahli W , Laudet V Reciprocal regulation of brain and muscle Arnt-like protein 1 and peroxisome proliferator-activated receptor α defines a novel positive feedback loop in the rodent liver circadian clock.

Mol Endocrinol 20 : — Wang N , Yang G , Jia Z , Zhang H , Aoyagi T , Soodvilai S , Symons JD , Schnermann JB , Gonzalez FJ , Litwin SE , Yang T Vascular PPARγ controls circadian variation in blood pressure and heart rate through Bmal1.

Cell Metab 8 : — Chawla A , Lazar MA Induction of Rev-ErbA α, an orphan receptor encoded on the opposite strand of the α-thyroid hormone receptor gene, during adipocyte differentiation. Torra IP , Tsibulsky V , Delaunay F , Saladin R , Laudet V , Fruchart JC , Kosykh V , Staels B Circadian and glucocorticoid regulation of Rev-erb α expression in liver.

Fontaine C , Dubois G , Duguay Y , Helledie T , Vu-Dac N , Gervois P , Soncin F , Mandrup S , Fruchart JC , Fruchart-Najib J , Staels B The orphan nuclear receptor Rev-Erbα is a peroxisome proliferator-activated receptor PPAR γ target gene and promotes PPARγ-induced adipocyte differentiation.

Duez H , Staels B Rev-erb α gives a time cue to metabolism. FEBS Lett : 19 — Lau P , Nixon SJ , Parton RG , Muscat GE RORα regulates the expression of genes involved in lipid homeostasis in skeletal muscle cells: caveolin-3 and CPT-1 are direct targets of ROR.

Akashi M , Takumi T The orphan nuclear receptor RORα regulates circadian transcription of the mammalian core-clock Bmal1. Nat Struct Mol Biol 12 : — Ueda HR , Chen W , Adachi A , Wakamatsu H , Hayashi S , Takasugi T , Nagano M , Nakahama K , Suzuki Y , Sugano S , Iino M , Shigeyoshi Y , Hashimoto S A transcription factor response element for gene expression during circadian night.

Liu C , Li S , Liu T , Borjigin J , Lin JD Transcriptional coactivator PGC-1α integrates the mammalian clock and energy metabolism. Grimaldi B , Sassone-Corsi P Circadian rhythms: metabolic clockwork. Yanagihara H , Ando H , Hayashi Y , Obi Y , Fujimura A High-fat feeding exerts minimal effects on rhythmic mRNA expression of clock genes in mouse peripheral tissues.

Chronobiol Int 23 : — Satoh Y , Kawai H , Kudo N , Kawashima Y , Mitsumoto A Time-restricted feeding entrains daily rhythms of energy metabolism in mice. Kohsaka A , Laposky AD , Ramsey KM , Estrada C , Joshu C , Kobayashi Y , Turek FW , Bass J High-fat diet disrupts behavioral and molecular circadian rhythms in mice.

Cell Metab 6 : — Havel PJ , Townsend R , Chaump L , Teff K High-fat meals reduce h circulating leptin concentrations in women.

Cha MC , Chou CJ , Boozer CN High-fat diet feeding reduces the diurnal variation of plasma leptin concentration in rats. Metabolism 49 : — Cano P , Jiménez-Ortega V , Larrad A , Reyes Toso CF , Cardinali DP , Esquifino AI Effect of a high-fat diet on h pattern of circulating levels of prolactin, luteinizing hormone, testosterone, corticosterone, thyroid-stimulating hormone and glucose, and pineal melatonin content, in rats.

Endocrine 33 : — Barnea M , Madar Z , Froy O High-fat diet delays and fasting advances the circadian expression of adiponectin signaling components in mouse liver. Kaneko K , Yamada T , Tsukita S , Takahashi K , Ishigaki Y , Oka Y , Katagiri H Obesity alters circadian expressions of molecular clock genes in the brainstem.

Brain Res : 58 — Rudic RD , McNamara P , Curtis AM , Boston RC , Panda S , Hogenesch JB , Fitzgerald GA BMAL1 and CLOCK, two essential components of the circadian clock, are involved in glucose homeostasis. PLoS Biol 2 : e Mendoza J , Pévet P , Challet E High-fat feeding alters the clock synchronization to light.

J Physiol : — Gorman MR Differential effects of multiple short day lengths on body weights of gonadectomized Siberian hamsters. Physiol Biochem Zool 76 : — Morgan PJ , Ross AW , Mercer JG , Barrett P Photoperiodic programming of body weight through the neuroendocrine hypothalamus.

J Endocrinol : 27 — Bocquier F , Bonnet M , Faulconnier Y , Guerre-Millo M , Martin P , Chilliard Y Effects of photoperiod and feeding level on perirenal adipose tissue metabolic activity and leptin synthesis in the ovariectomized ewe. Reprod Nutr Dev 38 : — Faulconnier Y , Bonnet M , Bocquier F , Leroux C , Chilliard Y Effects of photoperiod and feeding level on adipose tissue and muscle lipoprotein lipase activity and mRNA level in dry non-pregnant sheep.

Br J Nutr 85 : — Di Lorenzo L , De Pergola G , Zocchetti C , L'Abbate N , Basso A , Pannacciulli N , Cignarelli M , Giorgino R , Soleo L Effect of shift work on body mass index: results of a study performed in glucose-tolerant men working in a Southern Italian industry.

Int J Obes Relat Metab Disord 27 : — Karlsson B , Knutsson A , Lindahl B Is there an association between shift work and having a metabolic syndrome? Results from a population based study of 27, people. Occup Environ Med 58 : — Karlsson BH , Knutsson AK , Lindahl BO , Alfredsson LS Metabolic disturbances in male workers with rotating three-shift work.

Results of the WOLF study. Int Arch Occup Environ Health 76 : — Licinio J Longitudinally sampled human plasma leptin and cortisol concentrations are inversely correlated. J Clin Endocrinol Metab 83 : Heptulla R , Smitten A , Teague B , Tamborlane WV , Ma YZ , Caprio S Temporal patterns of circulating leptin levels in lean and obese adolescents: relationships to insulin, growth hormone, and free fatty acids rhythmicity.

J Clin Endocrinol Metab 86 : 90 — Perfetto F , Tarquini R , Cornélissen G , Mello G , Tempestini A , Gaudiano P , Mancuso F , Halberg F Circadian phase difference of leptin in android versus gynoid obesity. Peptides 25 : — Zhang Y , Proenca R , Maffei M , Barone M , Leopold L , Friedman JM Positional cloning of the mouse obese gene and its human homologue.

Montague CT , Farooqi IS , Whitehead JP , Soos MA , Rau H , Wareham NJ , Sewter CP , Digby JE , Mohammed SN , Hurst JA , Cheetham CH , Earley AR , Barnett AH , Prins JB , O'Rahilly S Congenital leptin deficiency is associated with severe early-onset obesity in humans.

Navbar Search Filter Endocrine Reviews This issue Endocrine Society Journals Clinical Medicine Endocrinology and Diabetes Medicine and Health Books Journals Oxford Academic Mobile Enter search term Search.

Endocrine Society Journals. Advanced Search. Search Menu. Article Navigation. Close mobile search navigation Article Navigation. Volume Article Contents I Introduction. II Circadian Rhythms. III The Biological Clock. IV The Biological Clock and Energy Homeostasis.

V The Biological Clock and Obesity. VI Summary and Conclusions. Journal Article. Metabolism and Circadian Rhythms—Implications for Obesity. Oren Froy Oren Froy.

Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot , Israel. Oxford Academic. PDF Split View Views. Select Format Select format. ris Mendeley, Papers, Zotero. enw EndNote. bibtex BibTex. txt Medlars, RefWorks Download citation.

Permissions Icon Permissions. Close Navbar Search Filter Endocrine Reviews This issue Endocrine Society Journals Clinical Medicine Endocrinology and Diabetes Medicine and Health Books Journals Oxford Academic Enter search term Search.

Open in new tab Download slide. Fatty acyl-coenzyme A synthetase 1;. adipocyte differentiation-related protein;. adipocyte fatty acid-binding protein 2;.

CCAAT enhancer binding protein α;. circadian locomotor output cycles kaput;. nicotinamide adenine dinucleotide;. peroxisome proliferator-activated receptor-coactivator 1α;. peroxisome proliferator-activated receptor;. This would help explain why people exposed to light at the wrong time are more likely to develop metabolic diseases such as diabetes, they concluded.

Irregular sleep can mean not getting enough sleep less than 6 hours , staying up past your usual bedtime and sleeping later than normal social jetlag , or sleeping during the day and working during the night night shift work.

That has negative consequences for your blood sugar and can lead to metabolic dysfunction. Indeed, research has found that people 60 years or younger with social jetlag of greater than one hour —meaning the difference in time between when you usually go to bed and when you do on your days off, which includes 69 percent of the population—have more than 1.

Similar associations are seen in shift workers and people who work late at night. All three of these irregular sleep patterns have been linked with poor glucose control and insulin resistance, though the how and why is still unclear and likely multilayered.

Some possible factors include:. In studies, eating late at night has been associated with high blood sugar levels and insulin resistance. Much of that, researchers have thought, was because people who eat late at night also tend to consume larger portion sizes and calorie-dense foods that can lead to those issues.

But regardless of calorie intake, the actual timing of when you eat appears to be just as important a factor. Our peripheral clocks are also involved here, says Kristen Knutson, PhD , associate professor of neurology and preventive medicine at the Feinberg School of Medicine at Northwestern University in Chicago.

A study in mice suggests that these changes in circadian clock rhythms affect us at both the genetic and protein levels. While fed a high-fat diet, mice exposed to dim light at night gained more weight than their peers who were not exposed.

The researchers found that nighttime light exposure decreased the expression of fundamental clock genes; these genes encode tiny messengers that help keep our internal clocks running correctly.

This genetic disruption suggests a mechanism connecting circadian imbalance with weight gain. The good news is that, despite not knowing the direct cause and effects, experts can say for sure that there are things you can do to help keep your body in sync and operating at its peak.

The hormone most closely associated with sleep also seems to impact glucose and insulin. Ashley Welch. Murdoc Khaleghi, MD, Base Chief Medical Officer.

The Explainer. Sleep quality and quantity have a profound effect on metabolic health. This article describes the mechanisms by which improving sleep hygiene can improve glucose control.

Alex Moskov. Research Highlight. A recent study in people without diabetes found that getting poor sleep or deviating from normal sleep patterns can impact glucose control the next day. Kristen Fischer. Matthew Laye, PhD. The glycemic index provides insight into how particular foods affect glucose but has limitations.

Stephanie Eckelkamp. Ami Kapadia. Metabolic Basics. Being aware of these causes of inaccurate data can help you identify—and avoid—surprising and misleading feedback. Joy Manning, RD. Inside Levels. Levels Co-Founder's new book—Good Energy: The Surprising Connection Between Metabolism and Limitless Health—releases May 14; available for pre-order today.

The Levels Team. Metabolic flexibility means that your body can switch easily between burning glucose and fat, which means you have better energy and endurance. Jennifer Chesak. Dominic D'Agostino, PhD. Written By Naomi Barr. Reviewed By Sara Gottfried, MD. Article highlights When the cells in our eyes perceive natural light, they alert the neurons in a part of our brain that serves as the central timepiece for our circadian system.

This triggers signals that help regulate most physiological processes, including hormone release. Working along with our central clock are tiny genetic timepieces in every organ and tissue in our body. Behaviors that can cause misalignment include a lack of exposure to natural light during the day, late-night eating, and irregular sleep schedules.

Circadian misalignment can cause decreased glucose tolerance.

Yeliz RhyythmNilüfer Acar Tek; Calculate BMI of Circadian Rhythm on Metabolic Processes and the Berry Cheesecake Recipes of Energy Balance. Multivitamin for women Nutr Metaholism 28 May ; 74 BMI for Disease Risk : mstabolism Background: The metaolism timing system or circadian clock plays a crucial role in many biological rhjthm, such ghythm the Circadian rhythm metabolism cycle, hormone secretion, cardiovascular health, glucose homeostasis, and body temperature regulation. Energy balance is also one of the most important cornerstones of metabolic processes, whereas energy imbalance is associated with many diseases i. Circadian clock is the main regulator of metabolism, and this analysis provides an overview of the bidirectional effect of circadian rhythm on metabolic processes and energy balance. According to recent research, long-term circadian disruptions are associated with many pathological conditions such as premature mortality, obesity, impaired glucose tolerance, diabetes, psychiatric disorders, anxiety, depression, and cancer progression, whereas short-term disruptions are associated with impaired wellness, fatigue, and loss of concentration. Thank you for visiting Circcadian. You are using a Circadkan version metaboolism limited support for Lycopene and sun protection. To obtain the Circadiam Berry Cheesecake Recipes, we recommend you use a Circadian rhythm metabolism up to Berry Cheesecake Recipes browser merabolism turn off compatibility mode in Internet Explorer. In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript. Humans, like all mammals, partition their daily behaviour into activity wakefulness and rest sleep phases that differ largely in their metabolic requirements. The circadian clock evolved as an autonomous timekeeping system that aligns behavioural patterns with the solar day and supports the body functions by anticipating and coordinating the required metabolic programmes.

Circadian rhythm metabolism -

Endocrinology 3 — Google Scholar. Gu YZ, Hogenesch JB, Bradfield CA The PAS superfamily: sensors of environmental and developmental signals. Annu Rev Pharmacol Toxicol — Guan XM, Hess JF, Yu H, Hey PJ, van der Ploeg LH Differential expression of mRNA for leptin receptor isoforms in the rat brain.

Mol Cell Endocrinol 1 :1—7. Guilding C, Piggins HD Challenging the omnipotence of the suprachiasmatic timekeeper: are circadian oscillators present throughout the mammalian brain? Eur J Neurosci 25 11 — Harrington ME The ventral lateral geniculate nucleus and the intergeniculate leaflet: interrelated structures in the visual and circadian systems.

Neurosci Biobehav Rev 21 5 — Hatori M, Vollmers C, Zarrinpar A, DiTacchio L, Bushong EA, Gill S, Leblanc M, Chaix A, Joens M, Fitzpatrick JA, Ellisman MH, Panda S Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Cell Metab 15 6 — Hattar S, Kumar M, Park A, Tong P, Tung J, Yau KW, Berson DM Central projections of melanopsin-expressing retinal ganglion cells in the mouse.

J Comp Neurol 3 — PubMed Central PubMed Google Scholar. Haynes WG, Morgan DA, Walsh SA, Mark AL, Sivitz WI Receptor-mediated regional sympathetic nerve activation by leptin. J Clin Invest 2 — Hirota T, Okano T, Kokame K, Shirotani-Ikejima H, Miyata T, Fukada Y Glucose down-regulates Per1 and Per2 mRNA levels and induces circadian gene expression in cultured Rat-1 fibroblasts.

J Biol Chem 46 — Honma KI, Honma S, Hiroshige T Feeding-associated corticosterone peak in rats under various feeding cycles. Am J Physiol Regul Integr Comp Physiol 5 Pt 2 :R—R Inyushkin AN, Bhumbra GS, Dyball RE Leptin modulates spike coding in the rat suprachiasmatic nucleus.

J Neuroendocrinol 21 8 — Kahn BB, Alquier T, Carling D, Hardie DG AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab 1 1 — Kalsbeek A, Fliers E, Romijn JA, La Fleur SE, Wortel J, Bakker O, Endert E, Buijs RM The suprachiasmatic nucleus generates the diurnal changes in plasma leptin levels.

Endocrinology 6 — Kalsbeek A, Palm IF, La Fleur SE, Scheer FA, Perreau-Lenz S, Ruiter M, Kreier F, Cailotto C, Buijs RM SCN outputs and the hypothalamic balance of life. J Biol Rhythms 21 6 — Kalsbeek A, Verhagen LA, Schalij I, Foppen E, Saboureau M, Bothorel B, Buijs RM, Pevet P Opposite actions of hypothalamic vasopressin on circadian corticosterone rhythm in nocturnal versus diurnal species.

Eur J Neurosci 27 4 — Kalsbeek A, Yi CX, La Fleur SE, Fliers E The hypothalamic clock and its control of glucose homeostasis. Trends Endocrinol Metab 21 7 — Kaneko K, Yamada T, Tsukita S, Takahashi K, Ishigaki Y, Oka Y, Katagiri H Obesity alters circadian expressions of molecular clock genes in the brainstem.

Brain Res — Karlsson BH, Knutsson AK, Lindahl BO, Alfredsson LS Metabolic disturbances in male workers with rotating three-shift work. Results of the WOLF study. Int Arch Occup Environ Health 76 6 — Kiessling S, Eichele G, Oster H Adrenal glucocorticoids have a key role in circadian resynchronization in a mouse model of jet lag.

J Clin Invest 7 — Ko CH, Takahashi JS Molecular components of the mammalian circadian clock. Hum Mol Genet 15 Spec No 2 :R—R Kohsaka A, Laposky AD, Ramsey KM, Estrada C, Joshu C, Kobayashi Y, Turek FW, Bass J High-fat diet disrupts behavioral and molecular circadian rhythms in mice. Cell Metab 6 5 — Kreier F, Fliers E, Voshol PJ, Van Eden CG, Havekes LM, Kalsbeek A, Van Heijningen CL, Sluiter AA, Mettenleiter TC, Romijn JA, Sauerwein HP, Buijs RM Selective parasympathetic innervation of subcutaneous and intra-abdominal fat — functional implications.

J Clin Invest 9 — Diabetologia 47 8 — Kurumiya S, Kawamura H Damped oscillation of the lateral hypothalamic multineuronal activity synchronized to daily feeding schedules in rats with suprachiasmatic nucleus lesions. J Biol Rhythms 6 2 — La Fleur SE, Kalsbeek A, Wortel J, Buijs RM A suprachiasmatic nucleus generated rhythm in basal glucose concentrations.

J Neuroendocrinol 11 8 — Lambert CM, Weaver DR Peripheral gene expression rhythms in a diurnal rodent. J Biol Rhythms 21 1 — Lamia KA, Storch KF, Weitz CJ Physiological significance of a peripheral tissue circadian clock.

Proc Natl Acad Sci U S A 39 — Lamia KA, Sachdeva UM, DiTacchio L, Williams EC, Alvarez JG, Egan DF, Vasquez DS, Juguilon H, Panda S, Shaw RJ, Thompson CB, Evans RM AMPK regulates the circadian clock by cryptochrome phosphorylation and degradation.

Lamont EW, Diaz LR, Barry-Shaw J, Stewart J, Amir S Daily restricted feeding rescues a rhythm of period2 expression in the arrhythmic suprachiasmatic nucleus.

Lau P, Nixon SJ, Parton RG, Muscat GE RORalpha regulates the expression of genes involved in lipid homeostasis in skeletal muscle cells: caveolin-3 and CPT-1 are direct targets of ROR.

J Biol Chem 35 — Le Minh N, Damiola F, Tronche F, Schutz G, Schibler U Glucocorticoid hormones inhibit food-induced phase-shifting of peripheral circadian oscillators. EMBO J 20 24 — Li AJ, Wiater MF, Oostrom MT, Smith BR, Wang Q, Dinh TT, Roberts BL, Jansen HT, Ritter S a Leptin-sensitive neurons in the arcuate nuclei contribute to endogenous feeding rhythms.

Am J Physiol Regul Integr Comp Physiol 11 :R—R Li JD, Hu WP, Zhou QY b The circadian output signals from the suprachiasmatic nuclei. Liu C, Li S, Liu T, Borjigin J, Lin JD Transcriptional coactivator PGC-1alpha integrates the mammalian clock and energy metabolism.

Nature — Lowden A, Moreno C, Holmback U, Lennernas M, Tucker P Eating and shift work — effects on habits, metabolism and performance. Scand J Work Environ Health 36 2 — Malek ZS, Sage D, Pevet P, Raison S Daily rhythm of tryptophan hydroxylase-2 messenger ribonucleic acid within raphe neurons is induced by corticoid daily surge and modulated by enhanced locomotor activity.

Endocrinology 11 — 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, Bass J Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes.

J Neurosci 25 6 — Mendoza J, Pevet P, Challet E High-fat feeding alters the clock synchronization to light. J Physiol Pt 24 — Mendoza J, Clesse D, Pevet P, Challet E Food-reward signalling in the suprachiasmatic clock.

J Neurochem 6 — Mendoza J, Lopez-Lopez C, Revel FG, Jeanneau K, Delerue F, Prinssen E, Challet E, Moreau JL, Grundschober C Dimorphic effects of leptin on the circadian and hypocretinergic systems of mice.

J Neuroendocrinol 23 1 — Mistlberger RE Neurobiology of food anticipatory circadian rhythms. Physiol Behav 4 — Mistlberger RE, Lukman H, Nadeau BG Circadian rhythms in the Zucker obese rat: assessment and intervention.

Appetite 30 3 — Morin LP Serotonin and the regulation of mammalian circadian rhythmicity. Ann Med 31 1 — Mrosovsky N, Edelstein K, Hastings MH, Maywood ES Cycle of period gene expression in a diurnal mammal Spermophilus tridecemlineatus : implications for nonphotic phase shifting.

J Biol Rhythms 16 5 — Mühlbauer E, Gross E, Labucay K, Wolgast S, Peschke E Loss of melatonin signalling and its impact on circadian rhythms in mouse organs regulating blood glucose.

Eur J Pharmacol 1—3 — Mulder H, Nagorny CL, Lyssenko V, Groop L Melatonin receptors in pancreatic islets: good morning to a novel type 2 diabetes gene. Diabetologia 52 7 — Nagai K, Nishio T, Nakagawa H, Nakamura S, Fukuda Y Effect of bilateral lesions of the suprachiasmatic nuclei on the circadian rhythm of food-intake.

Brain Res 2 — Oishi K, Amagai N, Shirai H, Kadota K, Ohkura N, Ishida N a Genome-wide expression analysis reveals adrenal gland-dependent circadian genes in the mouse liver. DNA Res 12 3 — Oishi K, Shirai H, Ishida N b CLOCK is involved in the circadian transactivation of peroxisome-proliferator-activated receptor alpha PPARalpha in mice.

Biochem J Pt 3 — Oster H, Damerow S, Kiessling S, Jakubcakova V, Abraham D, Tian J, Hoffmann MW, Eichele G The circadian rhythm of glucocorticoids is regulated by a gating mechanism residing in the adrenal cortical clock.

Cell Metab 4 2 — Otway DT, Frost G, Johnston JD Circadian rhythmicity in murine pre-adipocyte and adipocyte cells. Chronobiol Int 26 7 — Otway DT, Mantele S, Bretschneider S, Wright J, Trayhurn P, Skene DJ, Robertson MD, Johnston JD Rhythmic diurnal gene expression in human adipose tissue from individuals who are lean, overweight, and type 2 diabetic.

Diabetes 60 5 — Panda S, Antoch MP, Miller BH, Su AI, Schook AB, Straume M, Schultz PG, Kay SA, Takahashi JS, Hogenesch JB Coordinated transcription of key pathways in the mouse by the circadian clock.

Cell 3 — Peschke E, Peschke D Evidence for a circadian rhythm of insulin release from perifused rat pancreatic islets. Diabetologia 41 9 — Pevet P, Challet E Melatonin: both master clock output and internal time-giver in the circadian clocks network.

J Physiol Paris 4—6 — Prosser RA, Bergeron HE Leptin phase-advances the rat suprachiasmatic circadian clock in vitro.

Reick M, Garcia JA, Dudley C, McKnight SL NPAS2: an analog of clock operative in the mammalian forebrain. Reid KJ, Baron KG, Zee PC Meal timing influences daily caloric intake in healthy adults.

Nutr Res 34 11 — Rosenfeld P, Van Eekelen JA, Levine S, De Kloet ER Ontogeny of the type 2 glucocorticoid receptor in discrete rat brain regions: an immunocytochemical study. Rutter J, Reick M, Wu LC, McKnight SL Regulation of clock and NPAS2 DNA binding by the redox state of NAD cofactors.

Sack RL, Hughes RJ, Edgar DM, Lewy AJ Sleep-promoting effects of melatonin: at what dose, in whom, under what conditions, and by what mechanisms? Sleep 20 10 — Saderi N, Cazarez-Marquez F, Buijs FN, Salgado-Delgado RC, Guzman-Ruiz MA, del Carmen Basualdo M, Escobar C, Buijs RM The NPY intergeniculate leaflet projections to the suprachiasmatic nucleus transmit metabolic conditions.

Neuroscience — Sage D, Ganem J, Guillaumond F, Laforge-Anglade G, Francois-Bellan AM, Bosler O, Becquet D Influence of the corticosterone rhythm on photic entrainment of locomotor activity in rats. J Biol Rhythms 19 2 — Saini C, Morf J, Stratmann M, Gos P, Schibler U Simulated body temperature rhythms reveal the phase-shifting behavior and plasticity of mammalian circadian oscillators.

Genes Dev 26 6 — Salgado-Delgado R, Angeles-Castellanos M, Buijs MR, Escobar C Internal desynchronization in a model of night-work by forced activity in rats. Neuroscience 3 — Sans-Fuentes MA, Diez-Noguera A, Cambras T Light responses of the circadian system in leptin deficient mice.

Physiol Behav 99 4 — Sato M, Murakami M, Node K, Matsumura R, Akashi M The role of the endocrine system in feeding-induced tissue-specific circadian entrainment. Cell Rep 8 2 — Schmutz I, Ripperger JA, Baeriswyl-Aebischer S, Albrecht U The mammalian clock component PERIOD2 coordinates circadian output by interaction with nuclear receptors.

Genes Dev 24 4 — Sellix MT, Egli M, Poletini MO, McKee DT, Bosworth MD, Fitch CA, Freeman ME Anatomical and functional characterization of clock gene expression in neuroendocrine dopaminergic neurons.

Am J Physiol Regul Integr Comp Physiol 5 :R—R Shibata S, Liou SY, Ueki S, Oomura Y Inhibitory action of insulin on suprachiasmatic nucleus neurons in rat hypothalamic slice preparations.

Physiol Behav 36 1 — Shieh KR, Yang SC, Lu XY, Akil H, Watson SJ Diurnal rhythmic expression of the rhythm-related genes, rPeriod1, rPeriod2, and rClock, in the rat brain. J Biomed Sci 12 1 — Silver R, LeSauter J, Tresco PA, Lehman MN A diffusible coupling signal from the transplanted suprachiasmatic nucleus controlling circadian locomotor rhythms.

Sinha MK, Ohannesian JP, Heiman ML, Kriauciunas A, Stephens TW, Magosin S, Marco C, Caro JF Nocturnal rise of leptin in lean, obese, and non-insulin-dependent diabetes mellitus subjects. J Clin Invest 97 5 — Spiegel K, Tasali E, Leproult R, Van Cauter E Effects of poor and short sleep on glucose metabolism and obesity risk.

Nat Rev Endocrinol 5 5 — Steiner RA, Kabigting E, Lent K, Clifton DK Diurnal rhythm in proopiomelanocortin mRNA in the arcuate nucleus of the male rat.

J Neuroendocrinol 6 6 — Stokkan KA, Yamazaki S, Tei H, Sakaki Y, Menaker M Entrainment of the circadian clock in the liver by feeding. Striegel-Moore RH, Rosselli F, Wilson GT, Perrin N, Harvey K, DeBar L Nocturnal eating: association with binge eating, obesity, and psychological distress.

Int J Eat Disord 43 6 — Surjit M, Ganti KP, Mukherji A, Ye T, Hua G, Metzger D, Li M, Chambon P Widespread negative response elements mediate direct repression by agonist-liganded glucocorticoid receptor. Tahara Y, Otsuka M, Fuse Y, Hirao A, Shibata S Refeeding after fasting elicits insulin-dependent regulation of Per2 and Rev-erbalpha with shifts in the liver clock.

J Biol Rhythms 26 3 — Tahara Y, Kuroda H, Saito K, Nakajima Y, Kubo Y, Ohnishi N, Seo Y, Otsuka M, Fuse Y, Ohura Y, Komatsu T, Moriya Y, Okada S, Furutani N, Hirao A, Horikawa K, Kudo T, Shibata S In vivo monitoring of peripheral circadian clocks in the mouse.

Curr Biol 22 11 — Takeda S, Elefteriou F, Levasseur R, Liu X, Zhao L, Parker KL, Armstrong D, Ducy P, Karsenty G Leptin regulates bone formation via the sympathetic nervous system.

Teboul M, Guillaumond F, Grechez-Cassiau A, Delaunay F The nuclear hormone receptor family round the clock. Mol Endocrinol 22 12 — Tsai LL, Tsai YC, Hwang K, Huang YW, Tzeng JE Repeated light — dark shifts speed up body weight gain in male F rats. Am J Physiol Endocrinol Metab 2 :E—E Turek FW, Joshu C, Kohsaka A, Lin E, Ivanova G, McDearmon E, Laposky A, Losee-Olson S, Easton A, Jensen DR, Eckel RH, Takahashi JS, Bass J Obesity and metabolic syndrome in circadian Clock mutant mice.

Ulrich-Lai YM, Arnhold MM, Engeland WC Adrenal splanchnic innervation contributes to the diurnal rhythm of plasma corticosterone in rats by modulating adrenal sensitivity to ACTH.

Am J Physiol Regul Integr Comp Physiol 4 :R—R J Biol Chem 29 — Unger JW, Livingston JN, Moss AM Insulin receptors in the central nervous system: localization, signalling mechanisms and functional aspects.

Prog Neurobiol 36 5 — Wakamatsu H, Yoshinobu Y, Aida R, Moriya T, Akiyama M, Shibata S Restricted-feeding-induced anticipatory activity rhythm is associated with a phase-shift of the expression of mPer1 and mPer2 mRNA in the cerebral cortex and hippocampus but not in the suprachiasmatic nucleus of mice.

Eur J Neurosci 13 6 — Wang TA, Yu YV, Govindaiah G, Ye X, Artinian L, Coleman TP, Sweedler JV, Cox CL, Gillette MU Circadian rhythm of redox state regulates excitability in suprachiasmatic nucleus neurons. Wiater MF, Mukherjee S, Li AJ, Dinh TT, Rooney EM, Simasko SM, Ritter S Circadian integration of sleep-wake and feeding requires NPY receptor-expressing neurons in the mediobasal hypothalamus.

Williams KW, Elmquist JK From neuroanatomy to behavior: central integration of peripheral signals regulating feeding behavior. Nat Neurosci 15 10 — Xu B, Kalra PS, Farmerie WG, Kalra SP Daily changes in hypothalamic gene expression of neuropeptide Y, galanin, proopiomelanocortin, and adipocyte leptin gene expression and secretion: effects of food restriction.

Yagita K, Tamanini F, van Der Horst GT, Okamura H Molecular mechanisms of the biological clock in cultured fibroblasts. Yanagihara H, Ando H, Hayashi Y, Obi Y, Fujimura A High-fat feeding exerts minimal effects on rhythmic mRNA expression of clock genes in mouse peripheral tissues.

Chronobiol Int 23 5 — Yannielli PC, Molyneux PC, Harrington ME, Golombek DA Ghrelin effects on the circadian system of mice. J Neurosci 27 11 — Yi CX, van der Vliet J, Dai J, Yin G, Ru L, Buijs RM Ventromedial arcuate nucleus communicates peripheral metabolic information to the suprachiasmatic nucleus.

Endocrinology 1 — Yi CX, Challet E, Pevet P, Kalsbeek A, Escobar C, Buijs RM A circulating ghrelin mimetic attenuates light-induced phase delay of mice and light-induced Fos expression in the suprachiasmatic nucleus of rats.

Eur J Neurosci 27 8 — Yi CX, Serlie MJ, Ackermans MT, Foppen E, Buijs RM, Sauerwein HP, Fliers E, Kalsbeek A A major role for perifornical orexin neurons in the control of glucose metabolism in rats.

Diabetes 58 9 — Proc Natl Acad Sci U S A 15 — Yoon M The role of PPARalpha in lipid metabolism and obesity: focusing on the effects of estrogen on PPARalpha actions. Pharmacol Res 60 3 — Yu J, Auwerx J The role of sirtuins in the control of metabolic homeostasis.

Ann N Y Acad Sci Suppl 1 :E10—E Zigman JM, Jones JE, Lee CE, Saper CB, Elmquist JK Expression of ghrelin receptor mRNA in the rat and the mouse brain. Zvonic S, Ptitsyn AA, Conrad SA, Scott LK, Floyd ZE, Kilroy G, Wu X, Goh BC, Mynatt RL, Gimble JM Characterization of peripheral circadian clocks in adipose tissues.

Diabetes 55 4 — Download references. Regulation of Circadian Clocks team, Institute of Cellular and Integrative Neurosciences, CNRS, UPR, University of Strasbourg, Strasbourg, France. You can also search for this author in PubMed Google Scholar. Correspondence to Etienne Challet.

Perelman School of Medicine, University of Pennsylvania, Pennsylania, Pennsylvania, USA. Reprints and permissions. Grosbellet, E. Circadian Rhythms and Metabolism.

In: Ahima, R. eds Metabolic Syndrome. Springer, Cham. Received : 21 February Accepted : 22 April Published : 16 June Publisher Name : Springer, Cham. Online ISBN : eBook Packages : Springer Reference Medicine Reference Module Medicine.

Policies and ethics. Skip to main content. Abstract The circadian system relies on a master clock in the suprachiasmatic nucleus of the hypothalamus SCN , synchronizing a multitude of brain and peripheral oscillators that set physiological and metabolic functions in phase with the light—dark cycle.

Keywords Circadian rhythm Clock gene Feeding time Desynchronization Metabolic disturbances Obesity Diabetes. References Abe M, Herzog ED, Yamazaki S, Straume M, Tei H, Sakaki Y, Menaker M, Block GD Circadian rhythms in isolated brain regions.

J Neurosci 22 1 — CAS PubMed Google Scholar Ahima RS, Lazar MA Adipokines and the peripheral and neural control of energy balance. Mol Cell Neurosci 5 3 — CAS PubMed Google Scholar Ando H, Yanagihara H, Hayashi Y, Obi Y, Tsuruoka S, Takamura T, Kaneko S, Fujimura A Rhythmic messenger ribonucleic acid expression of clock genes and adipocytokines in mouse visceral adipose tissue.

Endocrinology 4 — CAS PubMed Google Scholar Asher G, Gatfield D, Stratmann M, Reinke H, Dibner C, Kreppel F, Mostoslavsky R, Alt FW, Schibler U SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell 2 — CAS PubMed Google Scholar Balland E, Dam J, Langlet F, Caron E, Steculorum S, Messina A, Rasika S, Falluel-Morel A, Anouar Y, Dehouck B, Trinquet E, Jockers R, Bouret SG, Prevot V Hypothalamic tanycytes are an ERK-gated conduit for leptin into the brain.

Cell Metab 19 2 — CAS PubMed Central PubMed Google Scholar Balsalobre A, Damiola F, Schibler U A serum shock induces circadian gene expression in mammalian tissue culture cells. Cell 93 6 — CAS PubMed Google Scholar Balsalobre A, Brown SA, Marcacci L, Tronche F, Kellendonk C, Reichardt HM, Schutz G, Schibler U a Resetting of circadian time in peripheral tissues by glucocorticoid signaling.

Science — CAS PubMed Google Scholar Balsalobre A, Marcacci L, Schibler U b Multiple signaling pathways elicit circadian gene expression in cultured Rat-1 fibroblasts. Curr Biol 10 20 — CAS PubMed Google Scholar Bartol-Munier I, Gourmelen S, Pevet P, Challet E Combined effects of high-fat feeding and circadian desynchronization.

Int J Obes Lond 30 1 —67 CAS Google Scholar Bechtold DA, Loudon AS Hypothalamic clocks and rhythms in feeding behaviour. Trends Neurosci 36 2 —82 CAS PubMed Google Scholar Bouatia-Naji N, Bonnefond A, Cavalcanti-Proenca C, Sparso T, Holmkvist J, Marchand M, Delplanque J, Lobbens S, Rocheleau G, Durand E, De Graeve F, Chevre JC, Borch-Johnsen K, Hartikainen AL, Ruokonen A, Tichet J, Marre M, Weill J, Heude B, Tauber M, Lemaire K, Schuit F, Elliott P, Jorgensen T, Charpentier G, Hadjadj S, Cauchi S, Vaxillaire M, Sladek R, Visvikis-Siest S, Balkau B, Levy-Marchal C, Pattou F, Meyre D, Blakemore AI, Jarvelin MR, Walley AJ, Hansen T, Dina C, Pedersen O, Froguel P A variant near MTNR1B is associated with increased fasting plasma glucose levels and type 2 diabetes risk.

Nat Genet 41 1 —94 CAS PubMed Google Scholar Brown SA, Zumbrunn G, Fleury-Olela F, Preitner N, Schibler U Rhythms of mammalian body temperature can sustain peripheral circadian clocks. Curr Biol 12 18 — CAS PubMed Google Scholar Brown TM, Coogan AN, Cutler DJ, Hughes AT, Piggins HD Electrophysiological actions of orexins on rat suprachiasmatic neurons in vitro.

Neurosci Lett 3 — CAS PubMed Google Scholar Buhr ED, Yoo SH, Takahashi JS Temperature as a universal resetting cue for mammalian circadian oscillators. Science — CAS PubMed Central PubMed Google Scholar Burdakov D, Luckman SM, Verkhratsky A Glucose-sensing neurons of the hypothalamus.

Philos Trans R Soc Lond B Biol Sci — CAS PubMed Central PubMed Google Scholar Cailotto C, van Heijningen C, van der Vliet J, van der Plasse G, Habold C, Kalsbeek A, Pevet P, Buijs RM Daily rhythms in metabolic liver enzymes and plasma glucose require a balance in the autonomic output to the liver.

Zhang, E. Cryptochrome mediates circadian regulation of cAMP signaling and hepatic gluconeogenesis. Yin, L. Rev-erbα, a heme sensor that coordinates metabolic and circadian pathways.

Nuclear receptor Rev-erbα: a heme receptor that coordinates circadian rhythm and metabolism. Preitner, N. Orphan nuclear receptors, molecular clockwork, and the entrainment of peripheral oscillators.

Novartis Found. Zhang, Y. Discrete functions of nuclear receptor Rev-erbα couple metabolism to the clock.

Report of the finding that REV-ERBAα has major metabolic functions outside the core clock mechanism. Feng, D. A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism. Peterson, T. mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway.

Le Martelot, G. REV-ERBα participates in circadian SREBP signaling and bile acid homeostasis. Woldt, E. Rev-erb-α modulates skeletal muscle oxidative capacity by regulating mitochondrial biogenesis and autophagy. Kallen, J. X-ray structure of the hRORα LBD at 1. Structure 10 , — Yang, X.

Nuclear receptor expression links the circadian clock to metabolism. Schmutz, I. The mammalian clock component PERIOD2 coordinates circadian output by interaction with nuclear receptors. Kriebs, A. Circadian repressors CRY1 and CRY2 broadly interact with nuclear receptors and modulate transcriptional activity.

Liu, C. Transcriptional coactivator PGC-1α integrates the mammalian clock and energy metabolism. High-fat diet disrupts behavioral and molecular circadian rhythms in mice.

Demonstration of the effect of high-fat diet on circadian clocks. Gill, S. Smartphone app reveals erratic diurnal eating patterns in humans that can be modulated for health benefits. Bolli, G. Demonstration of a dawn phenomenon in normal human volunteers.

Diabetes 33 , — Feigin, R. Circadian periodicity of blood amino-acids in adult men. Rivera-Coll, A. Circadian rhythmic variations in serum concentrations of clinically important lipids. Van Cauter, E. Roles of circadian rhythmicity and sleep in human glucose regulation.

Muscle insulin sensitivity and glucose metabolism are controlled by the intrinsic muscle clock. Marcheva, B. Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. The discovery of the role of the pancreatic circadian clock in insulin and glucose homeostasis.

Perelis, M. Pancreatic beta cell enhancers regulate rhythmic transcription of genes controlling insulin secretion.

Science , aac Shi, S. Circadian disruption leads to insulin resistance and obesity. Zhou, B. Hepatology 59 , — Transcriptional programming of lipid and amino acid metabolism by the skeletal muscle circadian clock. Yao, Z. Modeling circadian rhythms of glucocorticoid receptor and glutamine synthetase expression in rat skeletal muscle.

Benavides, A. Circadian rhythms of lipoprotein lipase and hepatic lipase activities in intermediate metabolism of adult rat. Mortola, J. Breathing around the clock: an overview of the circadian pattern of respiration. Rhythmic oxygen levels reset circadian clocks through HIF1α.

Emans, T. Circadian rhythm in kidney tissue oxygenation in the rat. Circadian clock interaction with HIF1α mediates oxygenic metabolism and anaerobic glycolysis in skeletal muscle. Rodrigo, G. Regulation of vascular function and blood pressure by circadian variation in redox signalling.

Free Radic. Franken, P. A role for clock genes in sleep homeostasis. Viola, A. PER3 polymorphism predicts sleep structure and waking performance.

Schmidt, M. The energy allocation function of sleep: a unifying theory of sleep, torpor, and continuous wakefulness. Jung, C. Energy expenditure during sleep, sleep deprivation and sleep following sleep deprivation in adult humans. Hirota, T. Glucose down-regulates Per1 and Per2 mRNA levels and induces circadian gene expression in cultured Rat-1 fibroblasts.

Oike, H. Feeding cues and injected nutrients induce acute expression of multiple clock genes in the mouse liver. PLOS ONE 6 , e Dang, F. Insulin post-transcriptionally modulates Bmal1 protein to affect the hepatic circadian clock.

Cell-specific resetting of mouse islet cellular clocks by glucagon, glucagon-like peptide 1 and somatostatin. Acta Physiol , e Google Scholar.

Resetting of circadian time in peripheral tissues by glucocorticoid signaling. Multiple signaling pathways elicit circadian gene expression in cultured Rat-1 fibroblasts. Hart, G. Cycling of O-linked β-N-acetylglucosamine on nucleocytoplasmic proteins.

Ma, Y. O-GlcNAcylation of BMAL1 regulates circadian rhythms in NIH3T3 fibroblasts. Li, M. Kaasik, K. Glucose sensor O-GlcNAcylation coordinates with phosphorylation to regulate circadian clock.

Hui, S. Glucose feeds the TCA cycle via circulating lactate. Rutter, J. Regulation of clock and NPAS2 DNA binding by the redox state of NAD cofactors.

Hogenesch, J. Characterization of a subset of the basic-helix-loop-helix-PAS superfamily that interacts with components of the dioxin signaling pathway. Wu, Y.

Reciprocal regulation between the circadian clock and hypoxia signaling at the genome level in mammals. Refs 90, 92 and are three related publications showing the interplay between oxygen, HIF and circadian clocks.

Klemz, R. Reciprocal regulation of carbon monoxide metabolism and the circadian clock. Dioum, E. NPAS2: a gas-responsive transcription factor.

Correa-Costa, M. Carbon monoxide protects the kidney through the central circadian clock and CD A genome-wide RNAi screen for modifiers of the circadian clock in human cells.

Ramsey, K. Nakahata, Y. Aguilar-Arnal, L. Asher, G. SIRT1 regulates circadian clock gene expression through PER2 deacetylation.

Chang, H. SIRT1 mediates central circadian control in the SCN by a mechanism that decays with aging. Partitioning circadian transcription by SIRT6 leads to segregated control of cellular metabolism.

Refs and are two related publications reporting that SIRT1 regulates circadian rhythmicity. Musiek, E. Circadian clock proteins regulate neuronal redox homeostasis and neurodegeneration.

Choi, J. Non-thermal plasma-induced apoptosis is modulated by ATR- and PARP1-mediated DNA damage responses and circadian clock. Oncotarget 7 , — Poly ADP-ribose polymerase 1 participates in the phase entrainment of circadian clocks to feeding. Zhao, H. PARP1- and CTCF-mediated interactions between active and repressed chromatin at the lamina promote oscillating transcription.

Cell 59 , — Purushotham, A. Hepatocyte-specific deletion of SIRT1 alters fatty acid metabolism and results in hepatic steatosis and inflammation.

Rodgers, J. Nutrient control of glucose homeostasis through a complex of PGC-1α and SIRT1. Liu, Y. Hirano, A. The pentose phosphate pathway regulates the circadian clock.

AMPK regulates the circadian clock by cryptochrome phosphorylation and degradation. Discovery that AMPK regulates circadian rhythmicity. Um, J. Activation of 5ʹ-AMP-activated kinase with diabetes drug metformin induces casein kinase Iepsilon CKIepsilon -dependent degradation of clock protein mPer2.

Canto, C. Zhang, C. Fructose-1,6-bisphosphate and aldolase mediate glucose sensing by AMPK. Yamamoto, H. Role of SCN in daily rhythms of plasma glucose, FFA, insulin and glucagon. Wellen, K. ATP-citrate lyase links cellular metabolism to histone acetylation.

Ariyannur, P. Nuclear-cytoplasmic localization of acetyl coenzyme a synthetase-1 in the rat brain. Zhao, S. Regulation of cellular metabolism by protein lysine acetylation. Sahar, S. Circadian control of fatty acid elongation by SIRT1 protein-mediated deacetylation of acetyl-coenzyme A synthetase 1.

Etchegaray, J. Rhythmic histone acetylation underlies transcription in the mammalian circadian clock. Doi, M. Circadian regulator CLOCK is a histone acetyltransferase. Circadian clock control by polyamine levels through a mechanism that declines with age.

Turek, F. Obesity and metabolic syndrome in circadian Clock mutant mice. Rudic, R. BMAL1 and CLOCK, two essential components of the circadian clock, are involved in glucose homeostasis. Ghiasvand, M.

Shift working and risk of lipid disorders: a cross-sectional study. Lipids Health Dis. Morgan, L. Circadian aspects of postprandial metabolism. Pan, A. Rotating night shift work and risk of type 2 diabetes: two prospective cohort studies in women.

PLOS Med. Thosar, S. Role of the circadian system in cardiovascular disease. Konturek, P. Gut clock: implication of circadian rhythms in the gastrointestinal tract. Schernhammer, E. Natl Cancer Inst. De Bacquer, D. Rotating shift work and the metabolic syndrome: a prospective study.

Zizi, F. Sleep duration and the risk of diabetes mellitus: epidemiologic evidence and pathophysiologic insights. Diabetes Rep. Lucassen, E. Interacting epidemics? Sleep curtailment, insulin resistance, and obesity.

NY Acad. Buxton, O. Adverse metabolic consequences in humans of prolonged sleep restriction combined with circadian disruption. Transl Med. Mullington, J. Sleep loss reduces diurnal rhythm amplitude of leptin in healthy men.

Schmid, S. A single night of sleep deprivation increases ghrelin levels and feelings of hunger in normal-weight healthy men. Beccuti, G. Sleep and obesity.

Care 14 , — Cui, H. The cellular and molecular bases of leptin and ghrelin resistance in obesity. The suprachiasmatic nucleus generates the diurnal changes in plasma leptin levels. Endocrinology , — Laermans, J. Role of the clock gene Bmal1 and the gastric ghrelin-secreting cell in the circadian regulation of the ghrelin-GOAT system.

Kettner, N. Circadian dysfunction induces leptin resistance in mice. Xu, Y. Functional consequences of a CKIδ mutation causing familial advanced sleep phase syndrome.

A cryptochrome 2 mutation yields advanced sleep phase in humans. eLife 5 , e Patke, A. Mutation of the human circadian clock gene CRY1 in familial delayed sleep phase disorder. Parsons, M.

Social jetlag, obesity and metabolic disorder: investigation in a cohort study. CAS Google Scholar. Roenneberg, T. Social jetlag and obesity. Chen, Z. Development and therapeutic potential of small-molecule modulators of circadian systems.

Ouyang, Y. Resonating circadian clocks enhance fitness in cyanobacteria. USA 95 , — Woelfle, M. The adaptive value of circadian clocks: an experimental assessment in cyanobacteria. Dodd, A. Plant circadian clocks increase photosynthesis, growth, survival, and competitive advantage.

Martino, T. Circadian rhythm disorganization produces profound cardiovascular and renal disease in hamsters. Wang, G. Cycling transcriptional networks optimize energy utilization on a genome scale.

Stokkan, K. Entrainment of the circadian clock in the liver by feeding. Demonstration that daytime feeding uncouples organ clocks from the SCN. Mendoza, J. Acosta-Rodriguez, V. Mice under caloric restriction self-impose a temporal restriction of food intake as revealed by an automated feeder system.

Potthoff, M. Bookout, A. FGF21 regulates metabolism and circadian behavior by acting on the nervous system. Hakansson, M. Leptin receptor immunoreactivity in chemically defined target neurons of the hypothalamus. Zigman, J. Expression of ghrelin receptor mRNA in the rat and the mouse brain.

Grosbellet, E. Biochimie , — Mistlberger, R. Food-anticipatory circadian rhythms: concepts and methods. Orozco-Solis, R.

The circadian clock in the ventromedial hypothalamus controls cyclic energy expenditure. Reprogramming of the circadian clock by nutritional challenge. Abbondante, S. Comparative circadian metabolomics reveal differential effects of nutritional challenge in the serum and liver.

Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Sherman, H. Timed high-fat diet resets circadian metabolism and prevents obesity.

FASEB J. Salgado-Delgado, R. Food intake during the normal activity phase prevents obesity and circadian desynchrony in a rat model of night work.

Branecky, K. Disruption of daily rhythms by high-fat diet is reversible. PLOS ONE 10 , e Tognini, P. Distinct circadian signatures in liver and gut clocks revealed by ketogenic diet.

Lynch, S. The human intestinal microbiome in health and disease. Mukherji, A. Homeostasis in intestinal epithelium is orchestrated by the circadian clock and microbiota cues transduced by TLRs. Thaiss, C. Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis.

Refs , and report the discovery of the metabolic benefits of time-restricted feeding. Liang, X. Rhythmicity of the intestinal microbiota is regulated by gender and the host circadian clock. Zarrinpar, A. Diet and feeding pattern affect the diurnal dynamics of the gut microbiome. Leone, V.

Effects of diurnal variation of gut microbes and high-fat feeding on host circadian clock function and metabolism. Cell Host Microbe 17 , — Murakami, M.

Gut microbiota directs PPARgamma-driven reprogramming of the liver circadian clock by nutritional challenge. EMBO Rep. Microbiota diurnal rhythmicity programs host transcriptome oscillations. Kil, I. Circadian oscillation of sulfiredoxin in the mitochondria. Edgar, R. Peroxiredoxins are conserved markers of circadian rhythms.

Circadian rhythms persist without transcription in a eukaryote. Grimaldi, B. PER2 controls lipid metabolism by direct regulation of PPARgamma. Wang, Y. Modulation of retinoic acid receptor-related orphan receptor alpha and gamma activity by 7-oxygenated sterol ligands.

Perrin, L. Human skeletal myotubes display a cell-autonomous circadian clock implicated in basal myokine secretion. Philippe, J. Thyroid circadian timing: roles in physiology and thyroid malignancies. Rhythms 30 , 76—83 A circadian rhythm of serum follicle-stimulating hormone in women.

Chung, S. Circadian rhythm of adrenal glucocorticoid: its regulation and clinical implications. Waldstreicher, J. Gender differences in the temporal organization of proclactin PRL secretion: evidence for a sleep-independent circadian rhythm of circulating PRL levels- a clinical research center study.

Lynch, H. Daily rhythm in human urinary melatonin. Eggink, H. Complex interaction between circadian rhythm and diet on bile acid homeostasis in male rats.

Ohashi, N. Circadian rhythm of blood pressure and the renin-angiotensin system in the kidney. Download references. The authors are grateful to R. Aviram and G. Manella for their valuable comments on the manuscript and for their assistance in the figure preparation. is recipient of the European Molecular Biology Organization EMBO young investigator award.

Nature Reviews Molecular Cell Biology thanks A. Weljie, F. Gachon and other anonymous reviewer s for their contribution to the peer review of this work. University of Düsseldorf, Medical Faculty, Institute of Clinical Chemistry and Laboratory Diagnostics, Düsseldorf, Germany.

IUF Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany. Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel. You can also search for this author in PubMed Google Scholar.

Correspondence to Hans Reinke or Gad Asher. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. DNA elements consensus sequence CANNTG bound by transcription factors, most commonly basic helix—loop—helix domain-containing proteins.

Light-induced molecular changes in cells that contribute to photoentrainment in the suprachiasmatic nucleus. A hypothalamic nucleus that contains neuroendocrine and centrally projecting neurons and that has pre-eminent roles in central homeostatic processes, such as energy metabolism.

A high-fat, low-carbohydrate diet that results in elevated levels of ketone bodies in the circulation by promoting the metabolism of lipids over the use of carbohydrates for energy generation.

Opsin of intrinsically photoactive retinal ganglion cells involved in non-image-forming visual functions including light-entrainment of the suprachiasmatic nucleus. Neurons in the ganglion cell layer of the retina; their axons form the optic nerve and the retinohypothalamic tract.

A pineal hormone that regulates circadian rhythms and wakefulness; its synthesis is suppressed by light. A porphyrin complex with a central iron atom that can bind and transport diatomic gases and can be used as a redox partner in electron transfer reactions.

Oxidized derivatives of cholesterol with biological activity, for example, as binding partners for nuclear receptors. Transmembrane receptors with homology to the Drosophila melanogaster Toll protein that recognize microbial pathogen structures.

Classifiers for clusters of closely related organisms, in particular used for prokaryotes owing to the lack of a traditional system of biological classification. Post-translational modification of proteins, whereby N -acetylglucosamine is covalently attached via an O -glycosidic linkage to serine or threonine residues.

Substrate use in a biochemical pathway determined as the turnover rate of a metabolite as opposed to its steady-state levels, which can be constant in different conditions despite widely varying flux rates. A small molecule that is covalently bound to a protein and is essential for its function.

An example of a prosthetic group is haem bound to haemoglobin. The tissue damage caused by oxidative stress when cells are resupplied with oxygen and nutrients after a period of anoxia, for example, after a stroke.

An adipocyte-derived hormone that can cross the blood—brain barrier and inhibit hunger by regulating the production of other satiety-controlling hormones in the hypothalamus.

The name comes from a Greek word meaning thin. Growth hormone release inducing. A gastrointestinal peptide hormone that stimulates growth hormone secretion from the anterior pituitary and can cross the blood—brain barrier to increase hunger in the hypothalamus antagonistically to leptin.

Reprints and permissions. Crosstalk between metabolism and circadian clocks. Nat Rev Mol Cell Biol 20 , — Download citation. Published : 11 January Issue Date : April Anyone you share the following link with will be able to read this content:.

Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative. Journal of Animal Science and Biotechnology Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Skip to main content Thank you for visiting nature. nature nature reviews molecular cell biology review articles article. Subjects Circadian mechanisms Circadian rhythms Metabolic disorders Metabolic pathways Metabolism. Abstract Humans, like all mammals, partition their daily behaviour into activity wakefulness and rest sleep phases that differ largely in their metabolic requirements.

Access through your institution. Buy or subscribe. Change institution. Learn more. References Teleman, A. CAS PubMed Google Scholar Hanahan, D. CAS PubMed Google Scholar Lopez-Otin, C.

CAS PubMed PubMed Central Google Scholar DeBerardinis, R. CAS PubMed PubMed Central Google Scholar Le Loir, Y. PubMed Google Scholar Reinke, H. PubMed Google Scholar Pittendrigh, C. CAS PubMed Google Scholar Lamia, K. CAS PubMed Google Scholar Borbely, A.

PubMed Google Scholar Greenham, K. CAS PubMed Google Scholar Allada, R. CAS PubMed Google Scholar Shultzaberger, R. CAS PubMed PubMed Central Google Scholar Takahashi, J. PubMed PubMed Central Google Scholar Feeney, K. CAS PubMed PubMed Central Google Scholar Henslee, E.

PubMed PubMed Central Google Scholar Zhang, R. CAS PubMed Google Scholar Hatori, M. CAS PubMed PubMed Central Google Scholar Buijs, F. PubMed Google Scholar Damiola, F. CAS PubMed PubMed Central Google Scholar Kalsbeek, A.

CAS PubMed Google Scholar Gerber, A. CAS PubMed Google Scholar Balsalobre, A. CAS PubMed Google Scholar Krishnaiah, S.

The circadian system metaboolism Berry Cheesecake Recipes a master clock in the suprachiasmatic Berry Cheesecake Recipes of the Non-stimulant fat burners SCNsynchronizing rhuthm multitude of brain and Circadia oscillators that set physiological ketabolism metabolic Circadian rhythm metabolism in phase with the light—dark cycle. Many evidences show that metabolism and circadian system are tightly interconnected. Peripheral oscillators, such as the liver and adipose tissue, can be shifted by mealtime. By contrast, feeding signals do not affect the master clock under light—dark conditions, although nutritional cues affect its functioning under metabolic challenges, such as calorie restriction and high-fat diet. Circadian desynchronization, such as shift work and chronic jet lag, is now recognized as a determinant of metabolic disturbances.

Video

Restore Your Circadian Rhythm ☀️🔄🌙 Singing Bowl Sound Bath

Author: Dajin

1 thoughts on “Circadian rhythm metabolism

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com