You are on page 1of 10

Physiology & Behavior 220 (2020) 112873

Contents lists available at ScienceDirect

Physiology & Behavior


journal homepage: www.elsevier.com/locate/physbeh

Review

Circadian regulation of appetite and time restricted feeding T


a,b,⁎ a c,d a,b
Amanda J. Page , Stewart Christie , Erin Symonds , Hui Li
a
Vagal Afferent Research Group and Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide Medical School, University of Adelaide,
Adelaide, SA 5000, Australia
b
Nutrition, Diabetes & Gut Health, Lifelong Health Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
c
Bowel Health Service, Flinders Medical Centre, Bedford Park, SA 5042, Australia
d
Flinders Centre for Innovation in Cancer, Flinders University, Bedford Park, SA 5042, Australia

A R T I C LE I N FO A B S T R A C T

Keywords: The circadian system plays an important role in the temporal regulation of metabolic processes as well as food
Circadian intake to ensure energy efficiency. The ‘master’ clock is located within the superchiasmatic nucleus and receives
Food intake input from the retina so that it can be entrained by the light:dark cycle. In turn, the master clock entrains other
Time-restricted feeding clocks in the central nervous system, including areas involved in energy homeostasis such as the arcuate nucleus,
Food-entrainable oscillator
and the periphery (e.g. adipose tissue and the gastrointestinal tract). This master clock is reinforced by other
zeitgebers such as the timing of food intake and activity. If these zeitgebers desynchronise, such as occurs in high
fat diet-induced obesity or shift work conditions, it can lead to a misalignment of circadian clocks, disruption of
metabolic processes and the development of metabolic disorders. The timing of food intake is a strong zeitgeber,
particularly in the gastrointestinal tract, and therefore time restricted feeding offers potential for the treatment
of diet and shift work induced metabolic disorders. This review will focus on the role of the circadian system in
food intake regulation and the effect of environment factors, such as high fat diet feeding or shift work, on the
temporal regulation of food intake along with the benefits of time restricted feeding.

1. Introduction light:dark cycle. The role of circadian rhythms in the synchronisation of


processes such as food intake and metabolism ensure efficiency of en-
In animals, the acquisition of energy to maintain life requires the ergy homeostasis. However, disruption of these rhythms can lead to
intake of food. As a consequence, evolution has developed a sophisti- metabolic disorders, such as obesity. Conversely, obesity may disrupt
cated and well-integrated system to finely control energy intake. circadian rhythms perpetuating the problem.
Centrally, the hypothalamus plays a key role in food intake regulation. The present review is focused on appetite regulation, the role of the
This area receives peripheral input from the gastrointestinal tract, for circadian system in controlling these signals and the potential of time
the short-term regulation of meal size and meal frequency, and adipose restricted feeding to reverse the adverse effects of shift work or obesity
tissue, for the long-term regulation of energy homeostasis. The pro- on disruption of these circadian satiety signals.
cesses that regulate energy homeostasis rely on a balance between or-
exigenic and anorexigenic factors. Circadian signals have an important 2. Circadian clock system
role in energy homeostasis [1], temporally regulating these opposing
factors and maximising energy efficiency. For example, circadian The circadian clock is complex but basically consists of a central
rhythms facilitate the temporal occurrence of related physiological clock, the ‘master’ clock, located in the suprachiasmatic nucleus (SCN)
functions, such as food intake and glycogenesis. of the anterior hypothalamus and peripheral clocks [2]. The master
‘Circadian’ is from the Latin words Circa and diem which translates clock coordinates the majority of daily rhythms. The SCN is composed
to ‘about a day’ and circadian rhythms occur over about a 24 h period. of multiple, single cell circadian oscillators, which generate coordinated
Many components of physiology, metabolism and behavior display circadian signals, if synchronised, to regulate physiological rhythms.
circadian rhythms. These rhythms are generated by endogenous clocks The SCN receives input from the retina via the retinohypothalamic tract
that possess highly conserved timing mechanisms enabling organisms which enables entrainment of the SCN oscillators to the 24 h light-dark
to participate and adapt to daily environmental fluctuations, such as the cycle [3].


Corresponding author at: Vagal Afferent Research Group, Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia.
E-mail address: amanda.page@adelaide.edu.au (A.J. Page).

https://doi.org/10.1016/j.physbeh.2020.112873
Received 9 December 2019; Received in revised form 1 February 2020; Accepted 10 March 2020
Available online 16 March 2020
0031-9384/ © 2020 Elsevier Inc. All rights reserved.
A.J. Page, et al. Physiology & Behavior 220 (2020) 112873

The molecular mechanisms, which drive these circadian oscilla- Food Intake
tions, consists of a series of interlocking molecular loops, involving
rhythmic transcription of specific clock genes and interactions of the
proteins they encode. In very basic terms, clock genes consist of positive
elements, such as Brain and Muscle ARNT-Like 1 (BMAL1), Circadian PVN
Locomotor Output Cycles Kaput (Clock) and neuronal PAS domain
protein 2 (NPAS2). BMAL1 forms a heterodimer with either Clock or
NPAS2, enters the nucleus and stimulates transcription of negative
elements, such as period 1, 2 and 3 (Per 1–3) and cryptochrome 1 and 2
DMN LHA
(Cry 1–2) [4-6]. The protein products of these genes heterodimerise,
translocate to the nucleus and inhibit the activity of the BMAL1/Clock BBB
complex. Further, a number of nuclear receptors, such as REV-ERB and
ROR interact with or can be considered core components of the circa-
dian clock [7]. For example, BMAL1/Clock activates REV-ERBα tran- Anorexigens POMC Dorsal
scription resulting in daily fluctuations in REV-ERBα, which, in turn, CART VMN
represses the transcription of BMAL1 [8,9]. In contrast, RORα stimu- NPY
lates BMAL1 expression [10]. These nuclear receptors act as a physio- AgRP
Orexigens ARC
logical signaling relay enabling the clock rhythms to be influenced by
nutrient signals (e.g. derivatives of fatty acids), hormonal signals (e.g. Ventral
cortisol) and cellular redox status and vice versa [11-13]. Similar clock Medial Lateral
oscillators have also been found in other central areas and in peripheral
Oscillatory signalling
tissues, such as the gastrointestinal tract, liver, muscle and adipose
tissue. The SCN sends signals, via neural connections or circulating Fig. 1. Hypothalamic nuclei involved in appetite regulation. The major hy-
humoral factors, to the peripheral clocks to prevent dampening of the pothalamic nuclei involved in appetite regulation are the arcuate nucleus
circadian rhythms in the peripheral tissues [14]. In addition to light, (ARC), dorsal medial nucleus (DMN), ventral medial nucleus (VMN), para-
feeding episodes, signalled by the gastrointestinal tract, and availability ventricular nucleus (PVN), and lateral hypothalamic area (LHA). Green lines
represent activation, red lines represent inhibition. The ARC contains two op-
of metabolites, represents another important synchroniser for central
posing nuclei expressing either: neuropeptide Y (NPY) and agouti-related
and peripheral clocks [15]. Restriction of food availability to a narrow
peptide (AgRP) or pro-opiomelanocortin (POMC) and cocaine and ampheta-
window each day (time-restricted feeding) has been demonstrated to mine related transcript (CART). NPY/AgRP neurons from the ARC inhibit the
result in a profound reorganisation of behavior and physiology [16,17]. PVN, DMN, and VMN, and stimulates the LHA to increase food intake. POMC/
Such restriction of food availability induces a bout of activity, known as AgRP neurons from the ARC inhibit the LHA and stimulate the PVN, DMN, and
food anticipatory activity, in advance of food availability. These food VMN to inhibit food intake. The ARC is adjacent to a weak blood brain barrier
anticipatory activities display properties consistent with control by (BBB) and is therefore capable of being regulated by blood borne signals such as
endogenous circadian clocks rather than a food-driven phenomenon. orexigens (e.g. ghrelin) and anorexigens (e.g. leptin). The daily feed-fast cycle is
For example, if the period of food availability is abruptly delayed the controlled by oscillations in all areas of the hypothalamic nuclei, involved in
onset of food anticipatory activity takes multiple days to resynchronise. food intake, which receives input for the master clock in the suprachiasmatic
nuclei of the hypothalamus.
Further, if the animal is completely food deprived the onset of the food
anticipatory activity persists for as long as the fasting period can be
maintained [16,17]. MSH binding and the subsequent inhibition of food intake.
POMC/CART and NPY/AgRP neurones in the ARC project to other
3. Food intake regulation hypothalamic nuclei including the PVN, LHA, VMN and DMV [23]. The
PVN is linked with satiety [32] and is stimulated by POMC/CART
3.1. Central regulation of food intake neurones and inhibited by NPY/AgRP neurones [19]. Conversely, the
LHA is considered orexigenic and contains two populations of neurones
There are many brain regions, including the hypothalamus, hind- expressing either orexin or melanin concentrating hormone (MCH)
brain and corticolimbic system, that play a role in the control of food [33,34]. These subsets of neurones are stimulated by NPY/AgRp neu-
intake [18]. In particular, the hypothalamus is composed of many nu- rones and inhibited by POMC/CART neurones [19]. The VMN and DMN
clei with various functions associated with energy homeostasis [19]. In also receive projections from the ARC where they are stimulated by
relation to food intake the most relevant hypothalamic areas include POMC/CART neurones and inhibited by NPY/AgRP neurones [19].
the arcuate nucleus (ARC), paraventricular nucleus (PVN), lateral hy- It has been demonstrated that the circadian system orchestrates the
pothalamic area (LHA), ventral medial nucleus (VMN) and dorsal temporal regulation of metabolic processes over a 24 h cycle, giving rise
medial nucleus (DMN; Fig. 1). The ARC is located in the mediobasal to diurnal rhythms in energy expenditure [35,36], appetite [37-40] and
hypothalamus and contains two distinct opposing populations of nuclei: insulin sensitivity [41-43]. Anatomical studies have identified connec-
those that express pro-opiomelanocortin (POMC) [20] and cocaine and tions between the SCN and several hypothalamic and extra-hypotha-
amphetamine regulated transcript (CART) [21], and those that express lamic areas, including the subparaventricular region, PVN, ARC, lateral
neuropeptide Y (NPY) and agouti-related peptide (AgRP) [22]. The geniculate nucleus and raphe nuclei [44-47]. These connections are
POMC/CART neurones are considered anorexigenic with their activa- important for the coordination of daily physiological rhythms, such as
tion reducing food intake. These neurones can be stimulated by leptin food intake, energy expenditure, locomotor activity and temperature.
and insulin and inhibited by ghrelin [23]. POMC is a precursor for The rhythmic activity of neuroendocrine neurones within the hy-
many peptides, including α-melanocyte stimulating hormone (α-MSH) pothalamus, including gene and protein expression of many of the
[24]. α-MSH binds to the melanocortin receptor 3 (MCR3) or the MCR4 neurotransmitters within the hypothalamic region [48-50], is regulated
to inhibit food intake [25,26]. Conversely, NPY/AgRP neurones are through these connections [51] and may be required for the circadian
orexigenic with their activation increasing food intake [27,28]. These regulation of appetite. For example, it has been shown that the in-
neurones can be stimulated by ghrelin and inhibited by leptin and creased food intake at the onset of the dark phase is reduced in mice
peptide tyrosine-tyrosine (PYY) [29]. NPY binds to Y1 receptors [30] lacking NPY [52], suggesting circadian rhythmicity of NPY in the hy-
and AgRP acts as an antagonist at MCR3 and MCR4 [31] to prevent α- pothalamus is driving diurnal rhythms in food intake. In addition,

2
A.J. Page, et al. Physiology & Behavior 220 (2020) 112873

expression of the clock gene REV-ERBα in the brain has been shown to regulation of food intake. This axis is comprised of neural vagal afferent
control the circadian organisation of food intake [53]. Circadian and endocrine signals that coordinate to regulate food intake. There is
rhythms may also explain the persistent basic eating patterns in hu- evidence that all aspect of this axis displays diurnal rhythms to regulate
mans, which has been observed in individuals isolated from external the timing of food intake, maximising digestion and nutrient absorption
time cues, such as the light-dark cycle [54]. as well as aligning food intake with energy requirements.
Central reward mechanisms also play an important role in influen-
cing food intake behavior. The reward system exerts distinct actions 3.3.1. Vagal afferents
through brain systems and neural pathways, such as the corticolimbic Gastrointestinal vagal afferents play an important role in the control
(e.g. amygdala, prefrontal cortex and hippocampus) and mesolimbic of food intake. Vagal afferents respond to mechanical and chemical
(e.g. nucleus accumbens, striatum and ventral tegmental area (VTA)) stimuli relaying information to the nucleus tractus solitarius (NTS). It is
systems [19] which represent the ‘liking’ and ‘wanting’ areas respec- generally believed that this information is then passed to the hy-
tively [55,56]. Further, the mesolimbic dopamine system is thought to pothalamus where it is integrated with other signals eventually leading
be the major reward seeking pathway [57]. Rhythmic dopamine release to feelings of fullness and satiety with subsequent changes in feeding
has been reported in the dorsal striatum [58] and this rhythmicity behavior. However, decerebrate rats, that lack the hindbrain to fore-
drives rhythmicity of Per2 in the rat dorsal striatum but not the SCN brain connections, are also capable of terminating a meal suggesting
[59]. that some aspects of food intake control are represented in the caudal
The metabolic status of an individual is also relayed, in a circadian- brain areas [78]. Gastric vagal afferents are predominantly mechan-
dependent manner, via humoral and neural signals from the peripheral osensitive [79,80], responding to the arrival and amount of food in the
tissues to the brain regions that control appetite [60]. In the following stomach. Gut hormones such as leptin [81] and ghrelin [82,83] are able
sections the input from adipose tissue and the gastrointestinal tract, (i.e. to modulate the sensitivity of these mechanosensitive afferents. In the
involved in the long and short-term regulation of energy homeostasis small intestine, there is an enteroendocrine response to nutrients, where
respectively) will be discussed. specific nutrients interact with a nutrient receptor initiating an in-
tracellular process that culminates in the release of a gut hormone
3.2. Adipose tissue [84,85], such as cholecystokinin (CCK). These gut hormones can then
either act directly on vagal afferent endings or enter the circulation to
White adipose tissue generally serves as the body's energy store act in an endocrine manner on central sites of appetite regulation.
where it can be utilised when energy intake is low. However, white Further, it has recently been demonstrated that duodenal mechan-
adipose tissue also secretes many peptides or adipokines that can reg- osensitive afferents play a major role in food intake regulation [86].
ulate energy balance and metabolism, such as leptin. After secretion There is limited information available on the role of the circadian
from adipose tissue, leptin enters the circulation and, via a saturable system in the regulation of vagal afferent function. However, it has been
transport protein [61], can cross the hypothalamic blood brain barrier. demonstrated that gastric vagal afferents display diurnal rhythms in
Here it binds to its receptor in the ARC causing inhibition of NPY and response to mechanical stimuli [87]. These rhythms are lost in high fat
activation of POMC neurones with a resultant reduction in food intake diet-induced obese mice [88] and/or simulated shift work conditions
[62] and increase in energy expenditure [63, 64]. It should be noted, [89], and associated with a ‘grazing-like’ behavior in food intake
that leptin can also act on other areas throughout the brain to control (Fig. 2).
feeding and feeding behavior, including the hindbrain, hippocampus
and limbic brain regions [65]. Leptin is secreted in proportion to adi- 3.3.2. Nutrient sensors
pocyte size and adipose mass [66]. However, it is also secreted in a Although there is evidence that intestinal nutrient transporters, such
circadian manner [67,68]; under the control of the SCN via sympathetic as sodium-glucose transporter 1 (SGLT1), glucose transporters (GLUT;
input to the adipocytes [69]. In humans, plasma leptin levels are high
during the night favouring fasting and nocturnal rest. In contrast, GVA signaling
during the daytime leptin levels are comparatively low permitting the
intake of food during the active period when energy demands are high.
In obese individuals, leptin levels are elevated, however, circulating ‘Normal’ Diurnal rhythms
diurnal level of leptin are still maintained but with lower amplitude condions in GVA signaling
[70,71]. With central leptin resistance the significance of circulating
diurnal leptin levels in appetite regulation is questionable [72]. This
indirect SCN signaling, via adipose tissue, will reinforce the temporal
regulation of food intake and metabolism centrally.
Loss of rhythms
High fat diet
3.3. Gastrointestinal tract
in GVA signaling
Grazing
The gastrointestinal tract plays a vital role in food intake, digestion
and absorption of nutrients. There is substantial evidence that the
gastrointestinal tract contains functional clock genes [73-75] and it also Loss of rhythms
receives input from the SCN via humoral and neural pathways. Clock Shi work in GVA signaling
genes are present throughout the gut, including the myenteric plexus
and the epithelial cells, suggesting a role in the generation of daily Grazing
rhythms in gastrointestinal functions, such as gastrointestinal motility Fig. 2. Schematic of the effect of light:dark cycle and diet on diurnal rhythms in
(e.g. gastric emptying), gastric secretions, enzymatic activity, and nu- gastric vagal afferent responses to food related stimuli in mice. Under ‘normal’
trient detection and absorption in the small intestine [76,77]. This conditions the light cycle and food intake patterns are coordinated leading to
circadian rhythmicity provides the gastrointestinal tract the ability to diurnal rhythms in gastric vagal afferent signaling. In high fat diet conditions
anticipate a meal and optimally prepare for the transit and absorption there is a disrupted ‘grazing-like’ feeding behavior resulting in a loss of diurnal
of nutrients. This review will focus on the gut-brain axis and appetite rhythms in gastric vagal afferent signaling. Similarly, in simulated shift work
regulation. conditions diurnal rhythms in food intake are disrupted leading to a loss of
The gut-brain axis plays an important role in the short-term diurnal rhythms in gastric vagal afferent signaling.

3
A.J. Page, et al. Physiology & Behavior 220 (2020) 112873

A Relative T1R2 mRNA


0.0005 pattern of secretion independent of nutrient stimulation.
0.0004 Leptin is not only expressed in adipose tissue but is also expressed in
expression the stomach [97,98]. Gastric leptin levels oscillate in a circadian
0.0003
manner with leptin levels high at night and low during the day in hu-
0.0002 mans [99]. Leptin modulates gastric vagal afferent mucosal receptor
0.0001 ** Vs
** ** ** mechanosensitivity [81] and, therefore, it is possible that diurnal
0.0000 12Hrs rhythms in gastric leptin levels are driving diurnal rhythms in gastric
0 3 6 9 12 15 18 21
Zeitgeber (Hrs) vagal afferent mucosal receptor mechanosensitivity. Further, the mod-
B ulatory effect of leptin on gastric vagal afferent mucosal receptors
Relative GPR93 mRNA

0.04 displays diurnal rhythms with potentiation of gastric vagal afferent


0.03 mucosal receptors during the light phase; an effect that diminishes
expression

during the dark phase in mice [88].


0.02 Ghrelin is also produced in the stomach [100,101] and is involved
0.01 in stimulating appetite via its central action on neuropeptide Y in the
lateral hypothalamus [102,103] and peripheral action on gastric vagal
0.00
0 3 6 9 12 15 18 21 afferents [82,83]. Ghrelin can also alter clock function in the SCN
Zeitgeber (Hrs) [104,105] and, hence, may be a signal involved in the cross-talk be-
C tween the peripheral and central clock system. Ghrelin oscillates with
Relave CaSR mRNA

0.0003
feeding [106], making this peptide a candidate for food-related en-
expression

0.0002 training signals. In humans, circulating levels of ghrelin are high during
the dark phase and relatively lower during the light phase [72].
0.0001 A number of other gut peptides, including glucagon-like peptide-1
(GLP-1) and PYY [107] and CCK [108] are thought to display circadian
0.0000 rhythms in circulating plasma levels, however, it is difficult to tease out
0 3 6 9 12 15 18 21
the impact of food intake from the circadian cycle on these diurnal
Zeitgeber (Hrs)
rhythms.
D
Relative GPR40 mRNA

0.0020
3.3.4. Microbiota
0.0015
expression

There is mounting evidence to suggest that the gut microbiota in-


0.0010 fluences the host's homeostatic processes and modulates the develop-
0.0005 ment of obesity and type 2 diabetes. Microbial products, such as short
chain fatty acids (SCFAs) and succinate, can alter intestinal gluconeo-
0.0000
0 3 6 9 12 15 18 21 genesis [109,110]. In addition, the microbiota affects circulating sig-
Zeitgeber (Hrs) nals, enteric neurons and vagal afferents that carry hunger and satiety
cues to the brain [111]. The gut microbiota also contributes to the
Fig. 3. Expression of nutrient receptors in the mouse proximal jejunum over a
diurnal communication between the gut and the brain participating in
24 h period. Expression of the (A) sweet taste component T1R2; (B) protein
the circadian regulation of food intake. The gut microbiota has been
hydrosylate receptor (GPR93); (C) amino acid receptor (calcium sensing re-
ceptor (CaSR)) and (D) long chain fatty acid receptor (GPR40 (aka FFAR1))
shown to modulate various host gastrointestinal clocks [112,113]. In
mRNA in the mouse (N = 6) proximal jejunum relative to GAPDH. **P < 0.01 turn, the hosts circadian system, feeding schedules as well as diet
vs ZT12; one-way ANOVA. Zeitgeber is hours since lights on. composition have a major effect on the daily fluctuations of the mi-
crobiota in mice and humans [114-116]. Removing the gut microbiota
has been shown to impair clock gene expression in the intestinal epi-
GLUT2 and GLUT5) and the proton-coupled oligopeptide transporter 1
thelial cells, liver and mediobasal hypothalamus [112,114]. The ques-
display diurnal rhythmicity [90-94], with peak expression during the
tion is whether the microbiota is controlling the gut clock or vice versa.
dark phase in rodents, there is little known on the diurnal expression of
The circadian clock within the intestinal epithelial cells is profoundly
nutrient receptors. Unpublished data (Fig. 3) indicate that a component
disrupted by depletion of the microbiota [112] and therefore it would
of the sweet taste receptor T1R2 displays diurnal rhythms in mRNA
appear that the microbiota is determining the function of the gut clock.
expression (Fig. 3A) in the mouse proximal jejunum; an effect not ob-
However, the composition of the microbiota undergoes diurnal oscil-
served in the rat jejunum [95]. This discrepancy may be due to the lack
lations in mice and humans [117] and these oscillations are disrupted in
of sufficient time points, particular the time-point of peak expression, in
mutant mice with a genetic mutation in the clock system and jet-lag
the study by Bhutta et al. [95] or simply due to species differences. In
experiments [96,117,118]. Therefore, the communication appears to be
contrast, no rhythmicity in the expression of protein hydrosylate re-
bi-directional. However, it is difficult to untangle direct communication
ceptor (GPR93; Fig. 3B), amino acid receptor (calcium sensing receptor;
between the gut and microbiota and indirect effects due to changes in
Fig. 3C) and the long chain fatty acid receptor mRNA (GPR40 (aka
feeding behavior.
FFAR1); Fig. 3D) were observed in the mouse proximal jejunum. These
differences may be related to their role in specific regions of the gas-
4. Timing of food intake and time-restricted feeding
trointestinal tract and the local drivers of these rhythms. For example,
diurnal fluctuations in microbial short chain fatty acid production is
The light-dark cycle is not the only regulator of circadian rhythms.
thought to drive diurnal expression of free fatty acid receptor 2 (FFAR2)
Food is a very potent zeitgeber especially for clocks in the gastro-
and FFAR3 in the colon [96].
intestinal tract. If rodents have access to food only during the light
phase, when they are normally docile, they will adjust to this feeding
3.3.3. Gut hormones schedule within a few days. This includes the display of food antici-
Gut hormones are released from the gastrointestinal tract to co- patory activity, such as increased locomotor activity, temperature, di-
ordinate the timing of food intake and the absorption and utilisation of gestive enzyme activity and gastrointestinal motility, a few hours be-
nutrients in the body. The majority are released in response to nu- fore food becomes available [17,119,120]. In addition, the rhythms in
trients, however, some are known to follow a circadian anticipatory clock gene expression realign with the new feeding schedule [121-123].

4
A.J. Page, et al. Physiology & Behavior 220 (2020) 112873

For example, there are studies that demonstrate that the liver clock can over 15 h per day [141], reducing the fasting period and disrupting the
be entrained by time-restricted feeding independently of the SCN and circadian alignment of the peripheral and central clocks. Male and fe-
the light cycle [124,125]. Under ‘normal’ conditions, animals and hu- male mice exposed to a dim light at night gained more weight than mice
mans have a diurnal rhythm in food intake with the major proportion of exposed to a normal light:dark cycle [126,142]. Interestingly, this was
24 h food intake consumed during the natural wake period. These daily not due to an increase in total daily food intake but rather a change in
eating-fasting rhythms are aligned with other zeitgebers, such as ac- eating patterns, with an increase in food intake during the light phase
tivity and the light-dark cycle, where they can act synergistically to compared to mice exposed to a normal light cycle [126].
drive daily rhythms in appetite, anabolism and catabolism. Further, it The availability of calorie dense diets can also impact on the cir-
has been shown that abandonment of regular eating patterns, due to the cadian system. For example, rodents fed a high fat diet shift their pat-
increased demands imposed by contemporary 24 h societies, disrupts tern of food intake so that they display a ‘grazing-like’ behavior, with
nutritional and metabolic processes leading to profound effects on long- increased food intake during the light phase compared to animals fed a
term health and well-being [126–128]. It should be noted that mis- standard laboratory diet [88,143]. These changes in behavioral rhyth-
alignment can occur for a number of reasons, including shift work, micity correlate with disrupted clock gene expression in the hypotha-
exposure to prolonged hours of artificial light and erratic eating pat- lamus, peripheral tissue, such as the liver and adipose tissue, as well as
terns. It is difficult to separate the outcomes of these perturbations as altered cycling hormones, such as leptin, thyroid stimulating hormone
one factor often leads to disruption of another factor. For example, shift and testosterone in rodents and humans [143–148]. Diurnal rhythms in
workers generally eat at the wrong time of day due to their altered peripheral gastric vagal afferent satiety signaling is also lost in high fat
sleep-wake patterns. Nonetheless, it is clear that anything that causes diet-induced obesity [88], however, it is still unclear whether the dis-
misalignment of the peripheral and central clocks will lead to disrup- ruption in food intake is causing the disruption in the circadian system
tion in food intake patterns along with metabolic misalignment that or vice versa. Nonetheless, the use of time restricted feeding to reinforce
ultimately contributes to the development and maintenance of obesity circadian rhythms and provide temporal regulation of appetite, as well
and its co-morbidities, such as type 2 diabetes mellitus and cardiovas- as anabolism and catabolism, is an attractive option.
cular disease.
In today's industrial society a large proportion (~15–20%) of the 4.1. Time-restricted feeding
working population are shift workers [129–131] including permanent
night shifts, rotating shifts or irregular schedules. During the night shift, The concept of time-restricted feeding as a potential intervention for
workers are exposed to prolonged hours of artificial light and increased metabolic disease arose from studies that examined the effect of time
activity. There are also changes in feeding patterns with shift workers restricted food intake on behavior and clock components in metabolic
tending to schedule their meals around their working hours resulting in organs, such as the liver (see [149]). As stated above animals and hu-
increased food intake during the normal resting phase [132–135]. mans have diurnal rhythms in food intake with the majority of food
Further, on days off shift workers revert back to a more social daytime consumed in the active natural wake period. Due to the reciprocal in-
schedule; imposing a jet-lag paradigm as the central and peripheral teraction between metabolism and the circadian clock, these diurnal
clocks try to adjust to the new schedule. As a consequence, working rhythms in food intake act synergistically with the molecular circadian
shift work is a risk factor for health with shift workers more likely to clock to drive daily rhythms in appetite, anabolism and catabolism. A
become overweight than their day-shift counterparts [136]. It has been high fat diet disrupts feed-fasting rhythms in animals, with food intake
demonstrated that although the amount of food consumed does not spread over a 24 h period. These erratic eating patterns can contribute
vary between the night shift and the day shift or days off [137] the to or perpetuate the disruption in circadian rhythms in metabolic or-
pattern of food intake is dramatically changed with participants eating gans such as the liver, leading to the development and maintenance of
around the clock, while they are on night shift, with no extended fasting metabolic disease. A similar scenario is observed in simulated shift
periods [138]. A study has demonstrated a difference in the energy work conditions. For example, simulated shift work conditions in mice
expenditure response to the same meal depending on the time the meal fed a standard laboratory diet led to the development of fatty liver to
was consumed; with morning diet-induced thermogenesis significantly the same extent as mice fed a high fat diet [150]. Time-restricted
higher than afternoon and night diet-induced thermogenesis [139]. feeding protocols offer a mechanism to realign and reinforce circadian
Therefore, although the same calories were consumed, the time-de- rhythms and have been shown to offer significant protection from diet-
pendent differences in thermogenesis may significantly impact on induced obesity and associated metabolic diseases [151]. In addition,
weight gain in shift workers and highlights the importance of co- time-restricted feeding has been shown to prevent obesity induced by
ordinating food intake with metabolic processes. Under a normal 12 chronic advances of the light/dark cycle in mouse models of jet-lag and
h:12 h light:dark cycle ad libitum standard laboratory diet fed mice shift work [152]. Further, time restricted feeding has been shown to
display diurnal rhythms in food intake, with 60–80% of food consumed entrain circadian clock-related mechanisms, including behavior, se-
during the dark phase [87]. These food intake patterns were shown to cretion of hormones, body temperature, and clock gene expression in
be negatively associated with gastric vagal afferent responses to food the periphery [153]. The benefits of time-restricted feeding on meta-
related stimuli [87]. In contrast, mice exposed to simulated shift work bolic processes, heart and brain health has been covered in details in a
conditions (a normal light cycle (lights on at ZT0: Lights off at ZT12) for review by Chaix et al. [149]. This review will focus on the effects of
3 days followed by a reverse light cycle (lights on at ZT12: lights off at time-restricted feeding on food anticipatory activity and appetite reg-
ZT0) for 4 days, repeated for 8 weeks) displayed a ‘grazing-like’ be- ulation.
havior and a dampening in diurnal rhythms in food intake [89]. This
was associated with a loss of diurnal rhythms in gastric vagal afferent 4.1.1. Time-restricted feeding and the anticipation of food intake
satiety signaling [89]. It is unclear whether the ‘grazing-like’ behavior It has been shown that during time-restricted feeding there is an
is causing the loss in diurnal rhythms in gastric vagal afferent satiety increase in locomotor activity and body temperature 2-3 h before the
signals or the other way around and longitudinal studies are required to availability of food, suggesting that mice anticipate feeding time
address this question. through an internal time keeping mechanism known as the food-en-
Artificial light has extended the day period leading to an extension trainable oscillator (FEO) system [153]. The exact location of the FEO is
of the feeding period. The increase in availability of light at night unknown, but brain lesion and transgenic mouse studies have revealed
parallels the increased rates of obesity [126,140]. A study, using a some of the mechanisms involved in the FEO.
smart-phone app to monitor food intake and eating patterns, demon- Although the SCN and peripheral tissue clocks use the circadian
strated that over half of the participants had feeding episodes spanning timekeeping mechanism, described above, there is increasing evidence

5
A.J. Page, et al. Physiology & Behavior 220 (2020) 112873

that the FEO does not require these feedback loops. For example, a routine paradigm, there is a trough in hunger during the morning (~8
recent study has demonstrated that time restricted feeding can override am) which rises in the afternoon, with a peak in hunger in the evening
otherwise compromised rhythms in various clock gene mutant mice (~8 pm) [37]. In rodents, exposed to a light-dark cycle or constant
leading to improvements in metabolic health [154]. Studies have darkness, feeding rhythms align with the onset of activity in the evening
shown that Clock mutant mice display food anticipatory activity during [186]. However, in mice with a point mutation in the clock gene Per1
restricted feeding protocols [155,156]. In addition, mice deficient in there was a disconnect between the feeding pattern and activity, with
the clock gene Cryptochrome still displayed food anticipatory activity the commencement of feeding occurring a few hours before the normal
even when the SCN was disabled (SCN-lesion or housing in constant feeding time of wild-type mice [186]. This suggests that the drive to eat
darkness) [157]. Similar observations have been made with Per1-3 is independent of daily rhythms in activity.
single, double and triple knockout mice [158–162], although there are In terms of food intake regulation, it has been shown, at the mole-
some discrepancies in the literature regarding Per2 [163]. Despite the cular level, that time-restricted feeding reinforces the circadian rhythms
fact that the SCN is not critical for FEO timekeeping, several studies in AgRP/NPY neurones, known to promote food intake, in the mouse
have shown that circadian clock genes modulate food anticipatory ac- hypothalamus [187]. In addition, the loss of diurnal rhythms in gastric
tivity. There is evidence to indicate that the SCN inhibits anticipatory vagal afferent satiety signaling in high fat diet-induced obesity was
behavior with SCN-lesioned mice exhibiting stronger anticipatory ac- prevented by time restricted feeding [188]. It remains to be determined
tivity than control mice [164,165]. Further, in mice lacking functional whether a time restricted feeding protocol can reverse the loss of
NPAS2 there was delayed food anticipatory activity [166] and the diurnal rhythms in vagal afferent satiety signaling observed in high fat
period of the FEO rhythm was shorter in Per 1-3 triple knockout mice diet-induced obesity [88] and simulated shift work conditions [89]. In
[160]. The SCN receives feeding-related information, possibly from the humans, it has been shown that early (0800–1400 h) time restricted
FEO, and SCN neural output has been shown to be decreased before and feeding reduced appetite; making hunger more even-keeled, increasing
during daytime feeding when neural activity is usually high [167]. In feelings of fullness and decreasing the desire to eat [189]. This was
addition, SCN neurons are glucose sensitive, with glucose phase shifting associated with a decrease in the mean levels of the hunger hormone
the neural activity of cultured SCN slices [168]. Therefore, although the ghrelin and an increase in the satiety hormone PYY [189]. Further, in
expression of the clock genes in the SCN is closely linked to the humans limiting food intake to a 8–11 h period during the daytime
light:dark cycle, the SCN does sense the nutritional status of the body [141,190, 191] or eating a large breakfast and small dinner [192–194]
and influence food anticipatory activity. decreased appetite and/or food intake. It remains to be determined
Mice with disrupted dopamine signaling in the striatum do not de- whether a time restricted protocol for a limited number of days per
monstrate normal food anticipatory activity. Dopamine is involved in week, allowing for a more relaxed weekend lifestyle, will provide suf-
reward seeking behavior [169] and dopamine deficient mice do not ficient entrainment to maintain oscillations in appetite regulation on
forage for food [170]. Mice co-treated with dopamine D1 and D2 re- the days when the time restricted feeding protocol is not being fol-
ceptor antagonists exhibited reduced food anticipatory activity [171]. lowed.
Conversely, mice developed food anticipatory activity in response to
timed treatment with a dopamine D1 receptor agonist [172]. Further, 5. Conclusion
mice lacking a functional D1 receptor exhibited reduced food antici-
patory activity compared to wildtype controls [165,172], an effect not The circadian system plays an important role in the temporal reg-
observed in D2 knockout mice [172]. Together this data suggests that ulation of metabolic processes, such as anabolism and catabolism, and
reward regulation in the striatum, through dopamine signaling, med- food intake to increase energy efficiency. In conditions such as high fat
iates food anticipatory activity. diet-induced obesity or shift work there is a disruption in circadian
A number of hormones or peptides could also be involved in the rhythms and food intake patterns leading to a misalignment in meta-
modulation of food anticipatory activity. Studies indicate that feeding bolic processes and the development and/or maintenance of the obese
regulation centres in the DMN and ARC, through the action of neuro- state. It is still unclear whether the disruption in circadian clocks is
peptides such as ghrelin and orexin [164,173-175], might play a role in causing the disruption in food intake patterns or vice versa but it is clear
the food anticipatory activity. However, there is a study, using thermal that the timing of food intake is a strong zeitgeber regulating many
DMN lesions, which demonstrated that the DMN is not necessary for peripheral clocks, particularly in the gastrointestinal tract. Therefore,
food anticipatory activity [176]. Plasma ghrelin levels increase prior to time restricted feeding provides a mechanism by which circadian clocks
a meal and ghrelin activates dopaminergic reward pathways in the can be realigned. In addition, time restricted feeding has the capacity to
brain [177,178]. Ghrelin receptor knockout mice and rats treated with realign the circadian and FEO system maximising efficiency of food
a ghrelin receptor antagonist exhibit reduced food anticipatory activity intake, digestion and absorption in line with metabolic processes.
during restricted feeding protocols compared to controls
[174,179,180]. Conversely, preproghrelin (precursor for ghrelin) defi- Supplementary materials
cient and ghrelin deficient mice displayed normal food anticipatory
activity in response to restricted feeding protocols [181,182]. There- Supplementary material associated with this article can be found, in
fore, although ghrelin may modulate food anticipatory activity it is not the online version, at doi:10.1016/j.physbeh.2020.112873.
essential for this activity to occur. Food anticipatory behavior might
also involve the orexin signaling pathway. Orexin A and B are neuro- References
peptides that regulate arousal [183]. In orexin mutant mice, food an-
ticipatory locomotor activity (not body temperature) was reduced [1] S. Armstrong, A chronometric approach to the study of feeding behavior, Neurosci.
compared to wildtype controls [184,185]. In contrast, another study Biobehav. Rev. 4 (1980) 27–53.
[2] A.P. Patton, M.H. Hastings, The suprachiasmatic nucleus, Curr. Biol 28 (2018)
demonstrated food anticipatory activity was unchanged in orexin R816–RR22.
knockout mice [181]. Therefore, orexin may play a modulatory role in [3] M.A. Ma, E.H. Morrison, Neuroanatomy, Nucleus Suprachiasmatic, StatPearls,
food anticipatory behaviour but is unlikely to be critical for this an- Treasure Island (FL), 2019.
[4] M.K. Bunger, L.D. Wilsbacher, S.M. Moran, C. Clendenin, L.A. Radcliffe,
ticipatory activity to occur. J.B. Hogenesch, et al., Mop3 is an essential component of the master circadian
pacemaker in mammals, Cell 103 (2000) 1009–1017.
4.1.2. Time restricted feeding and appetite regulation [5] N. Gekakis, D. Staknis, H.B. Nguyen, F.C. Davis, L.D. Wilsbacher, D.P. King, et al.,
Role of the CLOCK protein in the mammalian circadian mechanism, Science 280
Experiments, both in animals and humans, have shown there is a (1998) 1564–1569.
circadian drive to appetite and hunger. In humans, under a constant-

6
A.J. Page, et al. Physiology & Behavior 220 (2020) 112873

[6] M. Reick, J.A. Garcia, C. Dudley, S.L. McKnight, NPAS2: an analog of clock op- [39] C. Sargent, X. Zhou, R.W. Matthews, D. Darwent, G.D. Roach, Daily rhythms of
erative in the mammalian forebrain, Science 293 (2001) 506–509. hunger and satiety in healthy men during one week of sleep restriction and cir-
[7] D.J. Kojetin, T.P. Burris, REV-ERB and ROR nuclear receptors as drug targets, Nat. cadian misalignment, Int. J. Environ. Res. Public Health 13 (2016) 170.
Rev. Drug Discov. 13 (2014) 197–216. [40] S.M.T. Wehrens, S. Christou, C. Isherwood, B. Middleton, M.A. Gibbs, S.N. Archer,
[8] F. Guillaumond, H. Dardente, V. Giguere, N. Cermakian, Differential control of et al., Meal timing regulates the human circadian system, Curr. Biol 27 (2017)
Bmal1 circadian transcription by REV-ERB and ROR nuclear receptors, J. Biol. 1768–1775 e3.
Rhythms 20 (2005) 391–403. [41] E. Poggiogalle, H. Jamshed, C.M. Peterson, Circadian regulation of glucose, lipid,
[9] N. Preitner, F. Damiola, L. Lopez-Molina, J. Zakany, D. Duboule, U. Albrecht, and energy metabolism in humans, Metabolism 84 (2018) 11–27.
et al., The orphan nuclear receptor REV-ERBalpha controls circadian transcription [42] E. Van Cauter, J.D. Blackman, D. Roland, J.P. Spire, S. Refetoff, K.S. Polonsky,
within the positive limb of the mammalian circadian oscillator, Cell 110 (2002) Modulation of glucose regulation and insulin secretion by circadian rhythmicity
251–260. and sleep, J. Clin. Invest. 88 (1991) 934–942.
[10] T.K. Sato, S. Panda, L.J. Miraglia, T.M. Reyes, R.D. Rudic, P. McNamara, et al., A [43] M.P. Carrasco-Benso, B. Rivero-Gutierrez, J. Lopez-Minguez, A. Anzola, A. Diez-
functional genomics strategy reveals Rora as a component of the mammalian Noguera, J.A. Madrid, et al., Human adipose tissue expresses intrinsic circadian
circadian clock, Neuron 43 (2004) 527–537. rhythm in insulin sensitivity, FASEB J. 30 (2016) 3117–3123.
[11] A. Balsalobre, Clock genes in mammalian peripheral tissues, Cell Tissue Res 309 [44] R.Y. Moore, Organization of the mammalian circadian system, Ciba Found. Symp.
(2002) 193–199. 183 (1995) 88–99 discussion 100-6.
[12] N. Cermakian, D.B. Boivin, The regulation of central and peripheral circadian [45] F.K. Stephan, K.J. Berkley, R.L. Moss, Efferent connections of the rat suprachias-
clocks in humans, Obes. Rev. 10 (Suppl 2) (2009) 25–36. matic nucleus, Neuroscience 6 (1981) 2625–2641.
[13] E.E. Zhang, S.A. Kay, Clocks not winding down: unravelling circadian networks, [46] N. Vrang, P.J. Larsen, M. Moller, J.D. Mikkelsen, Topographical organization of
Nat. Rev. Mol. Cell Biol. 11 (2010) 764–776. the rat suprachiasmatic-paraventricular projection, J. Comp. Neurol. 353 (1995)
[14] S. Ohdo, Chronotherapeutic strategy: Rhythm monitoring, manipulation and dis- 585–603.
ruption, Adv. Drug Deliv. Rev. 62 (2010) 859–875. [47] E. Challet, The circadian regulation of food intake, Nat. Rev. Endocrinol. 15
[15] O. Froy, Metabolism and circadian rhythms–implications for obesity, Endocr. Rev. (2019) 393–405.
31 (2010) 1–24. [48] X.Y. Lu, K.R. Shieh, M. Kabbaj, G.S. Barsh, H. Akil, S.J. Watson, Diurnal rhythm of
[16] R.E. Mistlberger, Food-anticipatory circadian rhythms: concepts and methods, Eur. agouti-related protein and its relation to corticosterone and food intake,
J. Neurosci. 30 (2009) 1718–1729. Endocrinology 143 (2002) 3905–3915.
[17] F.K. Stephan, The “other” circadian system: food as a Zeitgeber, J. Biol. Rhythms. [49] A.M. Stutz, J. Staszkiewicz, A. Ptitsyn, G. Argyropoulos, Circadian expression of
17 (2002) 284–292. genes regulating food intake, Obesity (Silver Spring) 15 (2007) 607–615.
[18] N.R. Lenard, H.R. Berthoud, Central and peripheral regulation of food intake and [50] B. Xu, P.S. Kalra, W.G. Farmerie, S.P. Kalra, Daily changes in hypothalamic gene
physical activity: pathways and genes, Obesity (Silver Spring) 16 (Suppl 3) (2008) expression of neuropeptide Y, galanin, proopiomelanocortin, and adipocyte leptin
S11–S22. gene expression and secretion: effects of food restriction, Endocrinology 140
[19] H.R. Berthoud, Multiple neural systems controlling food intake and body weight, (1999) 2868–2875.
Neurosci. Biobehav. Rev. 26 (2002) 393–428. [51] K. Saeb-Parsy, S. Lombardelli, F.Z. Khan, K. McDowall, I.T. Au-Yong, R.E. Dyball,
[20] S.A. Joseph, W.H. Pilcher, K.M. Knigge, Anatomy of the corticotropin-releasing Neural connections of hypothalamic neuroendocrine nuclei in the rat, J.
factor and opiomelanocortin systems of the brain, Fed. Proc. 44 (1985) 100–107. Neuroendocrinol. 12 (2000) 635–648.
[21] P. Kristensen, M.E. Judge, L. Thim, U. Ribel, K.N. Christjansen, B.S. Wulff, et al., [52] D.K. Sindelar, R.D. Palmiter, S.C. Woods, M.W. Schwartz, Attenuated feeding re-
Hypothalamic CART is a new anorectic peptide regulated by leptin, Nature 393 sponses to circadian and palatability cues in mice lacking neuropeptide Y, Peptides
(1998) 72–76. 26 (2005) 2597–2602.
[22] C. Broberger, J. Johansen, C. Johansson, M. Schalling, T. Hokfelt, The neuro- [53] S. Sen, S. Dumont, D. Sage-Ciocca, S. Reibel, P. de Goede, A. Kalsbeek, et al.,
peptide Y/agouti gene-related protein (AGRP) brain circuitry in normal, anorectic, Expression of the clock gene Rev-erbalpha in the brain controls the circadian or-
and monosodium glutamate-treated mice, Proc. Natl. Acad. Sci. USA 95 (1998) ganisation of food intake and locomotor activity, but not daily variations of energy
15043–15048. metabolism, J. Neuroendocrinol. 30 (2018).
[23] C. Broberger, Brain regulation of food intake and appetite: molecules and net- [54] J. Aschoff, C. von Goetz, C. Wildgruber, R.A. Wever, Meal timing in humans
works, J. Internal Med. 258 (2005) 301–327. during isolation without time cues, J. Biol. Rhythms. 1 (1986) 151–162.
[24] R. Poggioli, A.V. Vergoni, A. Bertolini, ACTH-(1-24) and alpha-MSH antagonize [55] K.C. Berridge, C.Y. Ho, J.M. Richard, A.G. DiFeliceantonio, The tempted brain
feeding behavior stimulated by kappa opiate agonists, Peptides 7 (1986) 843–848. eats: pleasure and desire circuits in obesity and eating disorders, Brain Res. 1350
[25] A.A. Butler, R.A. Kesterson, K. Khong, M.J. Cullen, M.A. Pelleymounter, (2010) 43–64.
J. Dekoning, et al., A unique metabolic syndrome causes obesity in the melano- [56] E. Egecioglu, K.P. Skibicka, C. Hansson, M. Alvarez-Crespo, P.A. Friberg,
cortin-3 receptor-deficient mouse, Endocrinology 141 (2000) 3518–3521. E. Jerlhag, et al., Hedonic and incentive signals for body weight control, Rev.
[26] A.A. Butler, D.L. Marks, W. Fan, C.M. Kuhn, M. Bartolome, R.D. Cone, Endocr. Metab. Disord. 12 (2011) 141–151.
Melanocortin-4 receptor is required for acute homeostatic responses to increased [57] R.A. Wise, M.A. Bozarth, A psychomotor stimulant theory of addiction, Psychol.
dietary fat, Nat. Neurosci. 4 (2001) 605–611. Rev. 94 (1987) 469–492.
[27] M. Rossi, M.S. Kim, D.G.A. Morgan, C.J. Small, C.M.B. Edwards, D. Sunter, et al., A [58] M.J. Ferris, R.A. Espana, J.L. Locke, J.K. Konstantopoulos, J.H. Rose, R. Chen,
C-Terminal fragment of agouti-related protein increases feeding and antagonizes et al., Dopamine transporters govern diurnal variation in extracellular dopamine
the effect of alpha-melanocyte stimulating hormone in Vivo, Endocrinology 139 tone, Proc. Natl. Acad. Sci. USA 111 (2014) E2751–E2759.
(1998) 4428–4431. [59] S. Hood, P. Cassidy, M.P. Cossette, Y. Weigl, M. Verwey, B. Robinson, et al.,
[28] B.G. Stanley, S.F. Leibowitz, Neuropeptide Y injected in the paraventricular hy- Endogenous dopamine regulates the rhythm of expression of the clock protein
pothalamus: a powerful stimulant of feeding behavior, Proc. Natl. Acad. Sci. USA PER2 in the rat dorsal striatum via daily activation of D2 dopamine receptors, J.
82 (1985) 3940–3943. Neurosci. 30 (2010) 14046–14058.
[29] T.M. Hahn, J.F. Breininger, D.G. Baskin, M.W. Schwartz, Coexpression of Agrp and [60] S.P. Kalra, M. Bagnasco, E.E. Otukonyong, M.G. Dube, P.S. Kalra, Rhythmic, re-
NPY in fasting-activated hypothalamic neurons, Nat. Neurosci. 1 (1998) 271–272. ciprocal ghrelin and leptin signaling: new insight in the development of obesity,
[30] Y. Hu, B.T. Bloomquist, L.J. Cornfield, L.B. DeCarr, J.R. Flores-Riveros, Regul. Pept 111 (2003) 1–11.
L. Friedman, et al., Identification of a novel hypothalamic neuropeptide Y receptor [61] W.A. Banks, A.J. Kastin, W. Huang, J.B. Jaspan, L.M. Maness, Leptin enters the
associated with feeding behavior, J. Biol. Chem. 271 (1996) 26315–26319. brain by a saturable system independent of insulin, Peptides 17 (1996) 305–311.
[31] J. Voisey, L. Carroll, A van Daal, Melanocortins and their receptors and antago- [62] C.F. Elias, J.F. Kelly, C.E. Lee, R.S. Ahima, D.J. Drucker, C.B. Saper, et al.,
nists, Curr. Drug Targets 4 (2003) 586–597. Chemical characterization of leptin-activated neurons in the rat brain, J. Comp.
[32] J.S. Sims, J.F. Lorden, Effect of paraventricular nucleus lesions on body weight, Neurol. 423 (2000) 261–281.
food intake and insulin levels, Behav. Brain Res. 22 (1986) 265–281. [63] H. Doring, K. Schwarzer, B. Nuesslein-Hildesheim, I. Schmidt, Leptin selectively
[33] J.C. Bittencourt, C.F. Elias, Melanin-concentrating hormone and neuropeptide EI increases energy expenditure of food-restricted lean mice, Int. J. Obes. Relat.
projections from the lateral hypothalamic area and zona incerta to the medial Metab. Disord. 22 (1998) 83–88.
septal nucleus and spinal cord: a study using multiple neuronal tracers, Brain Res. [64] P.J. Scarpace, M. Matheny, B.H. Pollock, N Tumer, Leptin increases uncoupling
805 (1998) 1–19. protein expression and energy expenditure, Am. J. Physiol.– Endocrinol. Metab.
[34] T. Sakurai, A. Amemiya, M. Ishii, I. Matsuzaki, R.M. Chemelli, H. Tanaka, et al., 273 (1997) E226–E230.
Orexins and orexin receptors: a family of hypothalamic neuropeptides and G [65] A. Caron, D. Richard, Neuronal systems and circuits involved in the control of food
protein-coupled receptors that regulate feeding behavior, Cell 92 (1998) 573–585. intake and adaptive thermogenesis, Ann. N.Y. Acad. Sci. 1391 (2017) 35–53.
[35] K. Krauchi, A. Wirz-Justice, Circadian rhythm of heat production, heart rate, and [66] Y. Zhang, R. Proenca, M. Maffei, M. Barone, L. Leopold, J.M. Friedman, Positional
skin and core temperature under unmasking conditions in men, Am. J. Physiol. cloning of the mouse obese gene and its human homologue, Nature 372 (1994)
267 (1994) R819–R829. 425–432.
[36] C.M. Spengler, C.A. Czeisler, S.A. Shea, An endogenous circadian rhythm of re- [67] V. Lecoultre, E. Ravussin, L.M. Redman, The fall in leptin concentration is a major
spiratory control in humans, J. Physiol. 526 (Pt 3) (2000) 683–694. determinant of the metabolic adaptation induced by caloric restriction in-
[37] F.A. Scheer, C.J. Morris, S.A. Shea, The internal circadian clock increases hunger dependently of the changes in leptin circadian rhythms, J. Clin. Endocrinol.
and appetite in the evening independent of food intake and other behaviors, Metab. 96 (2011) E1512–E1516.
Obesity (Silver Spring) 21 (2013) 421–423. [68] M.L. Wong, J. Licinio, B.O. Yildiz, C.S. Mantzoros, P. Prolo, M. Kling, et al.,
[38] D.S. Owens, I. Macdonald, D. Benton, N. Sytnik, P. Tucker, S. Folkard, A pre- Simultaneous and continuous 24-h plasma and cerebrospinal fluid leptin mea-
liminary investigation into individual differences in the circadian variation of surements: dissociation of concentrations in central and peripheral compartments,
meal tolerance: effects on mood and hunger, Chronobiol. Int. 13 (1996) 435–447. J. Clin. Endocrinol. Metab. 89 (2004) 258–265.

7
A.J. Page, et al. Physiology & Behavior 220 (2020) 112873

[69] A. Kalsbeek, E. Fliers, J.A. Romijn, S.E. La Fleur, J. Wortel, O. Bakker, et al., The [99] S. Cinti, R. De Matteis, E. Ceresi, C. Pico, J. Oliver, P. Oliver, et al., Leptin in the
suprachiasmatic nucleus generates the diurnal changes in plasma leptin levels, human stomach, Gut 49 (2001) 155.
Endocrinology 142 (2001) 2677–2685. [100] Y. Date, M. Kojima, H. Hosoda, A. Sawaguchi, M.S. Mondal, T. Suganuma, et al.,
[70] R. Heptulla, A. Smitten, B. Teague, W.V. Tamborlane, Y.Z. Ma, S. Caprio, Ghrelin, a novel growth hormone-releasing acylated peptide, is synthesized in a
Temporal patterns of circulating leptin levels in lean and obese adolescents: re- distinct endocrine cell type in the gastrointestinal tracts of rats and humans,
lationships to insulin, growth hormone, and free fatty acids rhythmicity, J. Clin. Endocrinology 141 (2000) 4255–4261.
Endocrinol. Metab. 86 (2001) 90–96. [101] M. Kojima, H. Hosoda, Y. Date, M. Nakazato, H. Matsuo, K. Kangawa, Ghrelin is a
[71] J. Licinio, Longitudinally sampled human plasma leptin and cortisol concentra- growth-hormonereleasing acylated peptide from stomach, Nature 402 (1999)
tions are inversely correlated, J. Clin. Endocrinol. Metab. 83 (1998) 1042. 656–660.
[72] B.O. Yildiz, M.A. Suchard, M.L. Wong, S.M. McCann, J. Licinio, Alterations in the [102] H.Y. Chen, M.E. Trumbauer, A.S. Chen, D.T. Weingarth, J.R. Adams, E.G. Frazier,
dynamics of circulating ghrelin, adiponectin, and leptin in human obesity, Proc. et al., Orexigenic action of peripheral ghrelin is mediated by neuropeptide Y and
Natl. Acad. Sci. USA 101 (2004) 10434–10439. agouti-related protein, Endocrinology 145 (2004) 2607–2612.
[73] W.A. Hoogerwerf, H.L. Hellmich, G. Cornélissen, F. Halberg, V.B. Shahinian, [103] D. Hagemann, J.J. Meier, B. Gallwitz, W.E. Schmidt, Appetite regulation by
J. Bostwick, et al., Clock gene expression in the murine gastrointestinal tract: ghrelin - a novel neuro-endocrine gastric peptide hormone in the gut-brain-axis, Z
endogenous rhythmicity and effects of a feeding regimen, Gastroenterology 133 Gastroenterol 41 (2003) 929–936.
(2007) 1250–1260. [104] P.C. Yannielli, P.C. Molyneux, M.E. Harrington, D.A. Golombek, Ghrelin effects on
[74] M. Sladek, M. Rybova, Z. Jindrakova, Z. Zemanova, L. Polidarova, L. Mrnka, et al., the circadian system of mice, J. Neurosci. 27 (2007) 2890–2895.
Insight into the circadian clock within rat colonic epithelial cells, Gastroenterology [105] C.X. Yi, E. Challet, P. Pevet, A. Kalsbeek, C. Escobar, R.M. Buijs, A circulating
133 (2007) 1240–1249. ghrelin mimetic attenuates light-induced phase delay of mice and light-induced
[75] L. Polidarova, M. Sladek, M. Sotak, J. Pacha, A. Sumova, Hepatic, duodenal, and Fos expression in the suprachiasmatic nucleus of rats, Eur. J. Neurosci. 27 (2008)
colonic circadian clocks differ in their persistence under conditions of constant 1965–1972.
light and in their entrainment by restricted feeding, Chronobiol. Int. 28 (2011) [106] D.E. Cummings, D.S. Weigle, R.S. Frayo, P. Breen, M.K. Ma, P.E. Dellinger, et al.,
204–215. Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery, N.
[76] G.A. Bjarnason, R. Jordan, Rhythms in human gastrointestinal mucosa and skin, Engl. J. Med 346 (2002) 1623–1630.
Chronobiol. Int. 19 (2002) 129–140. [107] A.A. Moghadam, T.H. Moran, M.J. Dailey, Alterations in circadian and meal-in-
[77] L.A. Scheving, W.E. Russell, It's about time: clock genes unveiled in the gut, duced gut peptide levels in lean and obese rats, Exp. Biol. Med. (Maywood) 242
Gastroenterology 133 (2007) 1373–1376. (2017) 1786–1794.
[78] H.J. Grill, R Norgren, Chronically decerebrate rats demonstrate satiation but not [108] K. Lundberg, A. Josefsson, C. Nordin, Diurnal and seasonal variation of chole-
bait shyness, Science 201 (1978) 267–269. cystokinin peptides in humans, Neuropeptides 41 (2007) 59–63.
[79] A.J. Page, L.A. Blackshaw, An in vitro study of the properties of vagal afferent [109] M. Soty, A. Gautier-Stein, F. Rajas, G. Mithieux, Gut-brain glucose signaling in
fibres innervating the ferret oesophagus and stomach, J. Physiol. 512 (1998) energy homeostasis, Cell Metab. 25 (2017) 1231–1242.
907–916. [110] Y. Tahara, M. Yamazaki, H. Sukigara, H. Motohashi, H. Sasaki, H. Miyakawa,
[80] A.J. Page, C.M. Martin, L.A. Blackshaw, Vagal mechanoreceptors and chemor- et al., Gut microbiota-derived short chain fatty acids induce circadian clock en-
eceptors in mouse stomach and esophagus, J. Neurophysiol. 87 (2002) trainment in mouse peripheral tissue, Sci. Rep. 8 (2018) 1395.
2095–2103. [111] S.O. Fetissov, Role of the gut microbiota in host appetite control: bacterial growth
[81] S.J. Kentish, T.A. O’Donnell, N.J. Isaacs, R.L. Young, H. Li, A.M. Harrington, et al., to animal feeding behaviour, Nat. Rev. Endocrinol. 13 (2017) 11–25.
Gastric vagal afferent modulation by leptin is influenced by food intake status, J. [112] A. Mukherji, A. Kobiita, T. Ye, P. Chambon, Homeostasis in intestinal epithelium is
Physiol. 591 (2013) 1921–1934. orchestrated by the circadian clock and microbiota cues transduced by TLRs, Cell
[82] A.J. Page, J.A. Slattery, C. Milte, R. Laker, T. O'Donnell, C. Dorian, et al., Ghrelin 153 (2013) 812–827.
selectively reduces mechanosensitivity of upper gastrointestinal vagal afferents, [113] C.A. Thaiss, M. Levy, T. Korem, L. Dohnalova, H. Shapiro, D.A. Jaitin, et al.,
Am. J. Physiol. Gastrointest. Liver Physiol. 292 (2007) G1376–G1384. Microbiota diurnal rhythmicity programs host transcriptome oscillations, Cell. 167
[83] S. Kentish, H. Li, L.K. Philp, T.A. O'Donnell, N.J. Isaacs, R.L. Young, et al., Diet- (2016) 1495–1510 e12.
induced adaptation of vagal afferent function, J. Physiol. 590 (2012) 209–221. [114] V. Leone, S.M. Gibbons, K. Martinez, A.L. Hutchison, E.Y. Huang, C.M. Cham,
[84] E.L. Symonds, M. Peiris, A.J. Page, B. Chia, H. Dogra, A. Masding, et al., et al., Effects of diurnal variation of gut microbes and high-fat feeding on host
Mechanisms of activation of mouse and human enteroendocrine cells by nutrients, circadian clock function and metabolism, Cell Host Microbe 17 (2015) 681–689.
Gut 64 (2015) 618–626. [115] X. Liang, F.D. Bushman, G.A. FitzGerald, Rhythmicity of the intestinal microbiota
[85] A.J. Page, E. Symonds, M. Peiris, L.A. Blackshaw, R.L. Young, Peripheral neural is regulated by gender and the host circadian clock, Proc. Natl. Acad. Sci. USA 112
targets in obesity, Brit. J. Pharmacol. 166 (2012) 1537–1558. (2015) 10479–10484.
[86] L. Bai, S. Mesgarzadeh, K.S. Ramesh, E.L. Huey, Y. Liu, L.A. Gray, et al., Genetic [116] J.L. Kaczmarek, S.M. Musaad, H.D. Holscher, Time of day and eating behaviors are
identification of vagal sensory neurons that control feeding, Cell 179 (2019) associated with the composition and function of the human gastrointestinal mi-
1129–1143 e23. crobiota, Am. J. Clin. Nutr. 106 (2017) 1220–1231.
[87] S.J. Kentish, C.L. Frisby, D.J. Kennaway, G.A. Wittert, A.J. Page, Circadian var- [117] C.A. Thaiss, D. Zeevi, M. Levy, G. Zilberman-Schapira, J. Suez, A.C. Tengeler,
iation in gastric vagal afferent mechanosensitivity, J. Neurosci. 33 (2013) et al., Transkingdom control of microbiota diurnal oscillations promotes metabolic
19238–19242. homeostasis, Cell 159 (2014) 514–529.
[88] S.J. Kentish, A.D. Vincent, D.J. Kennaway, G.A. Wittert, A.J. Page, High-fat diet- [118] R.M. Voigt, C.B. Forsyth, S.J. Green, E. Mutlu, P. Engen, M.H. Vitaterna, et al.,
induced obesity ablates gastric vagal afferent circadian rhythms, J. Neurosci. 36 Circadian disorganization alters intestinal microbiota, PLoS One. 9 (2014) e97500.
(2016) 3199–3207. [119] A. Boulamery-Velly, N. Simon, J. Vidal, J. Mouchet, B. Bruguerolle, Effects of
[89] S.J. Kentish, S. Christie, A. Vincent, H. Li, G.A. Wittert, A.J. Page, Disruption of the three-hour restricted food access during the light period on circadian rhythms of
light cycle ablates diurnal rhythms in gastric vagal afferent mechanosensitivity, temperature, locomotor activity, and heart rate in rats, Chronobiol. Int. 22 (2005)
Neurogastroenterol. Motil. 31 (2019) e13711. 489–498.
[90] J. Fatima, C.W. Iqbal, S.G. Houghton, M.S. Kasparek, J.A. Duenes, Y. Zheng, et al., [120] C.A. Comperatore, F.K. Stephan, Entrainment of duodenal activity to periodic
Hexose transporter expression and function in mouse small intestine: role of feeding, J. Biol. Rhythms. 2 (1987) 227–242.
diurnal rhythm, J. Gastrointest. Surg. 13 (2009) 634–641. [121] S.A. Brown, G. Zumbrunn, F. Fleury-Olela, N. Preitner, U. Schibler, Rhythms of
[91] X. Pan, T. Terada, M. Irie, H. Saito, K. Inui, Diurnal rhythm of H+-peptide co- mammalian body temperature can sustain peripheral circadian clocks, Curr. Biol.
transporter in rat small intestine, Am. J. Physiol. Gastrointest. Liver Physiol. 283 12 (2002) 1574–1583.
(2002) G57–G64. [122] O.M. Buxton, C.W. Lee, M. L’Hermite-Baleriaux, F.W. Turek, E. Van Cauter,
[92] X. Pan, T. Terada, M. Okuda, K. Inui, The diurnal rhythm of the intestinal trans- Exercise elicits phase shifts and acute alterations of melatonin that vary with
porters SGLT1 and PEPT1 is regulated by the feeding conditions in rats, J. Nutr. circadian phase, Am. J. Physiol. Regul. Integr. Comp. Physiol. 284 (2003)
134 (2004) 2211–2215. R714–R724.
[93] D.B. Rhoads, D.H. Rosenbaum, H. Unsal, K.J. Isselbacher, L.L. Levitsky, Circadian [123] U. Schibler, J. Ripperger, S.A. Brown, Peripheral circadian oscillators in mammals:
periodicity of intestinal Na+/glucose cotransporter 1 mRNA levels is tran- time and food, J. Biol. Rhythms 18 (2003) 250–260.
scriptionally regulated, J. Biol. Chem. 273 (1998) 9510–9516. [124] F. Damiola, N. Le Minh, N. Preitner, B. Kornmann, F. Fleury-Olela, U. Schibler,
[94] H. Saito, T. Terada, J. Shimakura, T. Katsura, K. Inui, Regulatory mechanism Restricted feeding uncouples circadian oscillators in peripheral tissues from the
governing the diurnal rhythm of intestinal H+/peptide cotransporter 1 (PEPT1), central pacemaker in the suprachiasmatic nucleus, Genes. Dev. 14 (2000)
Am. J. .Physiol. Gastrointest. Liver Physiol. 295 (2008) G395–G402. 2950–2961.
[95] H. Bhutta, T. Deelman, S. Ashley, D. Rhoads, A. Tavakkoli, Disrupted circadian [125] K.A. Stokkan, S. Yamazaki, H. Tei, Y. Sakaki, M. Menaker, Entrainment of the
rhythmicity of the intestinal glucose transporter SGLT1 in Zucker diabetic fatty circadian clock in the liver by feeding, Science 291 (2001) 490–493.
rats, Digest. Dis. Sci. 58 (2013) 1537–1545. [126] L.K. Fonken, J.L. Workman, J.C. Walton, Z.M. Weil, J.S. Morris, A. Haim, et al.,
[96] Segers, A., Desmet, L., Thijs, T., Verbeke, K., Tack, J., Depoortere, I.The circadian Light at night increases body mass by shifting the time of food intake, Proc. Natl.
clock regulates the diurnal levels of microbial short-chain fatty acids and their Acad. Sci. USA 107 (2010) 18664–18669.
rhythmic effects on colon contractility in mice. Acta Physiol. (Oxf.). 2018:e13193. [127] A. Lowden, C. Moreno, U. Holmback, M. Lennernas, P. Tucker, Eating and shift
[97] A. Bado, S. Levasseur, S. Attoub, S. Kermorgant, J.P. Laigneau, M.N. Bortoluzzi, work - effects on habits, metabolism and performance, Scand. J. Work Environ.
et al., The stomach is a source of leptin, Nature 394 (1998) 790–793. Health 36 (2010) 150–162.
[98] S. Cinti, R. De Matteis, C. Picoa, E. Ceresi, A. Obrador, C. Maffeis, et al., Secretory [128] Y. Morikawa, H. Nakagawa, K. Miura, Y. Soyama, M. Ishizaki, T. Kido, et al., Effect
granules of endocrine and chief cells of human stomach mucosa contain leptin, Int. of shift work on body mass index and metabolic parameters, Scand. J. Work
J. Obes. (Lond.). 24 (2000) 789–793. Environ. Health 33 (2007) 45–50.

8
A.J. Page, et al. Physiology & Behavior 220 (2020) 112873

[129] Australian Bureau of Statistics, Working Time Arrangements, Australia, November activity, Sci. Rep. 7 (2017) 15510.
2012, ABS, Canberra, Australia, 2012. [162] K.F. Storch, C.J. Weitz, Daily rhythms of food-anticipatory behavioral activity do
[130] P.T. Agnes, B. Isabella, C. Jorge, V.L. Oscar, V. Greet, W., A., et al., Sixth European not require the known circadian clock, Proc. Natl. Acad. Sci. US.A. 106 (2009)
Working Conditions Survey—Overview Report, Eurofound, Brussels, Belgium, 6808–6813.
2016. [163] C.A. Feillet, J.A. Ripperger, M.C. Magnone, A. Dulloo, U. Albrecht, E. Challet, Lack
[131] Bureau of Labor Statistics, Workers on Flexible and Shift Schedules in 2004 of food anticipation in Per2 mutant mice, Curr. Biol. 16 (2006) 2016–2022.
Summary, U.S. Bureau of Labor Statistics, 2005. [164] G. Acosta-Galvan, C.X. Yi, J. van der Vliet, J.H. Jhamandas, P. Panula, M. Angeles-
[132] A. Knutsson, Health disorders of shift workers, Occup. Med. (Lond.) 53 (2003) Castellanos, et al., Interaction between hypothalamic dorsomedial nucleus and the
103–108. suprachiasmatic nucleus determines intensity of food anticipatory behavior, Proc.
[133] K.J. Vener, S. Szabo, J.G. Moore, The effect of shift work on gastrointestinal (GI) Natl. Acad. Sci. USA 108 (2011) 5813–5818.
function: a review, Chronobiologia 16 (1989) 421–439. [165] M. Michalik, A.D. Steele, R.E. Mistlberger, A sex difference in circadian food-an-
[134] M. Lennernas, L. Hambraeus, T. Akerstedt, Shift related dietary intake in day and ticipatory rhythms in mice: interaction with dopamine D1 receptor knockout,
shift workers, Appetite 25 (1995) 253–265. Behav. Neurosci. 129 (2015) 351–360.
[135] I.C. Pasqua, C.R. Moreno, The nutritional status and eating habits of shift workers: [166] C.A. Dudley, C. Erbel-Sieler, S.J. Estill, M. Reick, P. Franken, S. Pitts, et al., Altered
a chronobiological approach, Chronobiol. Int. 21 (2004) 949–960. patterns of sleep and behavioral adaptability in NPAS2-deficient mice, Science 301
[136] A. Pan, E.S. Schernhammer, Q. Sun, F.B. Hu, Rotating night shift work and risk of (2003) 379–383.
type 2 diabetes: two prospective cohort studies in women, PLoS Med. 8 (2011) [167] T. Dattolo, C.P. Coomans, H.C. van Diepen, D.F. Patton, S. Power, M.C. Antle,
e1001141. et al., Neural activity in the suprachiasmatic circadian clock of nocturnal mice
[137] E.A. Cayanan, N.A.B. Eyre, V. Lao, M. Comas, C.M. Hoyos, N.S. Marshall, et al., Is anticipating a daytime meal, Neuroscience 315 (2016) 91–103.
24-h energy intake greater during night shift compared to non-night shift patterns? [168] A.C. Hall, R.M. Hoffmaster, E.L. Stern, M.E. Harrington, D. Bickar,
A systematic review, Chronobiol. Int. 36 (2019) 1599–1612. Suprachiasmatic nucleus neurons are glucose sensitive, J. Biol. Rhythms 12 (1997)
[138] E. Shaw, J. Dorrian, A.M. Coates, G.K.W. Leung, R. Davis, E. Rosbotham, et al., 388–400.
Temporal pattern of eating in night shift workers, Chronobiol. Int. 36 (2019) [169] J.H. Baik, Dopamine signaling in reward-related behaviors, Front. Neural. Circuit.
1613–1625. 7 (2013) 152.
[139] M. Romon, J.L. Edme, C. Boulenguez, J.L. Lescroart, P. Frimat, Circadian variation [170] M.S. Szczypka, M.A. Rainey, D.S. Kim, W.A. Alaynick, B.T. Marck,
of diet-induced thermogenesis, Am. J. Clin. Nutr 57 (1993) 476–480. A.M. Matsumoto, et al., Feeding behavior in dopamine-deficient mice, Proc. Natl.
[140] C.L. Ogden, M.D. Carroll, B.K. Kit, K.M. Flegal, Prevalence of childhood and adult Acad. Sci. USA 96 (1999) 12138–12143.
obesity in the United States, 2011–2012, JAMA 311 (2014) 806–814. [171] Y.Y. Liu, T.Y. Liu, W.M. Qu, Z.Y. Hong, Y. Urade, Z.L. Huang, Dopamine is in-
[141] S. Gill, S. Panda, A smartphone app reveals erratic diurnal eating patterns in hu- volved in food-anticipatory activity in mice, J. Biol. Rhythms 27 (2012) 398–409.
mans that can be modulated for health benefits, Cell Metab. 22 (2015) 789–798. [172] C.M. Gallardo, M. Darvas, M. Oviatt, C.H. Chang, M. Michalik, T.F. Huddy, et al.,
[142] T.G. Aubrecht, R. Jenkins, R.J. Nelson, Dim light at night increases body mass of Dopamine receptor 1 neurons in the dorsal striatum regulate food anticipatory
female mice, Chronobiol. Int. 32 (2015) 557–560. circadian activity rhythms in mice, Elife 3 (2014) e03781.
[143] A. Kohsaka, A.D. Laposky, K.M. Ramsey, C. Estrada, C. Joshu, Y. Kobayashi, et al., [173] M. Akiyama, T. Yuasa, N. Hayasaka, K. Horikawa, T. Sakurai, S. Shibata, Reduced
High-fat diet disrupts behavioral and molecular circadian rhythms in mice, Cell food anticipatory activity in genetically orexin (hypocretin) neuron-ablated mice,
Metab. 6 (2007) 414–421. Eur. J. Neurosci. 20 (2004) 3054–3062.
[144] M. Barnea, Z. Madar, O. Froy, High-fat diet delays and fasting advances the cir- [174] J. LeSauter, N. Hoque, M. Weintraub, D.W. Pfaff, R. Silver, Stomach ghrelin-se-
cadian expression of adiponectin signaling components in mouse liver, creting cells as food-entrainable circadian clocks, Proc. Natl. Acad. Sci. USA 106
Endocrinology 150 (2009) 161–168. (2009) 13582–13587.
[145] M. Barnea, Z. Madar, O. Froy, High-fat diet followed by fasting disrupts circadian [175] M. Mieda, S.C. Williams, C.M. Sinton, J.A. Richardson, T. Sakurai, M. Yanagisawa,
expression of adiponectin signaling pathway in muscle and adipose tissue, Obesity Orexin neurons function in an efferent pathway of a food-entrainable circadian
(Silver Spring) 18 (2010) 230–238. oscillator in eliciting food-anticipatory activity and wakefulness, J. Neurosci. 24
[146] P. Cano, V. Jimenez-Ortega, A. Larrad, C.F. Reyes Toso, D.P. Cardinali, (2004) 10493–10501.
A.I. Esquifino, Effect of a high-fat diet on 24-h pattern of circulating levels of [176] T. Moriya, R. Aida, T. Kudo, M. Akiyama, M. Doi, N. Hayasaka, et al., The dor-
prolactin, luteinizing hormone, testosterone, corticosterone, thyroid-stimulating somedial hypothalamic nucleus is not necessary for food-anticipatory circadian
hormone and glucose, and pineal melatonin content, in rats, Endocrine 33 (2008) rhythms of behavior, temperature or clock gene expression in mice, Eur. J.
118–125. Neurosci. 29 (2009) 1447–1460.
[147] M.C. Cha, C.J. Chou, C.N. Boozer, High-fat diet feeding reduces the diurnal var- [177] A. Abizaid, Z.W. Liu, Z.B. Andrews, M. Shanabrough, E. Borok, J.D. Elsworth,
iation of plasma leptin concentration in rats, Metabolism 49 (2000) 503–507. et al., Ghrelin modulates the activity and synaptic input organization of midbrain
[148] P.J. Havel, R. Townsend, L. Chaump, K. Teff, High-fat meals reduce 24-h circu- dopamine neurons while promoting appetite, J. Clin. Invest. 116 (2006)
lating leptin concentrations in women, Diabetes 48 (1999) 334–341. 3229–3239.
[149] A. Chaix, E.N.C. Manoogian, G.C. Melkani, S Panda, Time-restricted eating to [178] B. Bodosi, J. Gardi, I. Hajdu, E. Szentirmai, F. Obal Jr., J.M. Krueger, Rhythms of
prevent and manage chronic metabolic diseases, Annu Rev Nutr 39 (2019) ghrelin, leptin, and sleep in rats: effects of the normal diurnal cycle, restricted
291–315. feeding, and sleep deprivation, Am. J. Physiol. Regul. Integr. Comp. Physiol. 287
[150] S. Christie, A.D. Vincent, H. Li, C.L. Frisby, S.J. Kentish, R. O’Rielly, et al., A ro- (2004) R1071–R1079.
tating light cycle promotes weight gain and hepatic lipid storage in mice, Am. J. [179] I.D. Blum, Z. Patterson, R. Khazall, E.W. Lamont, M.W. Sleeman, T.L. Horvath,
Physiol. Gastrointest. Liver Physiol. 315 (2018) G932–G942. et al., Reduced anticipatory locomotor responses to scheduled meals in ghrelin
[151] M. Hatori, C. Vollmers, A. Zarrinpar, L. DiTacchio, E.A. Bushong, S. Gill, et al., receptor deficient mice, Neuroscience 164 (2009) 351–359.
Time-restricted feeding without reducing caloric intake prevents metabolic dis- [180] M. Merkestein, M.A. Brans, M.C. Luijendijk, J.W. de Jong, E. Egecioglu,
eases in mice fed a high-fat diet, Cell Metab. 15 (2012) 848–860. S.L. Dickson, et al., Ghrelin mediates anticipation to a palatable meal in rats,
[152] H. Oike, M. Sakurai, K. Ippoushi, M. Kobori, Time-fixed feeding prevents obesity Obesity (Silver Spring) 20 (2012) 963–971.
induced by chronic advances of light/dark cycles in mouse models of jet-lag/shift [181] K.M. Gunapala, C.M. Gallardo, C.T. Hsu, A.D. Steele, Single gene deletions of
work, Biochem. Biophys. Res. Commun. 465 (2015) 556–561. orexin, leptin, neuropeptide Y, and ghrelin do not appreciably alter food antici-
[153] Y. Tahara, S Shibata, Chronobiology and nutrition, Neuroscience 253 (2013) patory activity in mice, PLoS One 6 (2011) e18377.
78–88. [182] E. Szentirmai, L. Kapas, Y. Sun, R.G. Smith, J.M. Krueger, Restricted feeding-in-
[154] A. Chaix, T. Lin, H.D. Le, M.W. Chang, S. Panda, Time-restricted feeding prevents duced sleep, activity, and body temperature changes in normal and preproghrelin-
obesity and metabolic syndrome in mice lacking a circadian clock, Cell Metab. 29 deficient mice, Am. J. Physiol. Regul. Integr. Comp. Physiol. 298 (2010)
(2019) 303–319 e4. R467–R477.
[155] K. Horikawa, Y. Minami, M. Iijima, M. Akiyama, S. Shibata, Rapid damping of [183] C. Alexandre, M.L. Andermann, T.E. Scammell, Control of arousal by the orexin
food-entrained circadian rhythm of clock gene expression in clock-defective per- neurons, Curr. Opin. Neurobiol. 23 (2013) 752–759.
ipheral tissues under fasting conditions, Neuroscience 134 (2005) 335–343. [184] S. Kaur, S. Thankachan, S. Begum, C. Blanco-Centurion, T. Sakurai,
[156] S. Pitts, E. Perone, R. Silver, Food-entrained circadian rhythms are sustained in M. Yanagisawa, et al., Entrainment of temperature and activity rhythms to re-
arrhythmic Clk/Clk mutant mice, Am. J. Physiol. Regul. Integr. Comp. Physiol. stricted feeding in orexin knock out mice, Brain Res 1205 (2008) 47–54.
285 (2003) R57–R67. [185] E.L. Clark, C.R. Baumann, G. Cano, T.E. Scammell, T. Mochizuki, Feeding-elicited
[157] M. Iijima, S. Yamaguchi, G.T. van der Horst, X. Bonnefont, H. Okamura, S. Shibata, cataplexy in orexin knockout mice, Neuroscience 161 (2009) 970–977.
Altered food-anticipatory activity rhythm in Cryptochrome-deficient mice, [186] Z. Liu, M. Huang, X. Wu, G. Shi, L. Xing, Z. Dong, et al., PER1 phosphorylation
Neurosci. Res. 52 (2005) 166–173. specifies feeding rhythm in mice, Cell Rep. 7 (2014) 1509–1520.
[158] Z. Li, Y. Wang, K.K. Sun, K. Wang, Z.S. Sun, M. Zhao, et al., Sex-related difference [187] J. Cedernaes, W. Huang, K.M. Ramsey, N. Waldeck, L. Cheng, B. Marcheva, et al.,
in food-anticipatory activity of mice, Horm. Behav. 70 (2015) 38–46. Transcriptional basis for rhythmic control of hunger and metabolism within the
[159] J. Mendoza, U. Albrecht, E. Challet, Behavioural food anticipation in clock genes AgRP neuron, Cell Metab. 29 (2019) 1078–1091 e5.
deficient mice: confirming old phenotypes, describing new phenotypes, Genes [188] S.J. Kentish, G. Hatzinikolas, H. Li, C.L. Frisby, G.A. Wittert, A.J. Page, Time-
Brain Behav. 9 (2010) 467–477. restricted feeding prevents ablation of diurnal rhythms in gastric vagal afferent
[160] J.S. Pendergast, G.A. Oda, K.D. Niswender, S. Yamazaki, Period determination in mechanosensitivity observed in high-fat diet-induced obese mice, J. Neurosci. 38
the food-entrainable and methamphetamine-sensitive circadian oscillator(s), Proc. (2018) 5088–5095.
Natl. Acad. Sci. USA 109 (2012) 14218–14223. [189] E. Ravussin, R.A. Beyl, E. Poggiogalle, D.S. Hsia, C.M. Peterson, Early time-re-
[161] J.S. Pendergast, R.H. Wendroth, R.C. Stenner, C.D. Keil, S. Yamazaki, mPeriod2 stricted feeding reduces appetite and increases fat oxidation but does not affect
(Brdm1) and other single Period mutant mice have normal food anticipatory energy expenditure in humans, Obesity (Silver Spring) 27 (2019) 1244–1254.

9
A.J. Page, et al. Physiology & Behavior 220 (2020) 112873

[190] K. Gabel, K.K. Hoddy, K.A. Varady, Safety of 8-h time restricted feeding in adults Obesity (Silver Spring) 21 (2013) 2504–2512.
with obesity, Appl. Physiol. Nutr. Metab. 44 (2019) 107–109. [193] H.R. Rabinovitz, M. Boaz, T. Ganz, D. Jakubowicz, Z. Matas, Z. Madar, et al., Big
[191] J.D. LeCheminant, E. Christenson, B.W. Bailey, L.A. Tucker, Restricting night-time breakfast rich in protein and fat improves glycemic control in type 2 diabetics,
eating reduces daily energy intake in healthy young men: a short-term cross-over Obesity (Silver Spring) 22 (2014) E46–E54.
study, Br. J. Nutr. 110 (2013) 2108–2113. [194] K.J. Reid, K.G. Baron, P.C. Zee, Meal timing influences daily caloric intake in
[192] D. Jakubowicz, M. Barnea, J. Wainstein, O. Froy, High caloric intake at breakfast healthy adults, Nutr. Res. 34 (2014) 930–935.
vs. dinner differentially influences weight loss of overweight and obese women,

10

You might also like