You are on page 1of 14

Pflügers Archiv - European Journal of Physiology

https://doi.org/10.1007/s00424-020-02381-6

INVITED REVIEW

Metabolic implications of circadian disruption


Narjis Fatima 1 & Sobia Rana 1

Received: 23 January 2020 / Revised: 16 April 2020 / Accepted: 17 April 2020


# Springer-Verlag GmbH Germany, part of Springer Nature 2020

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

Keywords Circadian rhythms . Metabolism . Clock genes . Shift-work . Gut microbiota . Metabolic disorders

Introduction cellular, biochemical, physiological, and behavioral processes


with changing solar cycles throughout the seasons. The envi-
Since the dawn of life, organisms have undergone evolutionary ronmental cues that synchronize the body’s internal clock with
adaptation as a response to daily environmental cycles resulting the external environment are known as zeitgebers (time givers)
from Earth’s rotation about its axis, in particular diurnal varia- [41, 62, 66, 108]. The activities regulated by circadian rhythms
tions in sunlight and temperature [26]. Many biological func- include sleep-wake cycle, body temperature, blood pressure,
tions such as cellular, physiological, and behavioral processes secretion of hormones, and metabolism [117, 132]. Taking into
exhibit rhythmic fluctuations that follow a 24-h cycle. For in- account the complex relationship between the circadian clock
stance, a rise in heart rate, blood pressure, and body temperature and metabolism, it is perhaps not surprising that disturbed cir-
is seen in the morning, while a decrease in these parameters is cadian rhythms that influence the normal metabolic pathways
observed in the evening. These variations in biological activi- and may lead to various cardio-metabolic disorders including
ties are referred as circadian rhythms and are generated by an obesity, diabetes, metabolic syndrome, and cardiovascular dis-
endogenous timing system, the circadian clock that produces ease. Similarly, as a consequence of altered sleep-wake cycle,
about 24-h rhythms [107]. In organisms as diverse as bacteria disrupted circadian rhythms may have a considerable impact on
and mammals, the circadian clock aligns the oscillations in behavior and work activities [133]. Recent studies have shown
that in mammals, around 80% of protein-coding genes present
Narjis Fatima and Sobia Rana contributed equally to this work. in various tissues and organs show circadian oscillations at
transcriptional level. For example, in baboons, 82% of genes
* Sobia Rana involved in cellular metabolism and other biochemical func-
molecularbiologist1@gmail.com; sobia.rana@iccs.edu tions are expressed in a circadian manner in at least one tissue
1
[96]. In mice, 43% of protein-coding genes show rhythmicity in
Molecular Biology and Human Genetics Laboratory, Dr. Panjwani
Center for Molecular Medicine and Drug Research (PCMD),
their expression [145]. In this review, wewill discuss the mo-
International Center for Chemical and Biological Sciences (ICCBS), lecular mechanism underlying the regulation of circadian clock
University of Karachi, Karachi 75270, Pakistan system. Moreover, we will review the role of clock genes in
Pflugers Arch - Eur J Physiol

regulating metabolism. Afterward, we will discuss how some signifying its role as a central pacemaker or master clock in
behaviors or lifestyles cause misalignment of the circadian synchronizing the various self-sustained clocks present in the
clock. peripheral organs [112]. SCN receives light from the external
environment through the retinohypothalamic tract that en-
trains the central clock to a 24-h cycle [43]. The central clock,
Central and peripheral clocks in turn, synchronizes the peripheral clocks and regulates phys-
iological processes and behavior in a circadian manner [6].
Circadian rhythms in mammals are regulated by a central Although circadian rhythms in the central clock are mainly
clock and various peripheral oscillators (Fig. 1). Central clock coupled with light-dark cycle, the most important zeitgeber
is located in suprachiasmatic nucleus (SCN) of anterior hypo- in peripheral organs such as liver, pancreas, and heart is food.
thalamus, while peripheral clocks exist in different tissues and Restricted feeding in mice was observed to alter rhythmic
organs of the body including brain, liver, adipose tissue, and gene expression in peripheral clocks without affecting the
muscles [9, 108]. Surgical removal of SCN in rats was found central clock and to uncouple the peripheral clocks from the
to abolish the circadian rhythms in peripheral organs central one [6, 33].

Fig. 1 Central and peripheral clocks in human. Suprachiasmatic nucleus tissues and organs (liver, adipose tissue, muscles, pancreas, heart, etc.)
(SCN) receives light signals from the external environment through in a circadian manner. Light-dark cycle is important zeitgeber for the
retinohypothalamic tract and entrains the central clock to a 24-h cycle. central clock, while food is the most dominant cue for the peripheral
Central clock synchronizes the peripheral clocks by humoral and neuro- clocks
nal signals and regulates various physiological processes in peripheral
Pflugers Arch - Eur J Physiol

Circadian clock at molecular level response element) that is present in the promoter region of
Bmal1. REV-ERBα binds to the promoter region of the
At molecular level, the circadian system is regulated by tran- Bmal1 gene and acts as a negative regulator for inhibiting its
scriptional and translational feedback loops of circadian clock transcription. On the other end, RORα protein initiates the
genes (Fig. 2). Core circadian clock genes include Clock, transcription of Bmal1 [53]. This additional feedback loop
Bmal1, Per (Per1 and Per2), and Cry (Cry1 and Cry2) genes. (REV-ERBα and RORα) links both negative (PER and
Both Clock and Bmal1 genes act as positive regulators, and CRY) and positive (BMAL1 and CLOCK) feedback loops.
their protein products dimerize to form a CLOCK-BMAL1 In addition to core clock genes, the circadian system also
complex that initiates transcription of Per and Cry genes. involves clock-controlled genes. The products of core clock
After reaching a significant concentration, protein products genes act as transcriptional factors for directly or indirectly
of Per and Cry genes act as negative regulators and inhibit regulating the expression of clock-controlled genes [107],
transcription of positive regulators [108, 126]. An additional while clock-controlled genes are involved in the regulation
negative feedback loop consisting of Rev-erbα and RORα of various biological processes including metabolism [97]. A
genes is also involved in the regulation of circadian rhythms. recent study involving Rev-erbα/β double-knockout mouse
CLOCK-BMAL1 heterodimer initiates the transcription of embryonic stem cells revealed that REV-ERBα/β has a sig-
Rev-erbα and RORα genes whereas protein products of Cry nificant role in the regulation of mammalian circadian clock
and Per genes inhibit this transcription in a circadian manner via controlling the expression of various clock-controlled
[106]. REV-ERBα and RORα compete for the Bmal1 gene genes [64]. Therefore, disruption of core clock genes may
binding element RORE (retinoic acid-related orphan receptor alter the expression of genes involved in various physiological

Fig. 2 Molecular framework of circadian clock. BMAL1-CLOCK het- of the Bmal1 gene by binding on the RORE site in the promoter region of
erodimer binds to E-Boxes in promoter regions and regulates expression Bmal1. PPARα binds to Bmal1 gene and regulates its transcription.
of Cry1/2, Per1/2, Rev-erbα, RORα, and various clock-controlled genes. BMAL1, brain and muscle ARNT-like 1; CRY1/2, cryptochrome1/2;
The protein products of Cry1/2 and Per1/2 genes act as negative regula- PER1/2, period1/2; REV-ERBs and RORs, retinoic acid related orphan
tors. When CRY1/2 and PER1/2 proteins reach a particular concentration nuclear receptors; RORE, retinoic acid-related orphan nuclear receptor
in cytoplasm, they form a heterodimer, which translocates to the nucleus response elements; PPARα, peroxisome proliferator-activated receptor
and interacts with the BMAL1-CLOCK and inhibits their transcription. alpha; PPRE, peroxisome proliferator response element; DBP, D-site
REV-ERBα acts as a negative regulator and inhibits Bmal1 transcription binding protein
whereas RORα acts as a positive regulator and initiates the transcription
Pflugers Arch - Eur J Physiol

processes such as metabolic activities suggesting a significant loss of diurnal variations in food intake and other metabol-
role of the circadian clock in regulation of key metabolic ic parameters such as blood pressure, heart rate, and heat
genes [86]. production [142]. PPARδ connects the circadian clock with
diurnal variations in body temperature [140]. Similarly,
Sirtuin genes are also involved in the regulation of clock-
Role of circadian clock in regulating controlled genes by controlling gene expression of core
metabolism clock genes Bmal1 and Clock [80]. Sirt1, Sirt3, and Sirt6
genes have a significant role in the regulation of circadian
The circadian clock regulates metabolism in mammals by clock, and these genes also link circadian clock with met-
interacting with multiple physiological, genetic, and envi- abolic homeostasis [88]. Sirt1 gene regulates metabolic
ronmental factors. Clock genes generate daily rhythms of processes such as glucose and lipid metabolism, insulin
sleep/wake cycle, feeding-fasting cycle, and metabolism in secretion, and fatty acid oxidation in mitochondria [147].
response to exogenous factors [90]. Circadian system reg- Sirt3 gene expression was found to be reduced up to 50%
ulates several metabolic processes including glucose and in the mouse model of diabetes whereas calorie restriction
cholesterol metabolism [57]. Regulation of other physio- led to increased expression of Sirt3 [67]. A high-fat diet in
logical functions such as blood pressure and heart rate is mice disturbed the expression of clock genes in different
also carried out in a circadian manner [25]. In addition, tissues by suppressing SIRT1 protein [122].
plasma levels of hormones involved in metabolic processes
such as glucagon and leptin display circadian oscillations
[69, 110]. Similarly, glucose transporters and glucagon re-
ceptors exhibit circadian rhythms [17]. Any disturbance in Circadian disruption and metabolic anomalies
the interaction of the circadian system with environmental
factors has been shown to have harmful effects on metab- Disruption of circadian rhythms as a consequence of itera-
olism, physiology, and behavioral processes of the organ- tive alterations in lifestyle may affect well-being and normal
ism. Besides, mutations in circadian clock genes may lead physiological processes in humans (Fig. 3). Disrupted cir-
to various metabolic disorders [90]. Genes involved in the cadian rhythms represent a well-known risk factor for pro-
metabolism of nutrients are expressed rhythmically. For moting various cardio-metabolic disorders like metabolic
instance, genes encoding enzymes involved in the regula- syndrome, diabetes, and cardiovascular disease [84].
tion of glucose and cholesterol metabolism show rhythmic Metabolic syndrome is the presence of at least three or more
expression [44]. Nuclear receptor genes involved in the of the five metabolic anomalies such as central obesity, hy-
regulation of metabolism such as PPARs, Rev-Erbs, and perglycemia, insulin resistance, dyslipidemia, and hyper-
RORs have also been found to display circadian oscilla- tension [61]. Metabolic syndrome increases the rate of mor-
tions in their expression [22, 73]. Both Rev-Erbs and tality worldwide as it leads to development of diabetes and
RORs connect the circadian clock with the metabolic pro- cardiovascular diseases [137]. Diabetes is a major metabolic
cesses [37, 36]. REV-ERBα/β regulates lipid metabolism disorder estimated to cause 380 million deaths worldwide
in the liver. Declined levels of REV-ERBα/β lower the by 2025 [42]. Majority of deaths in diabetic patients are due
expression of several genes involved in metabolism and to cardiovascular diseases [39]. Circadian misalignment
cause hepatic steatosis [15]. Likewise, PPAR genes are in- (shift work and eating at night) led to increased arterial
volved in energy homeostasis and regulation of lipid me- blood pressure, reduced plasma levels of leptin, increased
tabolism. PPARα is an important link between circadian glucose and insulin levels, and a significant reduction in
clock and metabolism [24]. It plays an important role in sleeping efficiency in humans [77, 114]. There is a great
the regulation of circadian rhythms and is involved in the variability in the way humans manage their sleep and wake-
regulation of lipid and protein metabolism [116]. fulness during the 24-h day. Many studies have reported a
Moreover, it also activates metabolic pathways in response strong association of evening-chronotype (the tendency of a
to starvation such as ketogenesis and hepatic fatty acid person to wake up late and sleep late) with the metabolic
oxidation [78]. PPARα expression is regulated by the disease [144]. Moreover, a recent study has reported that
CLOCK-BMAL1 heterodimer protein complex. PPARα, morning persons are mentally healthy as compared with
in turn, binds to the Bmal1 gene and regulates its transcrip- evening persons, but no evidence is found for the causal link
tion [116]. Bmal1 knockout and Clock mutant mice have of chronotype with the risk of being diabetic or obese [68].
been observed to have diminished expression of PPARα in Further studies are required to explore whether metabolic
the liver as compared with wild-type mice [18, 98]. PPARγ disorders are more significantly correlated with misalign-
gene is involved in adipogenesis and lipogenesis in adi- ment of circadian rhythms as a result of shift work or eating
pose tissues [82]. Deletion of PPARγ gene in mice led to at night as compared with the chronotype of individuals.
Pflugers Arch - Eur J Physiol

Fig. 3 Implications of iterative alterations of life style and mutations of clock genes have harmful effects on metabolism. Disrupted circadian
circadian clock genes for metabolic health. Disturbances in interaction of system causes various cardio-metabolic anomalies such as obesity, dia-
the circadian system with environmental factors disrupt the circadian betes, and cardiovascular disease. HDL-C, high density lipoprotein
clock. These circadian misalignments such as eating at night, exposure cholesterol
to artificial light at night, night shift work, and mutations in circadian

Clock genes’ mutants and metabolism gene mutation in mice was reported to result in the develop-
ment of diabetes [99]. Moreover, mutations in mice’s core
The disruption of circadian rhythms owing to either complete clock genes were observed to alter the expression of key genes
ablation or mutations of clock genes in animal models has involved in regulation of liver’s glucose and lipid metabolism
provided evidence for the association of circadian rhythms [74, 75]. It was shown in mice that pancreas specific Bmal1
with risk of developing various metabolic disorders (Fig. 1, gene expression is necessary for glucose metabolism, and its
Table 1). The circadian clock mutant mice were observed to deletion can lead to altered insulin secretion [111]. Likewise,
develop metabolic anomalies such as hyperphagia, obesity, liver-specific deletion of Bmal1 in mice was shown to result in
hyperglycemia, hyperlipidemia, hyperleptinemia, and hypoglycemia during the fasting phase, altered circadian ex-
hypoinsulinemia with accumulation of fats in the liver [44, pression of some liver genes involved in glucose metabolism
104, 131]. In Bmal1 knockout mice, increased levels of circu- and impaired clearance of circulating glucose, thus, suggest-
lating triglycerides, cholesterol, and free fatty acids resulted in ing a role of Bmal1 in the regulation of circadian homeostasis
deposition of fat in organs other than adipose tissues including of glucose [75]. Moreover, skeletal muscle-specific Bmal1
the liver and muscles [118]. An accumulating amount of stud- deletion in mice led to reduced glucose uptake and decreased
ies has shown that disruption of clock genes has a significant expression of glucose transporter GLUT4. Besides, the activ-
role in the development of diabetes. For instance, Bmal1 de- ity of key enzymes involved in glycolysis in skeletal muscles
ficient mice have been found to exhibit altered glucose me- was also found to be diminished and led to impaired glucose
tabolism and impaired insulin signaling leading to develop- metabolism in muscle tissues. Systematic homeostasis of glu-
ment of insulin resistance and obese phenotype [120]. In ad- cose was also disturbed in these mice [56]. Likewise, Bmal1
dition, Clock and Bmal1 mutant mice were found to have gene in mice adipose tissue was shown to be involved in lipid
impaired insulin secretion leading to the development of dia- metabolism and regulation of body temperature, fatty acid
betes [85]. Similarly, Per2 knockout mice have been observed oxidation, and energy homeostasis [19]. These pieces of evi-
to have aberrant blood insulin levels due to its reduced clear- dence suggest that peripheral clocks play a significant role in
ance as compared with wild-type mice [146]. In addition, Cry the regulation of glucose. Furthermore, both Clock mutant and
Pflugers Arch - Eur J Physiol

Table 1 Metabolic implications reported for circadian clock genes’ knockout/mutant organisms

Knockout/mutation Tissue-specific/whole body Phenotype Ref.

Bmal1−/− Liver-specific Elevated serum triglycerides (TGs), increased hepatic lipid, [21, 75]
and impaired clearance of circulating glucose
Bmal1−/− Whole-body Increased levels of free fatty acids, TGs, and [118]
cholesterol. Deposition of fat in adipose tissues,
liver, and muscles
Bmal1−/− Whole-body Glucose intolerance and reduced insulin secretion [120]
leading to insulin resistance and obesity
Bmal1−/− Whole-body Glucose intolerance, reduced insulin secretion, [85]
defective size, and multiplication of pancreatic islets
Bmal1−/− Smooth muscle-specific Disturbed rhythms of blood pressure [139]
Bmal1−/− Whole-body Elevated cholesterol and triglycerides, impaired [2]
vascular remodeling, vascular injury, and
endothelial dysfunction
Bmal1−/− Whole-body Disturbed rhythms of blood pressure and heart rate [31]
Bmal1−/− Adipose tissue-specific Altered feeding behavior and obese phenotype [104]
Bmal1−/− Pancreas specific Altered secretion of insulin [111]
Bmal1−/− Skeletal muscle-specific Impaired glucose metabolism in muscles and disturbed [56]
homeostasis of glucose in the whole body
Clock−/− Whole-body Slight weight gain [35]
Clock mutant Whole-body Glucose intolerance, delayed insulin signaling, defective [85]
size, and multiplication of pancreatic islets
Clock mutant Whole-body Vascular injury, thrombosis, and endothelial dysfunction [2]
Clock mutant Whole-body Disturbed circadian rhythms of blood pressure and heart rate [31]
Cry1−/−, Cry2−/− Whole-body Circadian variation in heart rate [89]
Cry1−/−, Cry2−/− Whole-body Weight gain and impaired glucose metabolism [21]
Cry1−/−, Cry2−/− Whole-body High-fat diet-induced obesity and hyperinsulinemia. [11]
Increased fatty acids storage in adipose tissues
Cry1 mutant Whole-body Diabetes [99]
Per2−/−/, Cry1−/− Whole-body Decreased energy metabolism [58]
Per2−/− Whole-body Circadian variation in feeding behaviorProne to obesity [141]
Per2−/− Whole-body Altered lipid profile, reduced levels of triacylglycerol, [51]
and increased adipocyte differentiation
Per2−/− Whole-body Reduced clearance of insulin [146]
Per1,2,3−/− Whole-body Prone to obesity [32]
PPARγ−/− Whole-body Circadian variation in food intake, blood pressure, heart [142]
rate, and heat production
Rev-erbα/β−/− Liver-specific Hepatic steatosis and dyslipidemia [21]

Bmal1 knockout mice were found to develop pathological were seen to be more susceptible to diet-induced obesity as
vascular remodeling and endothelial dysfunction in response compared with wild-type mice. Additionally, increased secre-
to vascular injury [2]. Also, smooth muscle Bmal1 deletion in tion of insulin and increased fatty acid storage in adipose
mice dampened the amplitude of blood pressure circadian tissues were also observed in these mice [11]. Likewise,
rhythm and decreased blood pressure. Similarly, altered circa- Clock deficient mice showed slight weight gain as compared
dian rhythms for blood pressure and heart rate were also seen with wild-type mice, but results were not statistically signifi-
in Bmal1 knockout, Cry1 and Cry2 knockout, and Clock mu- cant [35]. PER2 controls lipid metabolism and adipogenesis
tant mice [31, 89]. However, further mechanistic studies are by regulating the expression of PPARγ and PPARγ-target
needed to better comprehend the clock genes’ role in periph- genes. Mice lacking Per2 gene had variation in feeding be-
eral organs along with the molecular pathways and pathogenic havior and developed obesity. Altered lipid profile and severe
mechanism of cardiovascular disorders related to circadian reduction of triacylglycerol levels were observed in Per2
disruption of blood pressure. [30, 130, 139]. The products of knockout mice. Furthermore, fibroblasts of Per2 knockout
core clock genes Per2 and Cry1 were reported to control the mice were found to have enhanced adipocyte differentiation
expression of genes involved in the regulation of energy me- [51, 141]. Adipose tissue-specific Bmal1 knockout mice were
tabolism in zebrafish [58]. Also, the Cry gene knockout mice found to have altered feeding behavior and obesity [104]. In
Pflugers Arch - Eur J Physiol

the same way, Per1,2,3 knockout mice were found more importance of rodent models for future mechanistic studies
prone to obesity as compared with wild-type mice suggesting on shift work-related metabolic disorders. On the other hand,
a significant role of Per genes in the regulation of body weight conditions of shift work in humans are variable and not well-
[32]. In a similar manner, the Clock gene polymorphisms in defined. Therefore, it is difficult to design ideal animal
humans were found to be associated with metabolic syndrome models; thus, there is a need for developing a standard animal
[115]. Additionally, expression levels of circadian clock genes model having appropriate characteristics of shift work for fu-
in humans were also observed to be negatively associated with ture research [52, 100].
parameters of metabolic syndrome. For instance, expression
of Per2 in visceral adipose tissues was inversely associated
with waist circumference. Likewise, significant inverse asso- Impact of shift work on human metabolic
ciation of Bmal1, Cry, and Per2 gene expression levels with health
total and low-density lipoprotein cholesterol was observed
[49]. However, more studies are warranted to investigate the Disruption of circadian rhythms due to shift work is associated
tissue-specific role of circadian clock genes in lipid and glu- with obesity, diabetes, metabolic syndrome, and cardiovascu-
cose metabolism. lar illnesses [3, 4, 34]. The circadian clock is disrupted by
insufficient sleep and leads to arrhythmic physiological pro-
cesses, altered glucose metabolism, and overweight/obesity in
Metabolic status of rodent models of night humans [12, 92]. Altered sleep-wake cycle as a consequence
shift work of night shift work is associated with disruption of the internal
clock that leads to altered metabolic homeostasis [121].
Rodent models that mimic the night shift work schedule in Synthesis of melatonin occurs during the darkness that results
humans are being used to better understand the system behind in increased sleep tendency in humans [105]. Shift work and
the synchronization of circadian clock with the geophysical exposure to artificial lights at night resulted in an altered syn-
time [38]. Rodent model of shift work (male Wistar rats) were thesis of melatonin [16, 50]. Reduced levels of melatonin in
subjected to feeding and forced activity during their normal humans give rise to disturbed sleep-wake cycle, decreased
sleep period. These rats exhibited a loss of synchronization insulin sensitivity, and glucose intolerance, eventually leading
between SCN and liver. Moreover, expression levels of core to obesity [28]. Shift workers working at night and sleeping at
clock genes (Per1, Clock, and Bmal1) were altered, and the day time were found to have reduced energy expenditure
expression of Per2 lost rhythmicity in the liver. Furthermore, resulting in weight gain and obesity [93]. Glucose intolerance
clock-controlled genes involved in the regulation of metabolic in shift workers as a consequence of disrupted circadian
processes lost synchronization with circadian clock genes in rhythms increases the risk of diabetes [95]. Night shift work
rat liver [113]. Mice exposed to a 20-h light-dark cycle, which has also been found to be associated with increased levels of
was incompatible with their internal circadian clock, were cholesterol and triglycerides, low levels of HDL cholesterol,
found to have disrupted circadian clock. These conditions increased BMI (body mass index; a measure of obesity), and
resulted in negative effects on physiology including increased increased blood pressure ultimately leading to cardiovascular
levels of insulin and leptin, altered body temperature, weight diseases [14, 34, 54, 71]. Shift workers have been observed to
gain, and obesity [70]. In another mouse model of shift work, be more prone to the development of abdominal obesity [123].
early pathophysiological effects of shift work on molecular Years of shift work exposure and age of workers were found to
circadian machinery were observed. Circadian expression of be positively associated with an increase in BMI in individuals
core clock genes and clock-controlled genes was found to be working at night-shift. However, in individuals working dur-
severely disrupted in the liver, while Bmal1, Per1, Dbp, and ing the day-shift, the age of workers was found to be positive-
Rev-erbα genes’ expression was disrupted moderately in ly associated with the increase in BMI, but years of work
SCN. These early pathophysiological effects of shift work exposure were not associated with an increase in BMI [102].
may precede metabolic disorders. When these mice were sub- Shift workers were reported to have higher risk of having
jected to time-restricted feeding, improvement in disrupted obese phenotype and poor health [8, 65, 72, 83, 138].
glucose and lipid metabolism was observed suggesting that According to a study, energy intake between night and day
minimizing food intake during the night in shift workers can workers were similar suggesting that increasing rates of obe-
reduce the risk of metabolic disorders [10]. Poor sleep quality sity in shift workers were associated with circadian misalign-
along with having a high-fat diet led to impaired glucose tol- ment caused by the timing of food intake and variation in
erance in mice, while sleep recovery reversed the altered glu- metabolism at night. However, it remained uncertain whether
cose metabolism in mice exposed to a high-fat diet [59]. this redistribution of food and alteration in food preference at
Studies exploring metabolic disorders in shift workers using night is associated with the higher risk of obesity seen in the
rodents as a model of shift work have highlighted great shift workers [13]. A study reports that shift workers have
Pflugers Arch - Eur J Physiol

17% and 27% higher risk of developing obese phenotype and recent studies, light can also synchronize the peripheral clocks
diabetes, respectively. Additionally, association of shift work in SCN-independent manner [63]. Zebrafish cell lines and
with an increased risk of having cardiovascular diseases has embryos have also been reported as light-responsive, and light
also been reported [125]. Another study reports the impact of can directly entrain the circadian clock in these cell lines. [58,
disturbed sleep-wake cycle and fasting-feeding cycle on cir- 127, 128]. Nevertheless, further research is still required to
culating metabolites in human subjects under controlled con- fully elucidate the underlying mechanism by which the central
ditions. In this study, many metabolites lost or reversed their circadian clock synchronizes the peripheral clocks in light-
rhythmicity during simulated shift work. In addition, a great dependent manner.
majority of metabolites linked to nutrient metabolism
uncoupled their rhythmicity with SCN and were synchronized
with sleep-wake and eating schedule during shift work. [119]. Implications of irregular food timings
However, the mechanism underlying circadian misalignment on metabolism
with the external environment and to which extent dysregula-
tion of peripheral clocks is related to irregular working sched- The role of eating habits in regulation of circadian rhythm’s
ule is still unknown. Prospective cohort studies on large scale period has been indicated in several studies [5, 84]. Food is
are still needed to know the consequences of shift work and its one of the important zeitgebers that synchronizes the circadian
association with obesity and other metabolic disorders. clock [44]. Irregular timings of food intake in humans may
lead to disruption of metabolic homeostasis [90]. Feeding pat-
terns in mice were found to be coordinated mainly by SCN
Effect of exposure to light at night that regulate secretion of feeding-related hormones in a circa-
on metabolic well-being dian manner including ghrelin and leptin [69]. Leptin levels
were found low in individuals eating at night time that might
It is well-established fact that light can regulate circadian contribute to increased intake of food at night [94]. However,
clock that is involved in the regulation of metabolic pathways. some studies showed that leptin levels were similar between
Exposure to artificial lights at night is suggested to cause individuals eating at night (consuming about half of the food
disruption of circadian rhythms in humans, which in turn leads after the evening meal and awoke at night three times per
to deregulation of metabolic homeostasis and eventually met- week for taking food) and healthy controls (consuming less
abolic disorders like overweight and obesity [44, 91]. Mice than a quarter of food after dinner and did not wake at night for
exposed to constant 24-h bright light displayed glucose intol- taking food) [1, 55]. Disrupted circadian rhythms in
erance and weight gain without increasing the calorie intake as Drosophila (fruit fly) were reported to result in muscle dys-
compared with mice exposed to normal light-dark cycle [45]. function and obesity. On the other hand, metabolic glitches
Mice in response to 24-h continuous light exposure were ob- related to obesity were improved when the diet was restricted
served to have increased food intake and decreased energy in both clock mutant and wild type obese flies [134]. In circa-
expenditure. Weight gain was also more rapid in mice exposed dian clock mutant mice (whole-body Cry1, Cry2, and in liver-
to constant light as compared with mice having a high-fat diet. specific Bmal1 and Rev-erbα/β knockout mice), time-
Thus, indicating the role of constant light exposure in circadi- restricted feeding (TRF) prevented obesity and obesity-
an dysregulation of metabolism leading to insulin resistance related metabolic problems such as glucose intolerance, fatty
and obesity [29]. Mice exposed to constant dim light at night liver disease, and dyslipidemia as compared with clock mutant
developed altered food timings, increased white adipose tissue mice that developed obesity and related metabolic disorders as
mass, gained more body weight, and developed glucose intol- a consequence of free-feeding [21]. TRF was found to reduce
erance as compared with mice exposed to normal light-dark metabolic diseases (hepatic steatosis and hypercholesterol-
cycle. However, when these mice were kept back in a normal emia) arising as a result of various high-fat diets in mice.
light-dark cycle, the glucose clearance ability of mice was Also, TRF was observed to reverse the development of met-
recovered, and their food intake timings were also improved abolic disorders in mice with pre-existing obesity and type II
[47]. Similarly, male mice fed a high-fat diet and exposed to diabetes [20]. Moreover, TRF in mice resulted in rapid weight
constant dim light at night exhibited exaggerated weight gain loss, restoration of glucose tolerance, and decreased severity
as compared with mice exposed to normal light-dark cycle of hepatic steatosis. Besides, mice subjected to TRF were
and fed a normal diet. In addition, impaired glucose metabo- physically more active as compared with mice having free-
lism, abnormal insulin secretion, altered feeding timing, and feeding [27]. On the other hand, when juvenile mice having
peripheral inflammation were also observed in mice having a TRF in childhood period were subjected to free feeding in
high-fat diet and exposed to constant dim light at night [46]. In adulthood, they were found to have severe irreparable dam-
mammals, SCN receives light through the retinohypothalamic ages including metabolic disorders, increased body weight,
tract and synchronizes the peripheral clocks, but according to immune system suppression, delayed sexual maturity, and
Pflugers Arch - Eur J Physiol

increased risk of cardiovascular diseases [60]. Early TRF (eat- abundance of fecal microbiota [81]. Moreover, germ-free
ing early in the day) in humans was observed to increase (GF) mice displayed completely different central and hepatic
insulin sensitivity and improve β cell function, and to decrease circadian clock with altered metabolic pathways. In addition,
blood pressure and oxidative stress [124]. Furthermore, TRF the lack of gut microbiota in GF and antibiotic-induced mice
in humans was witnessed to decrease the body weight, lower deregulated the expression of the clock genes (Bmal1, Per1,
the circulating levels of glucose, triglycerides, and low- Per2, and Cry1) in the liver and intestinal epithelial cells [79].
density lipoprotein cholesterol, and to increase the circulating Gut microbes can alter host energy balance, metabolic pro-
levels of high-density lipoprotein cholesterol [109]. However, gramming, and microbial metabolites. Expression levels of
the mechanistic link between feeding patterns and metabolism key genes involved in the absorption of nutrients and metab-
are yet to be explored. According to a recent study, eating at olism in germ-free mice and specific pathogen-free mice were
night was found to be associated with metabolic syndrome in altered in peripheral tissues especially in the intestine when
women and dyslipidemia in both men and women [143]. these mice were conventionalized with fecal microbiota [40,
Abnormal food timings can disrupt the circadian clock partic- 76]. Several studies have reported a clear association of met-
ularly high-calorie diet at night that may lead to metabolic abolic syndrome and obesity with disruption of microbiota
disorders like obesity. Individuals having a high-calorie diet rhythm. Metabolites produced by gut microbiota are impor-
in breakfast easily lost their weight and had improved insulin tant mediators that link gut microbiota with the host clock.
sensitivity as compared with individuals having a high-calorie Thus, disturbance of this circadian network may lead to diet-
diet in dinner [48]. Temporal restriction of food intake in mice induced obesity [79, 135]. GF mice were found to have re-
can alter the phase of rhythmic expression of clock genes duced adiposity, improved glucose tolerance, and insulin sen-
(Per1, Per2, Per3, and Cry1) and clock-controlled genes sitivity as compared with specific pathogen-free mice.
(Rev-erbα, Dbp, and Cyp2a5) in peripheral organs without Moreover, GF mice were resistant to diet-induced obesity
affecting the phase of rhythmic expression of clock genes [7]. Impaired absorption and metabolism of lipids in the small
and clock-controlled genes in SCN, thus, uncoupling the pe- intestine of germ-free mice implied that gut microbiota has an
ripheral clocks from SCN. Unexpected large changes in food important role in the metabolism and absorption of lipids and
intake timings can reset the expression of genes that are can influence host circadian clock. Furthermore, GF mice
expressed in a circadian manner especially in the liver [33]. were partially protected from high fat diet-induced obesity
Nevertheless, further studies are warranted to estimate the role due to decreased dietary lipid digestion and absorption [87].
of meal timing in diurnal secretion of hormones. Gut microbes and circadian rhythms are linked via regulation
of metabolism, but the mechanisms underlying these interac-
tions are not well understood. Mechanistic studies are warrant-
Circadian rhythms, gut microbiota, ed to dissect the molecular mechanism underlying the corre-
and metabolism lation of gut microbiota with the metabolism of the host.

Gut microbiota and circadian clock have an interrelated regu-


latory relationship that plays an important role in metabolic Conclusion
health [101]. A growing body of evidence demonstrates that
gut microbiota (the microorganisms inhabiting the intestine) Organisms have evolved and adapted their biological clock to
play a significant role in the development of insulin resistance, synchronize with the cyclic nature of the environment such as
obesity, and metabolic syndrome [23]. Gut microbiota ex- light/dark cycle, availability of food, and changes in tempera-
hibits diurnal oscillations in both microbial community com- ture. Body’s endogenous clock produces 24-h rhythms in the
position and their metabolic activities. These changes in mi- body. The body’s clock is significantly affected by iterative
crobial rhythms are regulated by the time of eating and the alterations of life style that causes misalignment of internal
type of diet of the host. Circadian rhythms of host organism clock with geophysical time. As circadian clock synchronizes
can alter the composition and activity of the gut microbiota, behavioral and physiological processes in the body with envi-
while gut microbiota can affect the host metabolism [135]. ronmental cues, any disturbance in synchronization of the
Many studies have suggested that gut microbiota may affect circadian system with the external environment is found to
the rhythmic expression of the biological clock of the host have negative consequences and adverse effects on physiolo-
[103]. Similarly, mutations in host circadian clock genes also gy and metabolic processes. Disruption of the clock due to
influence the circadian rhythms of gut microbiota. Per1/2 shift work or modern lifestyles such as inappropriate timing
knockout mice exhibited a near-complete loss of diurnal os- of food intake or disturbance in the sleep-wake cycle has
cillations in the composition and function of commensal bac- harmful effects on health and may lead to metabolic syn-
teria in the intestine [129, 136]. Likewise, Bmal1 knockout drome, obesity, diabetes, and cardiovascular diseases. At pres-
mice showed abolished rhythmicity in composition and ent, research on chronobiology (the study of biological
Pflugers Arch - Eur J Physiol

rhythms) is becoming significant and constantly growing in Metab 304:E1053–E1063. https://doi.org/10.1152/ajpendo.


00512.2012
fields of biology and medicine. Even though the mechanism
12. Bo S, Ciccone G, Durazzo M, Ghinamo L, Villois P, Canil S,
of circadian clock disruption in knockout or mutant animal Gambino R, Cassader M, Gentile L, Cavallo-Perin P (2011)
models does not replicate the conditions of human physiology Contributors to the obesity and hyperglycemia epidemics. A pro-
and metabolism but it provides some understanding of meta- spective study in a population-based cohort. Int J Obes 35:1442–
1449. https://doi.org/10.1038/ijo.2011.5
bolic disorders driven by the circadian clock. Moreover, syn-
13. Bonham MP, Bonnell EK, Huggins CE (2016) Energy intake of
chronizing the feeding-fasting cycle and sleep-wake cycle shift workers compared to fixed day workers: a systematic review
with the timing of external signals can maintain a healthy and meta-analysis. Chronobiol Int 33:1086–1100. https://doi.org/
lifestyle that may in turn prevent metabolic diseases. 10.1080/07420528.2016.1192188
14. Buchvold HV, Pallesen S, Waage S, Bjorvatn BJ (2018) Shift
Furthermore, focusing on the crosstalk between circadian
work schedule and night work load: effects on body mass index
rhythm and metabolism can help us to elucidate the pathogen- – a four-year longitudinal study. Scand J Work Environ Health 44:
esis of circadian clock associated disorders, which in turn, can 251–257. https://doi.org/10.5271/sjweh.3702
provide support in devising new strategies for prevention and 15. Bugge A, Feng D, Everett LJ, Briggs ER, Mullican SE, Wang F,
Jager J, Lazar MA (2012) Rev-erbα and Rev-erbβ coordinately
therapy of these disorders.
protect the circadian clock and normal metabolic function. Genes
Dev 26:657–667. https://doi.org/10.1101/gad.186858.112
16. Burch JB, Yost MG, Johnson W, Allen E (2005) Melatonin, sleep,
and shift work adaptation. J Occup Environ Med 47:893–901.
https://doi.org/10.1097/01.jom.0000177336.21147.9f
References 17. Cailotto C, Lei J, van der Vliet J, van Heijningen C, van Eden CG,
Kalsbeek A, Pévet P, Buijs RM (2009) Effects of nocturnal light
1. Allison KC, Ahima RS, O’Reardon JP, Dinges DF, Sharma V, on (clock) gene expression in peripheral organs: a role for the
Cummings DE, Heo M, Martino NS, Stunkard AJ (2005) autonomic innervation of the liver. PLoS One 4:e5650. https://
Neuroendocrine profiles associated with energy intake, sleep, doi.org/10.1371/journal.pone.0005650
and stress in the night eating syndrome. J Clin Endocrinol 18. Canaple L, Rambaud J, Dkhissi-Benyahya O, Ba R, Tan NS,
Metab 90:6214–6217. https://doi.org/10.1210/jc.2005-1018 Michalik L, Delaunay F, Wahli W, Laudet V (2006) Reciprocal
2. Anea CB, Zhang M, Stepp DW, Simkins GB, Reed G, Fulton DJ, regulation of brain and muscle Arnt-like protein 1 and peroxisome
Rudic RD (2009) Vascular disease in mice with a dysfunctional proliferator-activated receptor α defines a novel positive feedback
circadian clock. Circulation 119:1510–1517. https://doi.org/10. loop in the rodent liver circadian clock. Mol Endocrinol 20:1715–
1161/CIRCULATIONAHA.108.827477 1727. https://doi.org/10.1210/me.2006-0052
3. Antunes LC, Levandovski R, Dantas G, Caumo W, Hidalgo MP 19. Castro C, Briggs W, Paschos GK, FitzGerald GA, Griffin JL
(2010) Obesity and shift work: chronobiological aspects. Nutr Res (2015) A metabolomic study of adipose tissue in mice with a
Rev 23:155–168. https://doi.org/10.1017/S0954422410000016 disruption of the circadian system. Mol BioSyst 11:1897–1906.
4. Ashby T, Louis M (2019) Circadian misalignment and cardiovas- https://doi.org/10.1039/c5mb00032g
cular risk. Cardiovasc Innov Appl 3:435–440. https://doi.org/10. 20. Chaix A, Zarrinpar A, Miu P, Panda S (2014) Time-restricted
15212/CVIA.2017.0070 feeding is a preventative and therapeutic intervention against di-
verse nutritional challenges. Cell Metab 20:991–1005. https://doi.
5. Asher G, Sassone-Corsi P (2015) Time for food: the intimate
org/10.1016/j.cmet.2014.11.001
interplay between nutrition, metabolism, and the circadian clock.
21. Chaix A, Lin T, Le HD, Chang MW, Panda S (2019) Time-
Cell 161:84–92. https://doi.org/10.1016/j.cell.2015.03.015
restricted feeding prevents obesity and metabolic syndrome in
6. Astiz M, Heyde I, Oster H (2019) Mechanisms of communication
mice lacking a circadian clock. Cell Metab 29:303–319.e304.
in the mammalian circadian timing system. Int J Mol Sci 20:343.
https://doi.org/10.1016/j.cmet.2018.08.004
https://doi.org/10.3390/ijms20020343
22. Chaudhari A, Gupta R, Makwana K, Kondratov R (2017)
7. Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI (2007) Circadian clocks, diets and aging. Nutr Healthy Aging 4:101–
Mechanisms underlying the resistance to diet-induced obesity in 112. https://doi.org/10.3233/NHA-160006
germ-free mice. Proc Natl Acad Sci U S A 104:979–984. https:// 23. Chen X, Devaraj S (2018) Gut microbiome in obesity, metabolic
doi.org/10.1073/pnas.0605374104 syndrome, and diabetes. Curr Diab Rep 18:129. https://doi.org/10.
8. Balieiro LCT, Rossato LT, Waterhouse J, Paim SL, Mota MC, 1007/s11892-018-1104-3
Crispim CA (2014) Nutritional status and eating habits of bus 24. Chen L, Yang G (2014) PPARs integrate the mammalian clock
drivers during the day and night. Chronobiol Int 31:1123–1129. and energy metabolism. PPAR Res 2014:6–6. https://doi.org/10.
https://doi.org/10.3109/07420528.2014.957299 1155/2014/653017
9. Balsalobre A (2002) Clock genes in mammalian peripheral tis- 25. Chen L, Yang G (2015) Recent advances in circadian rhythms in
sues. Cell Tissue Res 309:193–199. https://doi.org/10.1007/ cardiovascular system. Front Pharmacol 6:71–71. https://doi.org/
s00441-002-0585-0 10.3389/fphar.2015.00071
10. Barclay JL, Husse J, Bode B, Naujokat N, Meyer-Kovac J, 26. Chung S, Son GH, Kim K (2011) Circadian rhythm of adrenal
Schmid SM, Lehnert H, Oster H (2012) Circadian desynchrony glucocorticoid: its regulation and clinical implications. Biochim
promotes metabolic disruption in a mouse model of shiftwork. Biophys Acta (BBA) - Mol Basis Dis 1812:581–591. https://doi.
PLoS One 7:e37150. https://doi.org/10.1371/journal.pone. org/10.1016/j.bbadis.2011.02.003
0037150 27. Chung H, Chou W, Sears DD, Patterson RE, Webster NJG, Ellies
11. Barclay JL, Shostak A, Leliavski A, Tsang AH, Jöhren O, Müller- LG (2016) Time-restricted feeding improves insulin resistance and
Fielitz H, Landgraf D, Naujokat N, van der Horst GT, Oster H hepatic steatosis in a mouse model of postmenopausal obesity.
(2013) High-fat diet-induced hyperinsulinemia and tissue-specific Metabolism 65:1743–1754. https://doi.org/10.1016/j.metabol.
insulin resistance in cry-deficient mice. Am J Physiol Endocrinol 2016.09.006
Pflugers Arch - Eur J Physiol

28. Cipolla-Neto J, Amaral FG, Afeche SC, Tan DX, Reiter RJ (2014) 45. Fonken LK, Workman JL, Walton JC, Weil ZM, Morris JS, Haim
Melatonin, energy metabolism, and obesity: a review. J Pineal Res A, Nelson RJ (2010) Light at night increases body mass by
56:371–381. https://doi.org/10.1111/jpi.12137 shifting the time of food intake. Proc Natl Acad Sci U S A 107:
29. Coomans CP, van den Berg SA, Houben T, van Klinken JB, van 18664–18669. https://doi.org/10.1073/pnas.1008734107
den Berg R, Pronk AC, Havekes LM, Romijn JA, van Dijk KW, 46. Fonken LK, Lieberman RA, Weil ZM, Nelson RJ (2013) Dim
Biermasz NR, Meijer JH (2013) Detrimental effects of constant light at night exaggerates weight gain and inflammation associated
light exposure and high-fat diet on circadian energy metabolism with a high-fat diet in male mice. Endocrinology 154:3817–3825.
and insulin sensitivity. FASEB J 27:1721–1732. https://doi.org/10. https://doi.org/10.1210/en.2013-1121
1096/fj.12-210898 47. Fonken LK, Weil ZM, Nelson RJ (2013) Dark nights reverse
30. Crnko S, Du Pré BC, Sluijter JPG, Van Laake LW (2019) metabolic disruption caused by dim light at night. Obesity
Circadian rhythms and the molecular clock in cardiovascular bi- (Silver Spring) 21:1159–1164. https://doi.org/10.1002/oby.20108
ology and disease. Nat Rev Cardiol 16:437–447. https://doi.org/ 48. Garaulet M, Gomez-Abellan P (2014) Timing of food intake and
10.1038/s41569-019-0167-4 obesity: a novel association. Physiol Behav 134:44–50. https://
31. Curtis AM, Cheng Y, Kapoor S, Reilly D, Price TS, Fitzgerald GA doi.org/10.1016/j.physbeh.2014.01.001
(2007) Circadian variation of blood pressure and the vascular re- 49. Gómez-Abellán P, Hernández-Morante JJ, Luján JA, Madrid JA,
sponse to asynchronous stress. Proc Natl Acad Sci U S A 104: Garaulet M (2007) Clock genes are implicated in the human met-
3450–3455. https://doi.org/10.1073/pnas.0611680104 abolic syndrome. Int J Obes 32:121–128. https://doi.org/10.1038/
32. Dallmann R, Weaver DR (2010) Altered body mass regulation in sj.ijo.0803689
male mPeriod mutant mice on high-fat diet. Chronobiol Int 27: 50. Gooley JJ, Chamberlain K, Smith KA, Khalsa SBS, Rajaratnam
1317–1328. https://doi.org/10.3109/07420528.2010.489166 SMW, Van Reen E, Zeitzer JM, Czeisler CA, Lockley SW (2011)
33. Damiola F, Le Minh N, Preitner N, Kornmann B, Fleury-Olela F, Exposure to room light before bedtime suppresses melatonin onset
Schibler U (2000) Restricted feeding uncouples circadian oscilla- and shortens melatonin duration in humans. J Clin Endocrinol
tors in peripheral tissues from the central pacemaker in the supra- Metab 96:E463–E472. https://doi.org/10.1210/jc.2010-2098
chiasmatic nucleus. Genes Dev 14:2950–2961. https://doi.org/10. 51. Grimaldi B, Bellet MM, Katada S, Astarita G, Hirayama J, Amin
1101/gad.183500 RH, Granneman JG, Piomelli D, Leff T, Sassone-Corsi P (2010)
34. Di Lorenzo L, De Pergola G, Zocchetti C, L'Abbate N, Basso A, PER2 controls lipid metabolism by direct regulation of PPARγ.
Pannacciulli N, Cignarelli M, Giorgino R, Soleo L (2003) Effect Cell Metab 12:509–520. https://doi.org/10.1016/j.cmet.2010.10.
of shift work on body mass index: results of a study performed in 005
319 glucose-tolerant men working in a southern Italian industry. 52. Guerrero-Vargas NN, Espitia-Bautista E, Buijs RM, Escobar C
Int J Obes 27:1353–1358. https://doi.org/10.1038/sj.ijo.0802419 (2018) Shift-work: is time of eating determining metabolic health?
35. Dubrovsky YV, Samsa WE, Kondratov RV (2010) Deficiency of Evidence from animal models. Proc Nutr Soc 77:199–215. https://
circadian protein CLOCK reduces lifespan and increases age- doi.org/10.1017/S0029665117004128
related cataract development in mice. Aging 2:936–944. https:// 53. Guillaumond F, Dardente H, Giguère V, Cermakian N (2005)
doi.org/10.18632/aging.100241 Differential control of Bmal1 circadian transcription by REV-
36. Duez H, Staels B (2008) The nuclear receptors rev-erbs and RORs ERB and ROR nuclear receptors. J Biol Rhythm 20:391–403.
integrate circadian rhythms and metabolism. Diab Vasc Dis Res 5: https://doi.org/10.1177/0748730405277232
82–88. https://doi.org/10.3132/dvdr.2008.0014 54. Ha M, Park J (2005) Shiftwork and metabolic risk factors of car-
37. Duez H, Staels B (2009) Rev-erb-alpha: an integrator of circadian diovascular disease. J Occup Health 47:89–95. https://doi.org/10.
rhythms and metabolism. J Appl Physiol (1985) 107:1972–1980. 1539/joh.47.89
https://doi.org/10.1152/japplphysiol.00570.2009 55. Haraguchi A, Fukuzawa M, Iwami S, Nishimura Y, Motohashi H,
38. Eckel-Mahan K, Sassone-Corsi P (2013) Metabolism and the cir- Tahara Y, Shibata S (2018) Night eating model shows time-
cadian clock converge. Physiol Rev 93:107–135. https://doi.org/ specific depression-like behavior in the forced swimming test.
10.1152/physrev.00016.2012 Sci Rep 8:1081. https://doi.org/10.1038/s41598-018-19433-8
39. Einarson TR, Acs A, Ludwig C, Panton UH (2018) Prevalence of 56. Harfmann BD, Schroder EA, Kachman MT, Hodge BA, Zhang X,
cardiovascular disease in type 2 diabetes: a systematic literature Esser KA (2016) Muscle-specific loss of Bmal1 leads to disrupted
review of scientific evidence from across the world in 2007-2017. tissue glucose metabolism and systemic glucose homeostasis.
Cardiovasc Diabetol 17:83–83. https://doi.org/10.1186/s12933- Skelet Muscle 6:12–12. https://doi.org/10.1186/s13395-016-
018-0728-6 0082-x
40. El Aidy S, Merrifield CA, Derrien M, van Baarlen P, Hooiveld G, 57. Hatanaka F, Matsubara C, Myung J, Yoritaka T, Kamimura N,
Levenez F, Doré J, Dekker J, Holmes E, Claus SP, Reijngoud D-J, Tsutsumi S, Kanai A, Suzuki Y, Sassone-Corsi P, Aburatani H,
Kleerebezem M (2013) The gut microbiota elicits a profound met- Sugano S, Takumi T (2010) Genome-wide profiling of the core
abolic reorientation in the mouse jejunal mucosa during clock protein BMAL1 targets reveals a strict relationship with
conventionalisation. Gut 62:1306–1314. https://doi.org/10.1136/ metabolism. Mol Cell Biol 30:5636–5648. https://doi.org/10.
gutjnl-2011-301955 1128/MCB.00781-10
41. Farhud D, Aryan Z (2018) Circadian rhythm, lifestyle and health: 58. Hirayama J, Alifu Y, Hamabe R, Yamaguchi S, Tomita J,
a narrative review. Iran J Public Health 47:1068–1076 Maruyama Y, Asaoka Y, K-i N, Tamaru T, Takamatsu K,
42. Federation IDJBIDF (2006) Diabetes atlas. Available via Takamatsu N, Hattori A, Nishina S, Azuma N, Kawahara A,
DIALOG. https://www.idf.org/e-library/epidemiology-research/ Kume K, Nishina H (2019) The clock components Period2,
diabetes-atlas/22-atlas-3rd-edition.html Cryptochrome1a, and Cryptochrome2a function in establishing
43. Fisk AS, Tam SKE, Brown LA, Vyazovskiy VV, Bannerman DM, light-dependent behavioral rhythms and/or total activity levels in
Peirson SN (2018) Light and cognition: roles for circadian zebrafish. Sci Rep 9:196. https://doi.org/10.1038/s41598-018-
rhythms, sleep, and arousal. Front Neurol 9. https://doi.org/10. 37879-8
3389/fneur.2018.00056 59. Ho JM, Barf RP, Opp MR (2016) Effects of sleep disruption and
44. Fonken LK, Nelson RJ (2014) The effects of light at night on high fat intake on glucose metabolism in mice.
circadian clocks and metabolism. Endocr Rev 35:648–670. Psychoneuroendocrinology 68:47–56. https://doi.org/10.1016/j.
https://doi.org/10.1210/er.2013-1051 psyneuen.2016.02.024
Pflugers Arch - Eur J Physiol

60. Hu D, Mao Y, Xu G, Liao W, Ren J, Yang H, Yang J, Sun L, Chen 75. Lamia KA, Storch K-F, Weitz CJ (2008) Physiological signifi-
H, Wang W, Wang Y, Sang X, Lu X, Zhang H, Zhong S (2019) cance of a peripheral tissue circadian clock. Proc Natl Acad Sci
Time-restricted feeding causes irreversible metabolic disorders U S A 105:15172–15177. https://doi.org/10.1073/pnas.
and gut microbiota shift in pediatric mice. Pediatr Res 85:518– 0806717105
526. https://doi.org/10.1038/s41390-018-0156-z 76. Larsson E, Tremaroli V, Lee YS, Koren O, Nookaew I, Fricker A,
61. Huang PL (2009) A comprehensive definition for metabolic syn- Nielsen J, Ley RE, Bäckhed F (2012) Analysis of gut microbial
drome. Dis Model Mech 2:231–237. https://doi.org/10.1242/ regulation of host gene expression along the length of the gut and
dmm.001180 regulation of gut microbial ecology through MyD88. Gut 61:
62. Huang R-C (2018) The discoveries of molecular mechanisms for 1124–1131. https://doi.org/10.1136/gutjnl-2011-301104
the circadian rhythm: the 2017 Nobel Prize in Physiology or 77. Lebeche D, Davidoff AJ, Hajjar RJ (2008) Interplay between im-
Medicine. Biom J 41:5–8. https://doi.org/10.1016/j.bj.2018.02. paired calcium regulation and insulin signaling abnormalities in
003 diabetic cardiomyopathy. Nat Clin Pract Cardiovasc Med 5:715–
63. Husse J, Eichele G, Oster H (2015) Synchronization of the mam- 724. https://doi.org/10.1038/ncpcardio1347
malian circadian timing system: light can control peripheral clocks 78. Leone TC, Weinheimer CJ, Kelly DP (1999) A critical role for the
independently of the SCN clock. BioEssays 37:1119–1128. peroxisome proliferator-activated receptor α (PPARα) in the cel-
https://doi.org/10.1002/bies.201500026 lular fasting response: the PPARα-null mouse as a model of fatty
64. Ikeda R, Tsuchiya Y, Koike N, Umemura Y, Inokawa H, Ono R, acid oxidation disorders. Proc Natl Acad Sci U S A 96:7473–
Inoue M, Sasawaki Y, Grieten T, Okubo N, Ikoma K, Fujiwara H, 7478. https://doi.org/10.1073/pnas.96.13.7473
Kubo T, Yagita K (2019) REV-ERBα and REV-ERBβ function as 79. Leone V, Gibbons Sean M, Martinez K, Hutchison Alan L, Huang
key factors regulating mammalian circadian output. Sci Rep 9: Edmond Y, Cham Candace M, Pierre Joseph F, Heneghan Aaron
10171. https://doi.org/10.1038/s41598-019-46656-0 F, Nadimpalli A, Hubert N, Zale E, Wang Y, Huang Y, Theriault B,
65. Itani O, Kaneita Y, Murata A, Yokoyama E, Ohida T (2011) Dinner Aaron R, Musch Mark W, Kudsk Kenneth A, Prendergast
Association of onset of obesity with sleep duration and shift work Brian J, Gilbert Jack A, Chang Eugene B (2015) Effects of diurnal
among Japanese adults. Sleep Med 12:341–345. https://doi.org/ variation of gut microbes and high-fat feeding on host circadian
10.1016/j.sleep.2010.09.007 clock function and metabolism. Cell Host Microbe 17:681–689.
66. Jagannath A, Taylor L, Wakaf Z, Vasudevan SR, Foster RG https://doi.org/10.1016/j.chom.2015.03.006
(2017) The genetics of circadian rhythms, sleep and health. Hum 80. Li X (2013) SIRT1 and energy metabolism. Acta Biochim
Mol Genet 26:R128–R138. https://doi.org/10.1093/hmg/ddx240 Biophys Sin Shanghai 45:51–60. https://doi.org/10.1093/abbs/
67. Jing E, Emanuelli B, Hirschey MD, Boucher J, Lee KY, Lombard gms108
D, Verdin EM, Kahn CR (2011) Sirtuin-3 (Sirt3) regulates skeletal 81. Liang X, Bushman FD, FitzGerald GA (2015) Rhythmicity of the
muscle metabolism and insulin signaling via altered mitochondrial intestinal microbiota is regulated by gender and the host circadian
oxidation and reactive oxygen species production. Proc Natl Acad clock. Proc Natl Acad Sci U S A 112:10479–10484. https://doi.
Sci U S A 108:14608–14613. https://doi.org/10.1073/pnas. org/10.1073/pnas.1501305112
1111308108 82. Liu S, Downes M, Evans RM (2015) Metabolic regulation by
68. Jones SE, Lane JM, Wood AR, van Hees VT, Tyrrell J, Beaumont nuclear receptors. Innovative medicine. Springer, Japan, p 25-37
RN, Jeffries AR, Dashti HS, Hillsdon M, Ruth KS, Tuke MA, 83. Liu Q, Shi J, Duan P, Liu B, Li T, Wang C, Li H, Yang T, Gan Y,
Yaghootkar H, Sharp SA, Jie Y, Thompson WD, Harrison JW, Wang X, Cao S, Lu Z (2018) Is shift work associated with a higher
Dawes A, Byrne EM, Tiemeier H, Allebrandt KV, Bowden J, risk of overweight or obesity? A systematic review of observation-
Ray DW, Freathy RM, Murray A, Mazzotti DR, Gehrman PR, al studies with meta-analysis. Int J Epidemiol 47:1956–1971.
Lawlor DA, Frayling TM, Rutter MK, Hinds DA, Saxena R, https://doi.org/10.1093/ije/dyy079
Weedon MN (2019) Genome-wide association analyses of 84. Manoogian ENC, Panda S (2017) Circadian rhythms, time-
chronotype in 697,828 individuals provides insights into circadian restricted feeding, and healthy aging. Ageing Res Rev 39:59–67.
rhythms. Nat Commun 10:343. https://doi.org/10.1038/s41467- https://doi.org/10.1016/j.arr.2016.12.006
018-08259-7 85. Marcheva B, Ramsey KM, Buhr ED, Kobayashi Y, Su H, Ko CH,
69. Kalsbeek A, Fliers E, Romijn JA, la Fleur SE, Wortel J, Bakker O, Ivanova G, Omura C, Mo S, Vitaterna MH, Lopez JP, Philipson
Endert E, Buijs RM (2001) The suprachiasmatic nucleus generates LH, Bradfield CA, Crosby SD, JeBailey L, Wang X, Takahashi
the diurnal changes in plasma leptin levels. Endocrinology 142: JS, Bass J (2010) Disruption of the clock components CLOCK
2677–2685. https://doi.org/10.1210/endo.142.6.8197 and BMAL1 leads to hypoinsulinaemia and diabetes. Nature 466:
70. Karatsoreos IN, Bhagat S, Bloss EB, Morrison JH, McEwen BS 627–631. https://doi.org/10.1038/nature09253
(2011) Disruption of circadian clocks has ramifications for metab- 86. Marcheva B, Ramsey KM, Peek CB, Affinati A, Maury E, Bass J
olism, brain, and behavior. Proc Natl Acad Sc U S A 108:1657– (2013) Circadian clocks and metabolism. Handb Exp Pharmacol
1662. https://doi.org/10.1073/pnas.1018375108 217:127–155. https://doi.org/10.1007/978-3-642-25950-0_6
71. Karlsson B, Knutsson A, Lindahl B (2001) Is there an association 87. Martinez-Guryn K, Hubert N, Frazier K, Urlass S, Musch MW,
between shift work and having a metabolic syndrome? Results Ojeda P, Pierre JF, Miyoshi J, Sontag TJ, Cham CM, Reardon CA,
from a population based study of 27 485 people. Occup Environ Leone V, Chang EB (2018) Small intestine microbiota regulate
Med 58:747–752. https://doi.org/10.1136/oem.58.11.747 host digestive and absorptive adaptive responses to dietary lipids.
72. Kim M-J, Son K-H, Park H-Y, Choi D-J, Yoon C-H, Lee H-Y, Cho Cell Host Microbe 23:458–469.e455. https://doi.org/10.1016/j.
E-Y, Cho M-C (2013) Association between shift work and obesity chom.2018.03.011
among female nurses: Korean nurses’ survey. BMC Public Health 88. Masri S (2015) Sirtuin-dependent clock control: new advances in
13:1204. https://doi.org/10.1186/1471-2458-13-1204 metabolism, aging and cancer. Curr Opin Clin Nutr Metab Care
73. Kojetin DJ, Burris TP (2014) REV-ERB and ROR nuclear recep- 18:521–527. https://doi.org/10.1097/MCO.0000000000000219
tors as drug targets. Nat Rev Drug Discov 13:197–216. https://doi. 89. Masuki S, Todo T, Nakano Y, Okamura H, Nose H (2005)
org/10.1038/nrd4100 Reduced alpha-adrenoceptor responsiveness and enhanced baro-
74. Lamia KA, Evans RM (2010) Metabolism Tick, tock, a β-cell reflex sensitivity in cry-deficient mice lacking a biological clock. J
clock. Nature 466(7306):571–572. https://doi.org/10.1038/ Physiol 566:213–224. https://doi.org/10.1113/jphysiol.2005.
466571a 086728
Pflugers Arch - Eur J Physiol

90. Maury E (2019) Off the clock: from circadian disruption to met- 106. Preitner N, Damiola F, Luis Lopez M, Zakany J, Duboule D,
abolic disease. Int J Mol Sci 20:1597. https://doi.org/10.3390/ Albrecht U, Schibler U (2002) The orphan nuclear receptor
ijms20071597 REV-ERBα controls circadian transcription within the positive
91. Maury E, Ramsey KM, Bass J (2010) Circadian rhythms and limb of the mammalian circadian oscillator. Cell 110:251–260.
metabolic syndrome: from experimental genetics to human dis- https://doi.org/10.1016/S0092-8674(02)00825-5
ease. Circ Res 106(3):447–462. https://doi.org/10.1161/ 107. Rana S, Mahmood S (2010) Circadian rhythm and its role in
CIRCRESAHA.109.208355 malignancy. J Circadian Rhythms 8:3–3. https://doi.org/10.1186/
92. McHill AW, Wright KP Jr (2017) Role of sleep and circadian 1740-3391-8-3
disruption on energy expenditure and in metabolic predisposition 108. Reppert SM, Weaver DR (2002) Coordination of circadian timing
to human obesity and metabolic disease. Obes Rev 18:15–24. in mammals. Nature 418:935–941. https://doi.org/10.1038/
https://doi.org/10.1111/obr.12503 nature00965
93. McHill AW, Melanson EL, Higgins J, Connick E, Moehlman TM, 109. Rothschild J, Hoddy KK, Jambazian P, Varady KA (2014) Time-
Stothard ER, Wright KP (2014) Impact of circadian misalignment restricted feeding and risk of metabolic disease: a review of human
on energy metabolism during simulated nightshift work. Proc Natl and animal studies. Nutr Rev 72:308–318. https://doi.org/10.1111/
Acad Sci U S A 111:17302–17307. https://doi.org/10.1073/pnas. nure.12104
1412021111 110. Ruiter M, La Fleur SE, van Heijningen C, van der Vliet J,
94. Milano W, De Rosa M, Milano L, Capasso A (2012) Night eating Kalsbeek A, Buijs RM (2003) The daily rhythm in plasma gluca-
syndrome: an overview. J Pharm Pharmacol 64:2–10. https://doi. gon concentrations in the rat is modulated by the biological clock
org/10.1111/j.2042-7158.2011.01353.x and by feeding behavior. Diabetes 52:1709–1715. https://doi.org/
95. Morris CJ, Purvis TE, Mistretta J, Scheer FAJL (2016) Effects of 10.2337/diabetes.52.7.1709
the internal circadian system and circadian misalignment on glu- 111. Sadacca LA, Lamia KA, deLemos AS, Blum B, Weitz CJ (2011)
cose tolerance in chronic shift workers. J Clin Endocrinol Metab An intrinsic circadian clock 54:120-124. Of the pancreas is re-
101:1066–1074. https://doi.org/10.1210/jc.2015-3924 quired for normal insulin release and glucose homeostasis in mice.
96. Mure LS, Le HD, Benegiamo G, Chang MW, Rios L, Jillani N, Diabetologica. https://doi.org/10.1007/s00125-010-1920-8
Ngotho M, Kariuki T, Dkhissi-Benyahya O, Cooper HM, Panda S
112. Sakamoto K, Nagase T, Fukui H, Horikawa K, Okada T, Tanaka
(2018) Diurnal transcriptome atlas of a primate across major neu-
H, Sato K, Miyake Y, Ohara O, Kako K, Ishida N (1998)
ral and peripheral tissues. Science 359:eaao0318. https://doi.org/
Multitissue circadian expression of rat periodHomolog (rPer2)
10.1126/science.aao0318
mRNA is governed by the mammalian circadian clock, the supra-
97. Oishi K, Miyazaki K, Kadota K, Kikuno R, Nagase T, G-i A,
chiasmatic nucleus in the brain. J Biol Chem 273:27039–27042.
Ohkura N, Azama T, Mesaki M, Yukimasa S, Kobayashi H,
https://doi.org/10.1074/jbc.273.42.27039
Iitaka C, Umehara T, Horikoshi M, Kudo T, Shimizu Y, Yano
113. Salgado-Delgado RC, Saderi N, Basualdo MC, Guerrero-Vargas
M, Monden M, Machida K, Matsuda J, Horie S, Todo T, Ishida
NN, Escobar C, Buijs RM (2013) Shift work or food intake during
N (2003) Genome-wide expression analysis of mouse liver reveals
the rest phase promotes metabolic disruption and desynchrony of
CLOCK-regulated circadian output genes. J Biol Chem 278:
liver genes in male rats. PLoS One 8:e60052. https://doi.org/10.
41519–41527. https://doi.org/10.1074/jbc.M304564200
1371/journal.pone.0060052
98. Oishi K, Shirai H, Ishida N (2005) CLOCK is involved in the
114. Scheer FAJL, Hilton MF, Mantzoros CS, Shea SA (2009) Adverse
circadian transactivation of peroxisome-proliferator-activated re-
metabolic and cardiovascular consequences of circadian misalign-
ceptor alpha (PPARalpha) in mice. Biochem J 386:575–581.
ment. Proc Natl Acad Sci U S A 106:4453–4458. https://doi.org/
https://doi.org/10.1042/BJ20041150
10.1073/pnas.0808180106
99. Okano S, Akashi M, Hayasaka K, Nakajima O (2009) Unusual
circadian locomotor activity and pathophysiology in mutant 115. Scott EM, Carter AM, Grant PJ (2007) Association between poly-
CRY1 transgenic mice. Neurosci Lett 451:246–251. https://doi. morphisms in the clock gene, obesity and the metabolic syndrome
org/10.1016/j.neulet.2009.01.014 in man. Int J Obes 32:658–662. https://doi.org/10.1038/sj.ijo.
100. Opperhuizen A-L, van Kerkhof LWM, Proper KI, Rodenburg W, 0803778
Kalsbeek A (2015) Rodent models to study the metabolic effects 116. Serin Y, Acar Tek N (2019) Effect of circadian rhythm on meta-
of shiftwork in humans. Front Pharmacol 6:50–50. https://doi.org/ bolic processes and the regulation of energy balance. Ann Nutr
10.3389/fphar.2015.00050 Metab 74:322–330. https://doi.org/10.1159/000500071
101. Parkar SG, Kalsbeek A, Cheeseman JF (2019) Potential role for 117. Serón-Ferré M, Forcelledo ML, Torres-Farfan C, Valenzuela FJ,
the gut microbiota in modulating host circadian rhythms and met- Rojas A, Vergara M, Rojas-Garcia PP, Recabarren MP, Valenzuela
abolic health. Microorganisms 7:41. https://doi.org/10.3390/ GJ (2013) Impact of chronodisruption during primate pregnancy
microorganisms7020041 on the maternal and newborn temperature rhythms. PLoS One 8:
102. Parkes KR (2002) Shift work and age as interactive predictors of e57710. https://doi.org/10.1371/journal.pone.0057710
body mass index among offshore workers. Scand J Work Environ 118. Shimba S, Ogawa T, Hitosugi S, Ichihashi Y, Nakadaira Y,
Health 28:64–71. https://doi.org/10.5271/sjweh.648 Kobayashi M, Tezuka M, Kosuge Y, Ishige K, Ito Y, Komiyama
103. Paschos GK, FitzGerald GA (2017) Circadian clocks and metab- K, Okamatsu-Ogura Y, Kimura K, Saito M (2011) Deficient of a
olism: implications for microbiome and aging. Trends Genet 33: clock gene, brain and muscle Arnt-like protein-1 (BMAL1), in-
760–769. https://doi.org/10.1016/j.tig.2017.07.010 duces dyslipidemia and ectopic fat formation. PLoS One 6:
104. Paschos GK, Ibrahim S, Song W-L, Kunieda T, Grant G, Reyes e25231. https://doi.org/10.1371/journal.pone.0025231
TM, Bradfield CA, Vaughan CH, Eiden M, Masoodi M, Griffin 119. Skene DJ, Skornyakov E, Chowdhury NR, Gajula RP, Middleton
JL, Wang F, Lawson JA, FitzGerald GA (2012) Obesity in mice B, Satterfield BC, Porter KI, Van Dongen HPA, Gaddameedhi S
with adipocyte-specific deletion of clock component Arntl. Nat (2018) Separation of circadian- and behavior-driven metabolite
Med 18:1768–1777. https://doi.org/10.1038/nm.2979 rhythms in humans provides a window on peripheral oscillators
105. Potter GDM, Skene DJ, Arendt J, Cade JE, Grant PJ, Hardie LJ and metabolism. Proc Natl Acad Sci U S A 115:7825–7830.
(2016) Circadian rhythm and sleep disruption: causes, metabolic https://doi.org/10.1073/pnas.1801183115
consequences, and countermeasures. Endocr Rev 37:584–608. 120. Shi S-q, Ansari TS, McGuinness Owen P, Wasserman David H,
https://doi.org/10.1210/er.2016-1083 Johnson Carl H (2013) Circadian disruption leads to insulin
Pflugers Arch - Eur J Physiol

resistance and obesity. Curr Biol 23:372–381. https://doi.org/10. of obesity and circadian-rhythm disruption. Nat Commun 10.
1016/j.cub.2013.01.048 https://doi.org/10.1038/s41467-019-10563-9
121. Stenvers DJ, Scheer FAJL, Schrauwen P, la Fleur SE, Kalsbeek A 135. Voigt RM, Forsyth CB, Green SJ, Engen PA, Keshavarzian A
(2019) Circadian clocks and insulin resistance. Nat Rev (2016) Circadian rhythm and the gut microbiome. Int Rev
Endocrinol 15:75–89. https://doi.org/10.1038/s41574-018-0122-1 Neurobiol 131:193–205. https://doi.org/10.1016/bs.irn.2016.07.
122. Sun L, Wang Y, Song Y, Cheng X-R, Xia S, Rahman MRT, Shi Y, 002
Le G (2015) Resveratrol restores the circadian rhythmic disorder 136. Voigt RM, Summa KC, Forsyth CB, Green SJ, Engen P, Naqib A,
of lipid metabolism induced by high-fat diet in mice. Biochem Vitaterna MH, Turek FW, Keshavarzian A (2016) The circadian
Biophys Res Commun 458:86–91. https://doi.org/10.1016/j.bbrc. clock mutation promotes intestinal Dysbiosis. Alcohol Clin Exp
2015.01.072 Res 40:335–347. https://doi.org/10.1111/acer.12943
123. Sun M, Feng W, Wang F, Li P, Li Z, Li M, Tse G, Vlaanderen J, 137. Westermeier F, Sáez PJ, Villalobos-Labra R, Sobrevia L, Farías-
Vermeulen R, Tse LA (2018) Meta-analysis on shift work and Jofré M (2014) Programming of fetal insulin resistance in preg-
risks of specific obesity types. Obes Rev 19:28–40. https://doi. nancies with maternal obesity by ER stress and inflammation.
org/10.1111/obr.12621 Biomed Res Int 2014:13–13. https://doi.org/10.1155/2014/
124. Sutton EF, Beyl R, Early KS, Cefalu WT, Ravussin E, Peterson 917672
CM (2018) Early time-restricted feeding improves insulin sensi- 138. Wyse CA, Celis Morales CA, Graham N, Fan Y, Ward J, Curtis
tivity, blood pressure, and oxidative stress even without weight AM, Mackay D, Smith DJ, Bailey MES, Biello S, Gill JMR, Pell
loss in men with prediabetes. Cell Metab 27:1212–1221.e1213. JP (2017) Adverse metabolic and mental health outcomes associ-
https://doi.org/10.1016/j.cmet.2018.04.010 ated with shiftwork in a population-based study of 277,168
125. Sweeney E, Yu ZM, Dummer TJB, Cui Y, DeClercq V, Forbes C, workers in UK biobank. Ann Med 49:411–420. https://doi.org/
Grandy SA, Keats M, Parker L, Adisesh A (2019) The relationship 10.1080/07853890.2017.1292045
between anthropometric measures and cardiometabolic health in
139. Xie Z, Su W, Liu S, Zhao G, Esser K, Schroder EA, Lefta M,
shift work: findings from the Atlantic PATH cohort study. Int Arch
Stauss HM, Guo Z, Gong MC (2015) Smooth-muscle BMAL1
Occup Environ Health 93(1):67–76. https://doi.org/10.1007/
participates in blood pressure circadian rhythm regulation. J Clin
s00420-019-01459-8
Investig 125:324–336. https://doi.org/10.1172/JCI76881
126. Takahashi JS, Hong H-K, Ko CH, McDearmon EL (2008) The
genetics of mammalian circadian order and disorder: implications 140. Yang X, Downes M, Yu RT, Bookout AL, He W, Straume M,
for physiology and disease. Nat Rev Genet 9:764–775. https://doi. Mangelsdorf DJ, Evans RM (2006) Nuclear receptor expression
org/10.1038/nrg2430 links the circadian clock to metabolism. Cell 126:801–810. https://
127. Tamai TK, Vardhanabhuti V, Foulkes NS, Whitmore D (2004) doi.org/10.1016/j.cell.2006.06.050
Early embryonic light detection improves survival. Curr Biol 14: 141. Yang S, Liu A, Weidenhammer A, Cooksey RC, McClain D, Kim
R104–R105 MK, Aguilera G, Abel ED, Chung JH (2009) The role of mPer2
128. Tamai TK, Carr AJ, Whitmore D (2005) Zebrafish circadian clock gene in glucocorticoid and feeding rhythms. Endocrinology
clocks: cells that see light. Biochem Soc Trans 33:962–966. 150:2153–2160. https://doi.org/10.1210/en.2008-0705
https://doi.org/10.1042/BST0330962 142. Yang G, Jia Z, Aoyagi T, McClain D, Mortensen RM, Yang T
129. Thaiss Christoph A, Zeevi D, Levy M, Zilberman-Schapira G, (2012) Systemic PPARγ deletion impairs circadian rhythms of
Suez J, Tengeler Anouk C, Abramson L, Katz Meirav N, Korem behavior and metabolism. PLoS One 7:e38117. https://doi.org/
T, Zmora N, Kuperman Y, Biton I, Gilad S, Harmelin A, Shapiro 10.1371/journal.pone.0038117
H, Halpern Z, Segal E, Elinav E (2014) Transkingdom control of 143. Yoshida J, Eguchi E, Nagaoka K, Ito T, Ogino K (2018)
microbiota diurnal oscillations promotes metabolic homeostasis. Association of night eating habits with metabolic syndrome and
Cell 159:514–529. https://doi.org/10.1016/j.cell.2014.09.048 its components: a longitudinal study. BMC Public Health 18:
130. Thosar SS, Butler MP, Shea SA (2018) Role of the circadian 1366. https://doi.org/10.1186/s12889-018-6262-3
system in cardiovascular disease. J Clin Investig 128:2157– 144. Yu JH, Yun C-H, Ahn JH, Suh S, Cho HJ, Lee SK, Yoo HJ, Seo
2167. https://doi.org/10.1172/JCI80590 JA, Kim SG, Choi KM, Baik SH, Choi DS, Shin C, Kim NH
131. Turek FW, Joshu C, Kohsaka A, Lin E, Ivanova G, McDearmon (2015) Evening chronotype is associated with metabolic disorders
E, Laposky A, Losee-Olson S, Easton A, Jensen DR, Eckel RH, and body composition in middle-aged adults. J Clin Endocrinol
Takahashi JS, Bass J (2005) Obesity and metabolic syndrome in Metab 100:1494–1502. https://doi.org/10.1210/jc.2014-3754
circadian clock mutant mice. Science 308:1043–1045. https://doi. 145. Zhang R, Lahens NF, Ballance HI, Hughes ME, Hogenesch JB
org/10.1126/science.1108750 (2014) A circadian gene expression atlas in mammals: implica-
132. Valenzuela FJ, Torres-Farfan C, Richter HG, Mendez N, Campino tions for biology and medicine. Proc Natl Acad Sci U S A 111:
C, Torrealba F, Valenzuela GJ, Serón-Ferré M (2008) Clock gene 16219–16224. https://doi.org/10.1073/pnas.1408886111
expression in adult primate suprachiasmatic nuclei and adrenal: is 146. Zhao Y, Zhang Y, Zhou M, Wang S, Hua Z, Zhang J (2012) Loss
the adrenal a peripheral clock responsive to melatonin? of mPer2 increases plasma insulin levels by enhanced glucose-
Endocrinology 149:1454–1461. https://doi.org/10.1210/en.2007- stimulated insulin secretion and impaired insulin clearance in
1518 mice. FEBS Lett 586:1306–1311. https://doi.org/10.1016/j.
133. Valenzuela FJ, Vera J, Venegas C, Muñoz S, Oyarce S, Muñoz K, febslet.2012.03.034
Lagunas C (2016) Evidences of polymorphism associated with 147. Zhou S, Tang X, Chen H-Z (2018) Sirtuins and insulin resistance.
circadian system and risk of pathologies: a review of the literature. Front Endocrinol 9:748. https://doi.org/10.3389/fendo.2018.
Int J Endocrinol 2016:12–12. https://doi.org/10.1155/2016/ 00748
2746909
134. Villanueva JE, Livelo C, Trujillo AS, Chandran S, Woodworth B,
Andrade L, Le HD, Manor U, Panda S, Melkani GC (2019) Time- Publisher’s note Springer Nature remains neutral with regard to jurisdic-
restricted feeding restores muscle function in Drosophila models tional claims in published maps and institutional affiliations.

You might also like