You are on page 1of 11

nature reviews endocrinology https://doi.org/10.

1038/s41574-023-00927-z

Review article Check for updates

The intersection between ghrelin,


metabolism and circadian rhythms
Soumya S. Kulkarni1,2, Omprakash Singh 2
& Jeffrey M. Zigman 2,3,4

Abstract Sections

Despite the growing popular interest in sleep and diet, many gaps exist Introduction

in our scientific understanding of the interaction between circadian Circadian biology and
rhythms and metabolism. In this Review, we explore a promising, metabolism

bidirectional role for ghrelin in mediating this interaction. Ghrelin Regulation and metabolic
actions of ghrelin and LEAP2
both influences and is influenced by central and peripheral circadian
systems. Specifically, we focus on how ghrelin impacts outputs of Ghrelin action in the brain

circadian rhythm, including neuronal activity, circulating growth Effects of ghrelin on circadian
rhythm
hormone levels, locomotor activity and eating behaviour. We also
consider the effects of circadian rhythms on ghrelin expression and Influence of circadian
rhythms on ghrelin and
the consequences of disrupted circadian patterns, such as shift work GHSR expression
and jet lag, on ghrelin secretion. Our Review is aimed at both the casual
Relevance of the work in
reader interested in gaining more insight into the scientific context mouse models to human
health and disease
surrounding the trending topics of sleep and metabolism, as well as
experienced scientists in the fields of ghrelin and circadian biology Conclusions

seeking inspiration and a comprehensive overview of how these fields


are related.

Medical Scientist Training Program, UT Southwestern Medical Center, Dallas, TX, USA. 2Center for Hypothalamic
1

Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA. 3Division of
Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA. 4Department
of Psychiatry, UT Southwestern Medical Center, Dallas, TX, USA. e-mail: jeffrey.zigman@utsouthwestern.edu

Nature Reviews Endocrinology


Review article

Key points circadian and metabolic systems. A growing area of concern is the
effects of working the nightshift on human health. A study of over 300
health-care workers found that those who worked the nightshift had
•• Disrupted circadian and metabolic systems are a growing cause of significantly elevated waist circumference, fasting blood glucose and
societal health burden. high-sensitivity C-reactive protein (a marker for the risk of developing
coronary artery disease)14.
•• The ghrelin system regulates many key metabolic functions Another growing area of concern is abnormal eating patterns, such
including food intake, body weight, blood glucose, food anticipatory as eating during resting periods15. Healthy young women who engaged
activity, growth hormone secretion and body temperature. in night-time snacking for just 13 days exhibited decreases in whole
body fat oxidation and increases in LDL cholesterol, both of which
•• Ghrelin and GHSR (growth hormone secretagogue receptor, which are risk factors for obesity and metabolic disease16. Late-night snack-
serves as the receptor for ghrelin) mRNA and protein expression are ing is associated with increased hunger, decreased waketime energy
regulated by the circadian system. expenditure and 24-h core body temperature, altered lipid metabolism
pathways, and, of relevance to this Review, an increased ratio of plasma
•• Ghrelin affects neuronal firing, food anticipatory activity and ghrelin to the anorexigenic adipose tissue-derived hormone leptin17.
thermoregulation in a circadian-dependent manner. Although not all individuals who engage in night-time snacking
have an eating disorder, some individuals who frequently engage in
•• Given the expansion in scientific technology in the past 25 years, night-time snacking meet criteria for night-eating syndrome. These
many more tools now exist to better understand the role of the ghrelin criteria include morning anorexia, consumption of over 50% of daily
system in metabolism and circadian biology. energy intake after the last evening meal, waking up at least once a
night to eat, exclusion of bulimia nervosa or binge-eating disorder,
•• Future studies are needed to elucidate further the connections and persistence of symptoms for at least 3 months18. Individuals with
between the ghrelin system, metabolism and circadian systems, night-eating syndrome display altered circadian physiology, including
and to provide novel avenues for therapeutic approaches. phase delays in timing of meals and in leptin and insulin rhythms19. By
contrast, circulating ghrelin levels in individuals with night-eating
syndrome show a phase advance of 5.2 h19. Given the severity and preva-
Introduction lence of the effects of aberrant circadian and metabolic regulation
Ghrelin is an acylated peptide hormone secreted mainly from an elec- on society, more steps must be taken to better understand how the
tron microscopically distinct type of enteroendocrine cell that is now circadian and metabolic systems interact. Exploring whether and how
known as a ghrelin cell. Calorie restriction, stress and poor sleep all the ghrelin system may be involved in this relationship is a rational
increase ghrelin secretion. By contrast, food intake and obesity lower place to begin.
plasma ghrelin levels. In adults, ghrelin cells are localized primarily In this Review, we touch on some salient evidence linking circadian
to the lining of the stomach and duodenum (Fig. 1). About one out of biology and metabolism. We briefly provide an overview of the ghrelin
every 100–300 gastric mucosal cells is a ghrelin cell. The receptor to system and its key players — ghrelin, GHSR, LEAP2 and GOAT — and
which ghrelin binds and through which ghrelin acts is the growth hor- discuss their physiological functions and effects on the brain. We then
mone secretagogue receptor (GHSR). GHSRs are localized to several take a more specific look at the effects of ghrelin on circadian biology,
tissues, but have perhaps been best characterized within the brain, including its roles in regulating neuronal activity, locomotor and food
pituitary and pancreatic islets. Ghrelin binding to GHSRs depends on anticipatory behaviours and body temperature. Similarly, we consider
ghrelin’s acyl group — most usually, an octanoyl group — which is added the effects of circadian rhythms on ghrelin secretion and activity,
as a post-translational modification catalysed by the enzyme ghrelin including what is known regarding this topic in humans, peripheral and
O-acyltransferase (GOAT). Liver-expressed antimicrobial peptide 2 central effects of circadian rhythmicity on ghrelin, and time-restricted
(LEAP2), a peptide hormone that is secreted mainly from hepatocytes feeding. Finally, we explore some clinically relevant models such as
and jejunal enterocytes, also binds to GHSR. Unlike ghrelin, LEAP2 chronic jetlag, provide an overview of open research questions, and
serves as an antagonist and inverse agonist at GHSRs, powerfully block- give our suggestions on how to best begin to tackle them.
ing ghrelin action while also decreasing GHSR constitutive activity.
When first reported in 1999 (refs. 1,2), ghrelin’s actions as an agonist Circadian biology and metabolism
for GHSR and a potent growth hormone secretagogue were the focus. An overview of some basic circadian terminology is shown (Box 1).
Yet, ghrelin has since been implicated in several well-known functions The interaction between circadian biology and metabolism has been
including meal initiation3–5, appetite stimulation (reviewed elsewhere6), extensively discussed previously20–22. Here, we discuss only the key
reducing energy expenditure2, raising blood glucose and preventing findings that lay the foundation for the subsequent discussion. Dys-
life-threatening falls in blood glucose7–9, stimulating gastrointestinal regulation of circadian clock genes or zeitgebers can severely impact
motility2, and inducing reward behaviours related to food, alcohol and metabolism. Both Turek et al.23 and Oishi et al.24 noted increased appe-
addictive drugs10–13, among others. tite in homozygous Clock-mutant mice but had contradictory findings
A topic that is not as well studied is the interaction between the regarding body weight and liver enzyme levels; these discrepancies
ghrelin system and circadian biology. This interaction is of great were attributed to differences in the strains of mice used by the two
importance because of the links between ghrelin and metabolism groups. Mutations in BMAL and CRY are also associated with metabolic
and between metabolism and circadian biology that are already abnormalities25, including impaired glucose and adipocyte regulation.
well established. Moreover, a large and growing population of indi- Thus, most, if not all, of the core clock genes are involved in metabolic
viduals are facing the synergistic health burdens of dysregulated regulation.

Nature Reviews Endocrinology


Review article

The suprachiasmatic nucleus (SCN) is the body’s master circadian


regulator. It is a bilateral region of the brain located ventromedially in GHSR
Brain Ghrelin
the anterior hypothalamus, rostral to the arcuate nucleus (ARC). The LEAP2
Pituitary gland
SCN regulates many metabolic processes including energy homeo-
stasis and insulin release26. Indeed, organisms with disrupted SCNs
develop dysregulated metabolism23. Bilateral electrolytic SCN lesion-
ing experiments show an increase in body weight, loss of circadian
rhythms of oxygen consumption and food intake, and development of
hepatic insulin resistance26. In addition, SCN-neuron-specific Bmal1-
knockout animals under constant darkness (DD) conditions display
loss of circadian rhythmicity in feeding, oxygen consumption and
thermoregulation, increased body weight attributable to increased
adiposity, and impaired glucose tolerance27,28. Behavioural and molecu-
lar circadian rhythms are restored following 1 week of time-restricted
feeding27. Furthermore, tracing studies indicate an anatomical connec-
tion between the ventromedial ARC (ARCvm) and the SCN29. These ana-
tomical connections might help relay peripheral metabolic information
Liver
to the SCN, although functional evidence is needed to support this
Stomach
hypothesis. In a later section of the Review, we consider the potential
Pancreas
involvement of ghrelin in these maladaptive changes.
Duodenum

Regulation and metabolic actions of ghrelin Jejunum


and LEAP2
As already mentioned, ghrelin is an agonist for GHSR, which is a mem-
ber of the seven-transmembrane G protein-coupled receptor family
(Fig. 2). In adults, ghrelin is primarily secreted from the stomach and Pancreatic islets
duodenum2,30. As noted, plasma ghrelin level increases upon fasting,
with stress and in settings of poor sleep, while it declines in obesity31–33.
Plasma ghrelin levels are also dynamically affected by feeding status,
rising preprandially and falling after a meal4,34. In the case of calorie
restriction and stress, the increase in plasma ghrelin level is thought to
occur from activation of the sympathetic nervous system and engage-
ment of β1-adrenergic receptors localized to ghrelin cells32,35–38. By con- Fig. 1 | Major sites of expression of the hormones ghrelin and LEAP2 and
trast, meal-driven and obesity-driven increases in insulin, the ensuing their receptor, GHSR. In adults, ghrelin (red) is secreted mainly by a distinct
increased engagement of ghrelin cell-expressed insulin receptors, and type of enteroendocrine cells (ghrelin cells) in the mucosa of the stomach and
in turn reduced ghrelin secretion, are mainly responsible for the des­ duodenum. Growth hormone secretagogue receptor (GHSR; blue) serves as the
cribed postprandial and obesity-related reductions in plasma ghrelin receptor for ghrelin. GHSR is expressed in several regions of the brain, including
level39. Also contributing to meal and/or obesity-associated ghre- the suprachiasmatic nucleus of the hypothalamus. GHSR is also expressed
lin reductions are lowered sensitivity of ghrelin cells to the usual highly in growth hormone-secreting somatotrophs within the pituitary, as
potent ghrelin secretagogue noradrenaline, activation of ghrelin well as in several endocrine cell types within pancreatic islets. Liver-expressed
cell-expressed fatty acid receptors, and/or changed numbers of ghre- antimicrobial peptide 2 (LEAP2; yellow), is another endogenous GHSR ligand.
In contrast to ghrelin (which is an endogenous GHSR agonist), LEAP2 is an
lin cells36,39,40. Regarding effects on metabolism, administering ghrelin
endogenous antagonist and inverse agonist of GHSR. LEAP2 is primarily
or ghrelin mimetics increases food intake, body weight gain, adiposity,
produced by the liver and the jejunal region of the small intestine.
hepatic steatosis and blood glucose level2,33,41–44, lowers energy expendi-
ture, upregulates expression of fat storage-promoting enzymes, and
leads to hedonic eating behaviours including operant responding
for food rewards, sucrose self-administration and cue-potentiated rewards, reduced binge-like high-fat diet intake and reductions in other
feeding2,45–50. By contrast, neutralizing ghrelin or treatment with GHSR hedonic ingestive behaviours48,61–63.
antagonists lowers body weight, food intake, various food reward LEAP2 represents another endogenous GHSR ligand (Fig. 2). In
behaviours and hepatic fat44,47,49,51–55. Although neither ghrelin nor GHSR contrast to ghrelin, which is a GHSR agonist, LEAP2 is a GHSR antagonist
loss-of-function (knockout) mice exhibit obvious phenotypes under and inverse agonist that potently blocks ghrelin action and decreases
normal conditions, clear phenotypes emerge once challenged: when constitutive GHSR activity; it is primarily secreted from the liver and
exposed to a high-fat diet early in life, these mice eat less, gain less intestine58,64. Although LEAP2 is not as well characterized as ghrelin,
weight and develop less adiposity than wild-type mice56,57. Exposing plasma LEAP2 is known to change inversely to ghrelin in many settings.
these mice to an acute-on-chronic calorie restriction regimen results For instance, in both mice and humans, LEAP2 falls during fasting, rises
in marked hypoglycaemia and increased mortality32,58,59. Following in obesity and rises after a meal64,65. In contrast to ghrelin, LEAP2 and/or
forced high-intensity interval exercise, these mice eat much less than LEAP2 analogues reduce food intake, binge eating and body weight, and
exercised wild-type mice60. Further, these loss-of-function models block ghrelin-induced food intake in mice63,66,67. LEAP2 also has been
show reduced stress-induced conditioned place preference for food shown to reduce ad libitum food intake and postprandial blood glucose

Nature Reviews Endocrinology


Review article

engages GHSR-expressing neurons in other brain regions including


Box 1 the hippocampus, caudal brainstem and olfactory bulb65,73,74. Vari-
ous hypothalamic nuclei express GHSR, including the ventromedial
hypothalamus (VMH), which is involved in feeding cessation, and the
Key circadian biology definitions ARC, which helps balance hunger and satiety signals. Previous research
extensively supports the direct interaction of ghrelin with these two
Circadian rhythm: fluctuation pattern of any behavioural, mediobasal hypothalamic nuclei2,75.
physiological or molecular process over the course of In the ARC, ghrelin stimulates AgRP (agouti-related peptide)/NPY
approximately 24 h. (neuropeptide Y) neurons. Selective GHSR expression in those neurons
DD: experimental condition exposing subjects to constant darkness enables ghrelin to induce food intake and prevents marked hypogly-
conditions. caemia upon caloric restriction7. Conversely, selective GHSR deletion
LD: experimental condition exposing subjects to 12-h light–12-h dark. from ARC AgRP neurons or ablation of ARC AgRP neurons abolishes the
Peripheral clocks: tissue-specific circadian clocks, influenced by acute orexigenic effects of ghrelin24,44. In the VMH, ghrelin suppresses
but independent from the central clock, which drive the circadian the activity of glucose-sensing neurons23, and inhibiting AMPK robustly
expression of a variety of genes involved in various physiological impairs the central orexigenic effect of ghrelin23. GHSR-expressing
processes115. neurons in the paraventricular nucleus of the hypothalamus, the lateral
Phase shift: a measurable change in the phase of a rhythm, hypothalamic nucleus and dorsomedial hypothalamus (DMH) help
observed by comparing a reference point in the phase (for example, mediate the actions of ghrelin on the neuroendocrine axis, food intake
onset of locomotor activity) with external time. Phase shifts can and food reward24–26. However, the effects of ghrelin on the SCN and
be either advances or delays; a phase advance means that the vice versa are not yet thoroughly characterized.
reference point occurs earlier than normal, whereas a phase delay As mentioned previously, the SCN is the master circadian clock
means it occurs later than normal116. and is of critical importance for metabolic regulation. Notably, ghrelin
Transcription–translation feedback loop (TTFL): first described secretion fluctuates in a circadian pattern, suggesting a role for the
by Bass and Takahashi, mammalian cells possess a cell SCN in regulating ghrelin secretion. Evidence also exists suggesting
autonomous circadian clock generated by a transcriptional that ghrelin has direct actions on the SCN, as GHSR mRNA is expressed
autoregulatory feedback loop. Transcriptional activators, CLOCK in the SCN41,70,71 (Fig. 3). Thus, there is reason to suspect an interaction
and BMAL1, act on their target genes, Period and Cryptochrome. between ghrelin and the circadian system.
These products progressively accumulate to form a repressor Could the ghrelin/GHSR system be a bridge between circadian
complex which, in turn, inhibits transcription of CLOCK and rhythms and metabolism? So far, indications exist towards this pos-
BMAL1 (ref. 117). sibility, but there are no definitive answers. The following two sections
Zeitgeber: external time cues, such as light, access to food and explore the relationship between the ghrelin system and circadian
environmental temperature, which synchronize the circadian rhythm in further detail.
system with external (that is, geophysical) time118.
Zeitgeber time (ZT): standard notation in the circadian field where Effects of ghrelin on circadian rhythm
ZT0 indicates the beginning of the light phase and ZT12 indicates Effects of ghrelin or GHSR on SCN neuronal activity
the beginning of the dark phase116. Ghrelin modulates the activity of neurons in the SCN. In one study by
Zhou et al., 5–6-week-old mice were individually housed and initially
entrained to a 2-week 12-h light–12-h dark (LD) cycle, and were then
placed under DD conditions76. Maximal neuronal activity responses
excursions in healthy men68. In lean people, postprandial increases in were observed when the mice were given a bolus injection of the ghrelin
LEAP2 correlate positively with postprandial decreases in appetite mimetic GHRP-6 at the beginning of the subjective night (CT12; which
ratings and with postprandial decreases in food cue brain reactivity is the same as the beginning of the active phase for mice). Specifically,
(measured using a food picture evaluation functional MRI task)69. LEAP2 GHRP-6 injected at CT12 gradually increased SCN neuronal firing fre-
deletion augments ghrelin-induced food intake and cFos induction in quency as measured by in vivo extracellular electrode recordings. Also,
both the ARC and the olfactory bulb65. Female LEAP2-knockout mice Ca2+ mobilization in SCN neurons, as determined by flow cytometry,
fed a high-fat diet have higher body weight than wild-type mice, due was increased twofold76. Although Zhou et al. switched the animals
to lower energy expenditure and locomotor activity and higher food from LD to DD after 2 weeks, another study by Tokizawa et al.77 kept
intake65. Thus, lowering plasma LEAP2 levels and lowering the plasma the mice housed under LD conditions and then administered ghrelin
LEAP2 to ghrelin ratio reduces the phenomenon of ghrelin resistance — intraperitoneally either 1 h after lights on or 1 h after lights off. Neuro­
or rather, it enhances sensitivity to the actions of endogenous ghrelin. nal activity was measured using immunohistochemistry for cFos.
As LEAP2 was only recently identified as a GHSR ligand, only limited In the group that received ghrelin during the light phase, the number
specific interactions it might have with circadian biology have so far of cFos-positive neurons in the SCN was significantly greater in the
been reported. ghrelin group than in the vehicle group. In addition, the number of cFos
and NPY double-labelled neurons in the SCN was greater in the ghrelin
Ghrelin action in the brain group than in the vehicle group77. No difference in neuronal activity
One of the main targets of ghrelin is the brain, which makes sense was observed between the vehicle and ghrelin groups when ghrelin was
given the high expression of GHSR in the brain and the many brain administered during the dark phase. The results from the two studies
regions expressing GHSR70–72. This Review highlights some specific are challenging to compare, since the mice were housed under dif-
roles of ghrelin within the hypothalamus, although ghrelin also directly ferent circadian conditions. Still, it seems that although both groups

Nature Reviews Endocrinology


Review article

showed that ghrelin increases neuronal activity in the SCN, the study by In addition to affecting neuronal activity in the SCN, ghrelin also
Zhou et al. showed this effect predominantly during the traditional impacts neuronal activity in the ARC, specifically the ARCvm. The
active phase for mice, while the study by Tokizawa et al. showed this ARCvm, as described previously, is thought to have a role in relaying
effect during the traditional resting phase for mice. peripheral metabolic signals to the SCN29. Ghrelin administration
In similar experiments, rats were housed under DD conditions induces greater cFos expression in the mouse ARC in the dark phase
and exposed to a light stimulus 1 h into their subjective night78. Neuro­ than in the light phase77. The majority of the cFos-positive ARC neurons
nal activity was again measured using immunohistochemistry for in the dark phase are also NPY-positive. The ARC AgRP/NPY neuronal
cFos. The photic stimulation resulted in an increase in cFos-positive population is perhaps best known for its effects on feeding. Func-
neurons in the SCN; however, pretreatment with GHRP-6 attenuated tionally, these features of the ARC are relevant because they provide
the light-induced cFos expression by 50%. These results are contrary mechanistic insight into circadian patterns of eating — these find-
to the results of Zhou et al.76, but also conflict with the investigators’ ings are consistent with a role for ghrelin in promoting food-seeking
own results when the same paradigm was applied to mice, suggesting behaviours during an organism’s active phase. Not only does ghrelin
that species differences might exist in the effects of ghrelin on SCN administration globally increase cFos expression in the ARC, it also
neuronal activity. specifically increases cFos expression in the ARCvm. Similarly, GHRP-6
The effects of ghrelin on SCN neuronal activity have also been activated AgRP neurons exclusively in the ARCvm in rats78. Altogether,
studied using an in vitro approach. Coronal sections of mouse brains these findings suggest that ghrelin has both direct and indirect effects
containing the SCN region were obtained from mice that had been on the SCN and, therefore, on circadian biology.
housed under LD conditions and killed during the light phase79. Ghrelin
was applied to the sections on the first day ex vivo, at what would have Effects of the ghrelin system on circadian patterns of
been the middle of the light phase for the mice. Spontaneous neuronal locomotor activity
activity was recorded on the second day ex vivo. Sections that had been Locomotor activity is tightly regulated by circadian rhythm and is an
exposed to ghrelin showed a 3-h advance in the rhythm of spontane- easily observable behavioural output. Ghrelin has numerous effects
ous firing when compared with the firing pattern of neurons in brain on circadian patterns of locomotor activity. When mice housed under
sections from control mice that had received artificial cerebrospinal DD conditions were given GHRP-6 at the start of their subjective night,
fluid. These effects were not observed when ghrelin was applied to a clear phase delay occurred in the rhythm of locomotor activity76.
the sections at what would have been 2 h into the dark phase for the This effect was abolished when mice were pretreated with a GHSR
mice. Taken together, the results of the studies highlighted in this antagonist76. By contrast, in mice housed under LD conditions, no
section suggest that the effects of ghrelin on SCN neuronal activity effects on locomotor activity rhythm were observed when the mice
vary depending on phase and housing conditions (LD versus DD). were given ghrelin at the start of their dark phase78. Surprisingly, light
Specifically, ghrelin has a maximal impact on SCN neuronal activity stimulation 1 h into the dark phase produced a phase delay in loco-
during the resting phase when rodents are housed under LD condi- motor activity that was reduced when the animals received ghrelin
tions and during the active phase when rodents are housed under pretreatment78. In a GHSR-knockout mouse model, when animals were
DD conditions. switched from LD to constant light (LL), knockout animals had longer

• Fasting • Obesity
• Stress • Meals
• Poor sleep

Sympathetic Insulin acting


nervous system via insulin
acting via β1ARs receptors on
on ghrelin cells ghrelin cells

LEAP2
Ghrelin The
circadian • Food intake
clock • Body weight
• Blood glucose
• Growth hormone secretion
• Locomotor activity;
food anticipatory activity
GHSR • Thermoregulation

Fig. 2 | Overview of the ghrelin system and its physiological functions. increases ghrelin levels. Fasting reduces LEAP2 levels. Obesity and meals
Growth hormone secretagogue receptor (GHSR) is a seven-transmembrane decrease ghrelin secretion, in large part via increasing insulin followed by
G protein-coupled receptor expressed by CNS neurons, pituitary somatotrophs engagement of insulin receptors expressed by ghrelin cells. By contrast,
and pancreatic islet endocrine cells. Ghrelin and liver-expressed antimicrobial obesity and meals increase LEAP2 levels. Ghrelin and LEAP2 regulate food
peptide 2 (LEAP2) represent two endogenous GHSR ligands — one that activates intake, body weight, blood glucose, growth hormone secretion, locomotor and
GHSR (ghrelin) and the other that blocks ghrelin-dependent and constitutive GHSR food anticipatory activity, and body temperature. Both ghrelin and LEAP2 are
activity (LEAP2). Fasting and stress stimulate ghrelin secretion, in large part influenced by the circadian clock and have effects on the previously mentioned
via activation of the sympathetic nervous system and engagement of outputs (for example, food intake and body weight) that are dependent on the
β1-adrenergic receptors (β1ARs) expressed by ghrelin cells. Poor sleep also circadian rhythm.

Nature Reviews Endocrinology


Review article

GHSR-IRES-Cre: YFP DAPI nuclear stain

LV
vhc

PaAP
Pav LH
3V
AHA MPA
BMA LA

opt
SCN

Fig. 3 | GHSR-expressing neurons in SCN of GHSR-IRES-Cre mice. ventricle; AHA, anterior hypothalamic area, anterior part; BMA, basomedial
Photomicrographs demonstrating YFP-immunoreactive cell bodies (green) amygdaloid nucleus, anterior part; Cre, Cre recombinase; DAPI, 4′,6-diamidino-
in a coronal section of the hypothalamus (located approximately −0.58 mm 2-phenylindole; f, fornix; GHSR, growth hormone secretagogue receptor; IRES,
from the bregma, at the level of the SCN) of a representative GHSR-IRES- internal ribosome entry site; LA, lateroanterior hypothalamic nucleus; LH, lateral
Cre × ROSA26-YFP reporter mouse. The presence of GHSR-positive neurons hypothalamic area; LV, lateral ventricle; MPA, medial preoptic area; opt, optic
within the SCN provides evidence suggesting a direct effect of the ghrelin tract; PaAP, paraventricular hypothalamic nucleus, anterior parvicellular part;
system on the SCN. The methods for visualizing the YFP reporter and acquiring Pav, paraventricular hypothalamic nucleus, ventral part; SCN, suprachiasmatic
images are the same as those described by Mani et al.7. Scale bar 100 µm. 3V, third nucleus; vhc, ventral hippocampal commissure; YFP, yellow fluorescent protein.

average periods of locomotor activity and also had delayed phases80. anticipatory activity (FAA) is defined as an increase in activity preced-
In addition, under LL conditions, GHSR-knockout animals displayed ing a scheduled meal. In rodents on a restricted meal schedule, FAA is
increased daily activity when compared with wild-type animals80. A note- usually observed beginning several hours prior to the presentation of
worthy discrepancy in the literature pertains to the effects of ghrelin the meal with a peak at mealtime82.
on locomotor activity in a food-deprived state: when Yanielli et al.79 Of the many circadian processes reviewed here, the roles of ghre-
administered a high dosage of GHRP-6 in the middle of the subjective lin and GHSR are perhaps the most studied in relation to FAA. Some
day after 24 h of food deprivation, they noted phase advances of loco- groups found that ghrelin produces FAA and that in the absence of
motor activity. Zhou et al.76 recapitulated this experiment with a slight ghrelin signalling, FAA is reduced. LeSauter et al.83 found that periph-
variation, but they did not replicate the previous findings. Further, eral administration of ghrelin in the middle of the light phase increased
antisense GHSR shRNA-mediated knockdown of GHSR expression in FAA and food intake. In addition, FAA induced by ghrelin administra-
the VMH reduces wheel running activity in both ad libitum-fed rats and tion was decreased in GHSR-knockout mice83. Blum et al.84 also found
rats subjected to a restricted feeding schedule, while also attenuating that FAA was attenuated in GHSR-knockout mice. The decrease in FAA
body weight loss otherwise induced by the wheel running activity81. was associated with decreased cFos expression levels in several hypo-
Interestingly, VMH GHSR knockdown in ad libitum-fed rats increases thalamic areas, including the DMH, but not the SCN. In schedule-fed
food intake and body weight gain81. goldfish under LL conditions, ghrelin antagonist treatment abolished
Less is known regarding the role of LEAP2-mediated effects on the FAA85. However, several groups found no difference in FAA86,87 and
circadian patterns of locomotor activity. However, a study published body temperature rise87 between GHSR-knockout mice and wild-type
in 2021 found that in high-fat diet-fed female mice housed in a stand- animals. Several other groups have also noted delays in the onset of
ard LD cycle, LEAP2-knockout mice had a 49% reduction in locomotor FAA. For instance, Lamont et al.80 noted that GHSR-knockout mice are
activity during the first 5 h of the dark cycle compared with wild-type less active and develop FAA more slowly than wild-type animals under
mice, without any differences in the first 5 h of the light cycle65. Thus, DD conditions, although these observations did not reach statistical
although ghrelin and GHSR have effects on the circadian rhythmicity significance. Rats with GHSR knockdown in the VMH exhibit a delay
of locomotor activity, and although the effects of LEAP2 on locomotor in the onset of FAA81. Therefore, despite these many studies linking
activity depend on circadian rhythmicity, the wide variation in experi- ghrelin and FAA, the effects of ghrelin on FAA are complex and require
mental procedures and a somewhat limited literature make it difficult further investigation.
to generate any consensus on the nature of the effects of LEAP2.
Effects of the ghrelin system on thermoregulation
Effects of the ghrelin system on food anticipatory activity One interaction that is less intuitive than the ones previously mentioned
Besides its effects on locomotor activity in general, ghrelin also impacts is the effects of ghrelin on circadian-dependent thermoregulation.
circadian-dependent food intake and food-seeking behaviours. Food Tokizawa et al.77 investigated this relationship in detail. Mice injected

Nature Reviews Endocrinology


Review article

with either vehicle or ghrelin at the start of the light phase (ZT0) had Influence of circadian rhythms on ghrelin and
an immediate increase in body temperature, probably partially as an GHSR expression
effect of animal handling. However, the increase in body tempera- Circadian rhythmicity of ghrelin in humans
ture was less in the ghrelin group than in the vehicle group. Over the Circadian rhythms affect the expression of ghrelin and GHSR and are
next 2 h, body temperature decreased in both groups, with greater relevant to humans. The human ghrelin gene contains an Ebox ele-
decreases in the ghrelin group. At ZT2 (2 h after the start of the light ment, a sequence of DNA that allows protein binding and regulates
phase), mice were either exposed to 10°C or stayed under 27°C condi- gene expression88. Ebox elements are strongly associated with circadian
tions. At both temperatures, no change in body temperature occurred rhythm89. Functionally, plasma ghrelin has been measured in humans
in the vehicle-treated animals. Body temperature of the ghrelin-treated over 24-h periods. In humans with access to three meals a day on a fixed
mice that remained under 27°C conditions increased until it was not schedule, ghrelin fluctuates across 24 h both as a function of mealtime
significantly different from the body temperature in the vehicle-treated as well as in a diurnal rhythm4 (Fig. 4). Plasma ghrelin levels rise approxi-
group. However, the ghrelin-treated group exposed to 10°C condi- mately twofold immediately before every meal and fall to trough levels
tions showed a significant decrease in body temperature compared 1 h after eating4. These preprandial and postprandial plasma ghrelin
with all the other groups. No differences were observed when injec- patterns are influenced by the types of nutrients within meals: both
tions of vehicle or ghrelin were given during the dark phase. Related carbohydrate-containing beverages and protein-containing bever-
to this finding, the increase in oxygen consumption in the vehicle ages suppress post-meal nadirs in plasma ghrelin more robustly than
group exposed to cold conditions was significantly greater than the lipid-containing beverages90. Over the course of the day, ghrelin levels
increase in oxygen consumption in the cold-exposed ghrelin group. exhibit a diurnal rhythm, reaching a peak at 0100 and falling overnight
Interestingly, in a study of a cohort of female mice fed a high-fat diet, to their lowest point at 0900 (ref. 4).
heat production was reduced (by 9.5%) in LEAP2-knockout mice com-
pared with wild-type mice during the first 5 h of the dark cycle65. No Peripheral effects of circadian rhythmicity of ghrelin
differences were observed between groups during the first 5 h of the The effects of circadian rhythms on ghrelin and GHSR have been studied
light cycle65. These findings suggest that both activating GHSRs with in further detail in animal models, both in the periphery and in the cen-
ghrelin and knocking out the endogenous GHSR antagonist, LEAP2, tral nervous system (CNS). In the periphery in mice, ghrelin-secreting
impair an organism’s ability to generate heat, resulting in hypothermia, cells of the stomach express the circadian clock proteins Per1 and Per283.
but only during the dark phase. Many unanswered questions remain Ghrelin expression is rhythmic, antiphase to Per1 and Per2 expression,
regarding the intersection of thermogenesis, circadian rhythm and and does not require light input83. In ad libitum-fed rats, Sánchez et al.3
the ghrelin system. found that ghrelin mRNA expression in the gastric mucosa remains
Therefore, although ghrelin clearly influences circadian-driven high throughout the light phase (Fig. 4). At the onset of the dark phase,
processes such as neuronal activity, locomotor activity, food antici- ghrelin mRNA expression in the gastric mucosa decreases and reaches
patory behaviour and thermoregulation, much debate remains in the its lowest point in the middle of the dark phase3. By contrast, plasma
field regarding the exact role of the ghrelin system. More targeted ghrelin levels reach a peak at the start of the dark phase, drop to a
experiments with standardized protocols for light–dark conditions nadir shortly after, then gradually increase for the remainder of the
and fed versus fasted states, should be conducted to better define the dark phase3. Notably, Bodosi et al.91 observed a slightly earlier peak in
effects of ghrelin on circadian rhythms. plasma ghrelin levels in ad libitum-fed mice than did Sánchez et al.3;

Active phase Inactive phase Scheduled Fig. 4 | Representative traces of human and
mealtimes rat plasma ghrelin secretion patterns. Ghrelin
levels in humans and rodents are influenced both
by circadian rhythm and anticipated mealtimes.
Human Humans are largely a diurnal species whereas
Daily plasma ghrelin
fluctuations with three rodents are nocturnal. However, patterns of plasma
• In both humans and rats with scheduled meals ghrelin levels are similar between humans and
three scheduled meals, plasma rodents when the active and inactive phases for
ghrelin levels peak at
each species are compared, suggesting that ghrelin
mealtimes.
• During the active phase, secretion patterns are more heavily dependent on
plasma ghrelin levels continue circadian time than external time. Ghrelin levels also
Rat
to rise between mealtimes.
Daily plasma ghrelin peak at scheduled mealtimes in both humans and
fluctuations with three rodents. The graphs are representations inspired by
scheduled meals
multiple sources.

• With ad libitum access to food,


plasma ghrelin levels reach a
peak at the start of the active Rat
phase, then immediately drop Daily plasma ghrelin
down to a nadir. fluctuations with ad libitum
• Subsequently, plasma ghrelin meal access
levels rise until the beginning
of the inactive state.

Nature Reviews Endocrinology


Review article

the plasma ghrelin peak in the study by Bodosi et al. occurred during the food to the light phase in rats resulted in the complete reversal of the
middle of the light phase as opposed to at the start of the dark phase diurnal rhythmicity of plasma ghrelin, with the peak plasma ghrelin
in the study by Sánchez et al.3. Circadian patterns of peripheral ghrelin levels occurring towards the end of the dark phase preceding food
levels are also sensitive to the effects of chronic stress92. Mice exposed to presentation91. In mice, restriction of feeding to the light phase led to
a chronic social defeat stress model of clinical depression had increased increased adiposity, weight gain, hyperleptinaemia and hypothermia96.
plasma ghrelin levels in the late light phase immediately preceding lights Dark phase-restricted feeding (DRF) resulted in an increase in plasma
off, as compared with controls that did not face the chronic stressor92. ghrelin levels prior to the start of the dark phase that was significantly
greater than the increase seen in mice with ad libitum food access97.
CNS effects of circadian rhythmicity of ghrelin DRF led to an increase in expression of GHSR and AgRP 1 h into the
CNS effects of ghrelin are observed in mice with ad libitum access to dark phase compared with ad libitum feeding97. DRF also enhanced
food: administration of GHRP-6, a ghrelin mimetic, downregulates GHSR the obesogenic properties of ghrelin: a low dose of ghrelin in mice
expression in the SCN, suggesting a circadian-related feedback loop for receiving DRF was sufficient to significantly increase fat preservation
ghrelin and GHSR76. Ghrelin protein expression is significantly reduced in these mice compared with their counterparts fed ad libitum97.
in Clock-mutant mice at all time points of the LD cycle23. The importance Several groups have also investigated food anticipatory increases
of ghrelin mRNA expression in the brain has been hotly debated, with in plasma ghrelin levels. Drazen et al.98 noted that when rats were
Cabral et al.93 concluding that “no irrefutable and reproducible evidence restricted to eating during a 4-h period, an increase in plasma ghrelin
exists supporting the notion that ghrelin is synthetized, at physiologi- levels occurred in the 2 h preceding the meal, peaking 30 min before the
cally relevant levels, in the central nervous system of adult mammals”; anticipated mealtime. This rise in plasma ghrelin in time-restricted feed-
however, one study showed that ghrelin mRNA levels in the mediobasal ing rats corresponded to an increase in plasma ghrelin in ad libitum-fed
hypothalamus, as detected by PCR with reverse transcription, are mark- rats immediately before the onset of the dark phase, when rats typi-
edly decreased in Clock-mutant mice compared with wild-type mice23. cally eat their largest meal98. Frecka and Mattes5 made similar obser-
Thus, both ghrelin protein and/or mRNA expression in the periphery vations in humans: they placed two groups of participants on feeding
and in the CNS seem to be under circadian control. regimens with either small intervals between meals or large intervals
between meals and found that in both groups, the time of peak plasma
Effect of time-restricted feeding on ghrelin secretion ghrelin shifted to prior to the anticipated mealtimes. Interestingly,
Numerous studies have shown promising results in the improvement mealtime-associated increases in plasma ghrelin and subsequently
of metabolic health in humans in response to time-restricted feeding FAA are sensitive to perceived food palatability: ad libitum-fed rats that
protocols94. Understanding the effects of time-restricted feeding on anticipated chocolate had greater increases in plasma ghrelin and FAA
ghrelin secretion is, therefore, of interest. Mimicking the traditional than chow-fed control animals99. Taken together, these findings suggest
pattern of three meals a day in humans, rats that were restricted to three that ghrelin may be involved in priming the body for effective meal
meals a day also showed increases in spontaneous activity and ghrelin consumption, perhaps as part of the arousal state described by Saper100.
levels associated with the scheduled meals95 (Fig. 4). In rats, this inter-
mittent fasting was also associated with adipogenesis95. Restriction of Relevance of the work in mouse models to human
health and disease
Some concern always exists regarding how well an animal model sys-
tem replicates a given disease process in humans, whether because of
Ghrelin inherent differences in baseline physiology or in disease manifestations
• Nighttime snacking
between the two species or because of variations in the mechanisms
• Night-eating syndrome
• Shift work involved in regulating the underlying physiological processes. As sci-
• Chronic jetlag entists, we do our best to choose models based on some aspects of
• CLOCK gene SNP
similarity to the process we are studying, with the understanding that
not all of our findings will be directly translatable to humans. In the
Metabolism Circadian
rhythms
field of ghrelin biology and metabolism overall, as well as in circadian
Diet-induced obesity and biology, animal models have been the cornerstone for furthering our
other metabolic disorders
understanding of basic human physiology. Wang et al.15 offer a detailed
review of the current widely used animal models in circadian research,
the limitations and advantages of each of these models, the similarities
and differences between mouse and human biology overall and specifi-
Fig. 5 | Intersections of ghrelin, metabolism and circadian rhythms. cally with regard to circadian biology, and the advances in human health
The ghrelin system (ghrelin, growth hormone secretagogue receptor (GHSR), that have been made possible as a result of these models. Wang et al.
liver-expressed antimicrobial peptide 2 (LEAP2) and ghrelin O-acyltransferase
conclude that “considering the cost, life cycle, ethical issues, etc., the
(GOAT), circadian system (central and peripheral) and metabolism are
advantages of mouse models outweigh the limitations, and the major-
interconnected and affect one another. While some aspects of these relationships
ity of our knowledge in this field has been conducted in mice”15. Also,
are well understood, much remains to be further explained. Regardless of
the mechanisms, what is known is that when any one of these chain links is
numerous similarities exist in the ghrelin systems between rodents
disrupted — for instance, by night-time snacking, night-eating syndrome, shift and humans. The predicted amino acid sequences for GHSR and ghre-
work, chronic jetlag or certain CLOCK gene single nucleotide polymorphisms lin in humans and in rodents show high sequence homology101, and
(SNPs) — serious consequences such as diet-induced obesity and other metabolic LEAP2 is identical in humans and in rodents58,101,102. The overall effects
disorders ensue. The severity and prevalence of these consequences make it of ghrelin on glucose and insulin levels are conserved between rodents
critical to understand how all three systems interact. and humans, as are its overall effects103 on growth hormone secretion,

Nature Reviews Endocrinology


Review article

wakefulness, food intake and weight gain79,103. Differences also exist.


For example, islet ghrelin production, which is predominant in the fetal
period, persists at a low level in adult humans whereas it disappears
Box 2
in adult mice104. Thus, although it is certainly important to consider
the limitations of animal models, we find no reason to believe that the Open research questions
data presented in this Review from studies in animal models will not
be relevant to humans. •• What would be the effects on phase shift if ghrelin was
systematically administered at multiple timepoints throughout
Conclusions the day, as opposed to general timeframes such as early
Given the strong evidence for GHSR in the brain (including the SCN morning or early night, or limited timepoints?
and ARC), the diurnal rhythmicity of plasma ghrelin levels, and the •• Are variations in endogenous levels of ghrelin, such as through
bidirectional interactions between ghrelin and the circadian system, time-restricted feeding or fasting, sufficient to produce changes
ghrelin seems to be a promising candidate to explain the link between in circadian rhythm?
metabolism and circadian biology (Fig. 5). Several clinically relevant •• Is circadian rhythmicity of ghrelin expression in gastric mucosa
scenarios exist, in addition to the ones described above, where the role cells a cell-intrinsic pattern or does it depend on external
of ghrelin is less well understood (Box 2). cues such as nutrient sensing or suprachiasmatic nucleus
For instance, Garaulet et al.105 genotyped 1,290 individuals with (SCN) input?
overweight and obesity for the CLOCK3111T/C single nucleotide poly- •• Does ghrelin acting on growth hormone secretagogue receptor
morphism (SNP). Of the individuals genotyped, 606 had either one or (GHSR) in the SCN have some role in mediating appropriate
two copies of the minor allele C, and 684 were non-carriers for the SNP. mealtimes and meal-seeking behaviour, potentially through the
Individuals who had either one or two copies of the minor allele C had generation of an arousal state at the start of an organism’s active
higher plasma ghrelin concentrations, lower sleep duration, delayed phase?
breakfast time, subjective preference for evening-time as opposed to •• Where do GHSR neurons in the SCN project to? Where do they
morning-time, and less compliance with a Mediterranean diet pattern. receive input from?
Mechanistically, how higher plasma ghrelin concentrations could •• What is the role for liver-enriched antimicrobial peptide 2 (LEAP2)
explain the observed behavioural findings or vice versa is unknown, in circadian regulation of neuronal and locomotor activity?
although poor sleep is associated with higher plasma ghrelin levels106. •• Is LEAP2 expression and secretion influenced by circadian
Disrupted circadian rhythms are increasingly becoming part of rhythms?
day-to-day life. One such lifestyle that frequently produces chrono- •• Is there some role for ghrelin in linking phenotypes of poor
disruption is chronic jetlag due to frequent time zone travelling. In a metabolic health with disrupted circadian biology, such as that
mouse model of chronic jetlag, jetlagged mice were housed for 4 days a seen in humans and animal models of chronic jetlag, night-time
week under an LD schedule, were shifted 8 h forward for the remaining snacking and mutations in the CLOCK gene?
3 days of the week, after which the jetlagged mice were shifted back
to the normal LD schedule, for 4 consecutive weeks107. In the mucosa of
the stomach, ghrelin mRNA expression fluctuated diurnally in control To settle some discrepancies and advance our understanding
mice, consistent with previously described findings. By contrast, the of the role of the ghrelin system in linking metabolism and circadian
jetlagged mice did not display this rhythm. The study also looked at biology, we suggest applying ghrelin/GHSR-specific tools. For exam-
GOAT, the enzyme that catalyses the acylation reaction required for ple, conditional Ghsr-knockout mouse models can help determine
ghrelin to bind to GHSR. Stomach mucosa expression of the Mboat4 whether the ghrelin system has effects on the circadian system and
gene that encodes GOAT also fluctuated diurnally in control animals, metabolism during development or during adulthood. In vivo calcium
although this pattern was not significantly different in jetlagged mice. imaging of GHSR neurons in the SCN can provide real-time insight into
In addition, plasma ghrelin levels were also rhythmic in both groups107. how neuronal activity is changing over the course of the day. Viral
More work is needed to understand why chronic jetlag affects only tracing methods can be employed to better understand and manipu-
ghrelin mRNA but not GOAT mRNA in the stomach mucosa and what late the ghrelin-engaged neuronal circuitry involved, and to better
the functional significance of this finding could be. understand the connections between the DMH or ARCvm and the SCN.
Ghrelin is not alone as a potential link between metabolism and cir- Chemogenetic and optogenetic strategies can be targeted specifically
cadian biology. Roles for several other hormones have been described, to GHSR-expressing neurons in the SCN. Additionally, we propose
although it is beyond the scope of this Review to discuss them in detail. investigating further the role of ghrelin, a potent growth hormone
As just a few examples, rats with dopaminergic neuron-specific SCN secretagogue, in influencing the well-documented circadian regu­
lesions exhibited signs of metabolic syndrome including increased lation of growth hormone1,110–114. Finally, all of these studies should be
weight gain, increased levels of insulin, leptin and noradrenaline, and done in both male and female animals, as sex differences have not been
altered lipid synthesis, and were hypertensive108. Fibroblast growth well-studied in this field. These strategies are already being used in sev-
factor 21 (FGF21), a liver molecule that signals starvation, mediates cir- eral fields to answer questions that were previously difficult to answer.
cadian rhythm via B-Klotho receptors located in the SCN; mice housed Given the clinical potential and strong societal need to find solutions
under LD conditions and fed a ketogenic diet showed increases in FGF21 for dysregulation of circadian and metabolic systems, it seems reason-
as well as decreases in overall locomotor activity and increases in able to pursue the question of the role of the ghrelin system in these
locomotor activity in the light phase109. Leptin displays circadian rhyth- processes with increased urgency and vigour.
micity opposite to that of ghrelin and also has time-of-day-dependent
effects on feeding and gene expression28. Published online: xx xx xxxx

Nature Reviews Endocrinology


Review article

References 35. Gupta, D. et al. β1-adrenergic receptors mediate plasma acyl-ghrelin elevation
1. Kojima, M. et al. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. and depressive-like behavior induced by chronic psychosocial stress.
Nature 402, 656–660 (1999). Neuropsychopharmacology 44, 1319–1327 (2019).
2. Müller, T. D. et al. Ghrelin. Mol. Metab. 4, 437–460 (2015). 36. Gong, Z. et al. G protein-coupled receptor 120 signaling regulates ghrelin secretion
3. Sánchez, J., Oliver, P., Picó, C. & Palou, A. Diurnal rhythms of leptin and ghrelin in in vivo and in vitro. Am. J. Physiol. Endocrinol. Metab. 306, E28–E35 (2014).
the systemic circulation and in the gastric mucosa are related to food intake in rats. 37. Koyama, H. et al. Comprehensive profiling of GPCR expression in ghrelin-producing
Pflügers Arch. 448, 500–506 (2004). cells. Endocrinology 157, 692–704 (2016).
4. Cummings, D. E. et al. A preprandial rise in plasma ghrelin levels suggests a role in meal 38. Uchida, A. et al. Altered ghrelin secretion in mice in response to diet-induced obesity
initiation in humans. Diabetes 50, 1714–1719 (2001). and Roux-en-Y gastric bypass. Mol. Metab. 3, 717–730 (2014).
5. Frecka, J. M. & Mattes, R. D. Possible entrainment of ghrelin to habitual meal patterns in 39. Shankar, K. et al. Ghrelin cell-expressed insulin receptors mediate meal- and
humans. Am. J. Physiol. Gastrointest. Liver Physiol. 294, G699–G707 (2008). obesity-induced declines in plasma ghrelin. JCI Insight 6, e146983 (2021).
Plasma ghrelin measurements in humans indicate that ghrelin fluctuates across 24-h 40. Engelstoft, M. S. et al. Seven transmembrane G protein-coupled receptor repertoire
both as a function of mealtime as well as in a diurnal rhythm. of gastric ghrelin cells. Mol. Metab. 2, 376–392 (2013).
6. Inui, A. Ghrelin: An orexigenic and somatotrophic signal from the stomach. Nat. Rev. 41. Mani, B. K. et al. The role of ghrelin-responsive mediobasal hypothalamic neurons in
Neurosci. 2, 551–560 (2001). mediating feeding responses to fasting. Mol. Metab. 6, 882–896 (2017).
7. Mani, B. K., Shankar, K. & Zigman, J. M. Ghrelin’s relationship to blood glucose. 42. Davies, J. S. et al. Ghrelin induces abdominal obesity via GHS-R-dependent lipid
Endocrinology 160, 1247–1261 (2019). retention. Mol. Endocrinol. 23, 914–924 (2009).
8. Sakata, I. et al. Glucose-mediated control of ghrelin release from primary cultures of 43. Dallak, M. A. Acylated ghrelin induces but deacylated ghrelin prevents hepatic
gastric mucosal cells. Am. J. Physiol. Endocrinol. Metab. 302, E1300–E1310 (2012). steatosis and insulin resistance in lean rats: effects on DAG/PKC/JNK pathway.
9. Shankar, K. et al. Acyl-ghrelin is permissive for the normal counterregulatory response to Biomed. Pharmacother. 105, 299–311 (2018).
insulin-induced hypoglycemia. Diabetes 69, 228–237 (2020). 44. Li, Z. et al. Ghrelin promotes hepatic lipogenesis by activation of mTOR-PPARγ signaling
10. Decarie-Spain, L. & Kanoski, S. E. Ghrelin and glucagon-like peptide-1: a gut-brain axis pathway. Proc. Natl Acad. Sci. USA 111, 13163–13168 (2014).
battle for food reward. Nutrients 13, 977 (2021). 45. Asakawa, A. et al. A role of ghrelin in neuroendocrine and behavioral responses to stress
11. Farokhnia, M., Faulkner, M. L., Piacentino, D., Lee, M. R. & Leggio, L. Ghrelin: from a gut in mice. Neuroendocrinology 74, 143–147 (2001).
hormone to a potential therapeutic target for alcohol use disorder. Physiol. Behav. 204, 46. Theander-Carrillo, C. et al. Ghrelin action in the brain controls adipocyte metabolism.
49–57 (2019). J. Clin. Invest. 116, 1983–1993 (2006).
12. Tian, J., Wang, T. & Du, H. Ghrelin system in Alzheimer’s disease. Curr. Opin. Neurobiol. 47. Perello, M. et al. Ghrelin increases the rewarding value of high-fat diet in an
78, 102655 (2023). orexin-dependent manner. Biol. Psychiatry 67, 880–886 (2010).
13. Diano, S. et al. Ghrelin controls hippocampal spine synapse density and memory 48. Perello, M. & Zigman, J. M. The role of ghrelin in reward-based eating. Biol. Psychiatry 72,
performance. Nat. Neurosci. 9, 381–388 (2006). 347–353 (2012).
14. Bahinipati, J., Sarangi, R., Pathak, M. & Mohapatra, S. Effect of night shift on development 49. Skibicka, K. P., Hansson, C., Egecioglu, E. & Dickson, S. L. Role of ghrelin in food reward:
of metabolic syndrome among health care workers. J. Fam. Med. Prim. Care 11, 1710–1715 impact of ghrelin on sucrose self-administration and mesolimbic dopamine and
(2022). acetylcholine receptor gene expression. Addict. Biol. 17, 95–107 (2012).
15. Wang, Y., Guo, H. & He, F. Circadian disruption: from mouse models to molecular 50. Kanoski, S. E., Fortin, S. M., Ricks, K. M. & Grill, H. J. Ghrelin signaling in the ventral
mechanisms and cancer therapeutic targets. Cancer Metastasis Rev. 42, 297–322 (2023). hippocampus stimulates learned and motivational aspects of feeding via PI3K-Akt
16. Hibi, M. et al. Nighttime snacking reduces whole body fat oxidation and increases LDL signaling. Biol. Psychiatry 73, 915–923 (2013).
cholesterol in healthy young women. Am. J. Physiol. Regul. Integr. Comp. Physiol. 304, 51. Barnett, B. P. et al. Glucose and weight control in mice with a designed ghrelin
R94–R101 (2013). O-acyltransferase inhibitor. Science 330, 1689–1692 (2010).
17. Vujović, N. et al. Late isocaloric eating increases hunger, decreases energy expenditure, 52. Esler, W. P. et al. Small-molecule ghrelin receptor antagonists improve glucose
and modifies metabolic pathways in adults with overweight and obesity. Cell Metab. 34, tolerance, suppress appetite, and promote weight loss. Endocrinology 148, 5175–5185
1486–1498.e7 (2022). (2007).
18. Birketvedt, G. S. et al. Behavioral and neuroendocrine characteristics of the night-eating 53. Shearman, L. P. et al. Ghrelin neutralization by a ribonucleic acid-SPM ameliorates
syndrome. JAMA 282, 657–663 (1999). obesity in diet-induced obese mice. Endocrinology 147, 1517–1526 (2006).
19. Goel, N. et al. Circadian rhythm profiles in women with night eating syndrome. J. Biol. 54. Zorrilla, E. P. et al. Vaccination against weight gain. Proc. Natl Acad. Sci. USA 103,
Rhythm. 24, 85–94 (2009). 13226–13231 (2006).
20. Chaput, J. P. et al. The role of insufficient sleep and circadian misalignment in obesity. 55. Abegg, K. et al. Ghrelin receptor inverse agonists as a novel therapeutic approach
Nat. Rev. Endocrinol. 19, 82–97 (2023). against obesity-related metabolic disease. Diabetes Obes. Metab. 19, 1740–1750 (2017).
21. Reinke, H. & Asher, G. Crosstalk between metabolism and circadian clocks. Nat. Rev. Mol. 56. Sun, Y., Wang, P., Zheng, H. & Smith, R. G. Ghrelin stimulation of growth hormone release
Cell Biol. 20, 227–241 (2019). and appetite is mediated through the growth hormone secretagogue receptor. Proc. Natl
22. Marcheva, B. et al. in Circadian Clocks (eds Kramer, A. & Merrow, M.) 127–155 [Series ed. Acad. Sci. USA 101, 4679–4684 (2004).
Michel, M. C. Handbook of Experimental Pharmacology Vol. 217] (Springer, 2013). 57. Zigman, J. M. et al. Mice lacking ghrelin receptors resist the development of diet-induced
23. Turek, F. W. et al. Obesity and metabolic syndrome in circadian Clock mutant mice. obesity. J. Clin. Invest. 115, 3564–3572 (2005).
Science 308, 1043–1045 (2005). 58. Ge, X. et al. LEAP2 is an endogenous antagonist of the ghrelin receptor. Cell Metab. 27,
24. Oishi, K. et al. Disrupted fat absorption attenuates obesity induced by a high-fat diet in 461–469.e6 (2018).
Clock mutant mice. FEBS Lett. 580, 127–130 (2006). 59. Yi, C.-X. et al. The GOAT-ghrelin system is not essential for hypoglycemia prevention
25. Marcheva, B., Ramsey, K. M., Affinati, A. & Bass, J. Clock genes and disease. J. Appl. Physiol. during prolonged calorie restriction. PLoS ONE 7, e32100 (2012).
107, 1638–1646 (2009). 60. Mani, B. K. et al. Ghrelin mediates exercise endurance and the feeding response
26. Coomans, C. P. et al. The suprachiasmatic nucleus controls circadian energy metabolism post-exercise. Mol. Metab. 9, 114–130 (2018).
and hepatic insulin sensitivity. Diabetes 62, 1102–1108 (2013). 61. Chuang, J. C. et al. Ghrelin mediates stress-induced food-reward behavior in mice.
27. Kolbe, I., Leinweber, B., Brandenburger, M. & Oster, H. Circadian clock network J. Clin. Invest. 121, 2684–2692 (2011).
desynchrony promotes weight gain and alters glucose homeostasis in mice. Mol. Metab. 62. Perello, M. & Dickson, S. L. Ghrelin signalling on food reward: a salient link between the
30, 140–151 (2019). gut and the mesolimbic system. J. Neuroendocrinol. 27, 424–434 (2015).
28. Cedernaes, J. et al. Transcriptional basis for rhythmic control of hunger and metabolism 63. Cornejo, M. P. et al. Growth hormone secretagogue receptor signalling affects high-fat
within the AgRP neuron. Cell Metab. 29, 1078–1091.e5 (2019). intake independently of plasma levels of ghrelin and LEAP2, in a 4-day binge eating
29. Yi, C.-X. et al. Ventromedial arcuate nucleus communicates peripheral metabolic model. J. Neuroendocrinol. 31, e12785 (2019).
information to the suprachiasmatic nucleus. Endocrinology 147, 283–294 (2006). 64. Mani, B. K. et al. LEAP2 changes with body mass and food intake in humans and mice.
30. Gupta, D. et al. “A LEAP 2 conclusions? Targeting the ghrelin system to treat obesity and J. Clin. Invest. 129, 3909 (2019).
diabetes”. Mol. Metab. 46, 101128 (2021). 65. Shankar, K. et al. LEAP2 deletion in mice enhances ghrelin’s actions as an orexigen and
31. Ariyasu, H. et al. Stomach is a major source of circulating ghrelin, and feeding state growth hormone secretagogue. Mol. Metab. 53, 101327 (2021).
determines plasma ghrelin-like immunoreactivity levels in humans. J. Clin. Endocrinol. 66. Islam, M. N. et al. Liver-expressed antimicrobial peptide 2 antagonizes the effect
Metab. 86, 4753–4758 (2001). of ghrelin in rodents. J. Endocrinol. 244, 13–23 (2020).
32. Mani, B. K., Osborne-Lawrence, S., Vijayaraghavan, P., Hepler, C. & Zigman, J. M. 67. Wald, H. S., Ghidewon, M. Y., Hayes, M. R. & Grill, H. J. Hindbrain ghrelin and
β1-Adrenergic receptor deficiency in ghrelin-expressing cells causes hypoglycemia in liver-expressed antimicrobial peptide 2, ligands for growth hormone secretagogue
susceptible individuals. J. Clin. Invest. 126, 3467–3478 (2016). receptor, bidirectionally control food intake. Am. J. Physiol. Regul. Integr. Comp. Physiol.
33. Tschöp, M., Smiley, D. L. & Heiman, M. L. Ghrelin induces adiposity in rodents. Nature 324, R547–R555 (2023).
407, 908–913 (2000). 68. Hagemann, C. A. et al. LEAP2 reduces postprandial glucose excursions and ad libitum
34. Liu, J. et al. Novel ghrelin assays provide evidence for independent regulation of ghrelin food intake in healthy men. Cell Rep. Med. 3, 100582 (2022).
acylation and secretion in healthy young men. J. Clin. Endocrinol. Metab. 93, 1980–1987 69. Bhargava, R. et al. Postprandial increases in liver-gut hormone LEAP2 correlate with
(2008). attenuated eating behavior in adults without obesity. J. Endocr. Soc. 7, bvad061 (2023).

Nature Reviews Endocrinology


Review article

70. Guan, X.-M. et al. Distribution of mRNA encoding the growth hormone secretagogue 98. Drazen, D. L., Vahl, T. P., D’Alessio, D. A., Seeley, R. J. & Woods, S. C. Effects of a fixed meal
receptor in brain and peripheral tissues. Mol. Brain Res. 48, 23–29 (1997). pattern on ghrelin secretion: evidence for a learned response independent of nutrient
71. Zigman, J. M., Jones, J. E., Lee, C. E., Saper, C. B. & Elmquist, J. K. Expression of ghrelin status. Endocrinology 147, 23–30 (2006).
receptor mRNA in the rat and the mouse brain. J. Comp. Neurol. 494, 528–548 (2006). 99. Merkestein, M. et al. Ghrelin mediates anticipation to a palatable meal in rats. Obesity 20,
72. Ferrini, F., Salio, C., Lossi, L. & Merighi, A. Ghrelin in central neurons. Curr. 963–971 (2012).
Neuropharmacol. 7, 37–49 (2009). 100. Saper, C. B. in Hypothalamic Integration of Energy Metabolism (eds Kalsbeek et al.)
73. Kanoski, S. E. & Grill, H. J. Hippocampus contributions to food intake control: mnemonic, 243–252 (Elsevier, 2006). [Series Progress in Brain Research Vol. 153].
neuroanatomical, and endocrine mechanisms. Biol. Psychiatry 81, 748–756 (2017). 101. Palyha, O. C. et al. Ligand activation domain of human orphan growth hormone (GH)
74. Faulconbridge, L. F., Cummings, D. E., Kaplan, J. M. & Grill, H. J. Hyperphagic effects of secretagogue receptor (GHS-R) conserved from pufferfish to humans. Mol. Endocrinol.
brainstem ghrelin administration. Diabetes 52, 2260–2265 (2003). 14, 160–169 (2000).
75. Andrews, Z. B. The extra-hypothalamic actions of ghrelin on neuronal function. 102. Kojima, M. & Kangawa, K. Ghrelin: structure and function. Physiol. Rev. 85, 495–522 (2005).
Trends Neurosci. 34, 31–40 (2011). 103. Denney, W. S., Sonnenberg, G. E., Carvajal-Gonzalez, S., Tuthill, T. & Jackson, V. M.
76. Zhou, L. et al. Activation of growth hormone secretagogue receptor induces Pharmacokinetics and pharmacodynamics of PF-05190457: the first oral ghrelin receptor
time-dependent clock phase delay in mice. Am. J. Physiol. Endocrinol. Metab. 307, inverse agonist to be profiled in healthy subjects. Br. J. Clin. Pharmacol. 83, 326–338
E515–E526 (2014). (2017).
Bolus injection of ghrelin mimetic at CT12 produces phase delay under DD; this effect 104. Wierup, N., Sundler, F. & Heller, R. S. The islet ghrelin cell. J. Mol. Endocrinol. 52, R35–R49
is abolished by pretreatment with a GHSR agonist. (2014).
77. Tokizawa, K., Onoue, Y., Uchida, Y. & Nagashima, K. Ghrelin induces time-dependent 105. Garaulet, M. et al. Ghrelin, sleep reduction and evening preference: relationships to
modulation of thermoregulation in the cold. Chronobiol. Int. 29, 736–746 (2012). CLOCK 3111 T/C SNP and weight loss. PLoS ONE 6, e17435 (2011).
Wild-type animals exposed to ghrelin during the early light phase display diminished Individuals with overweight and obesity and a specific CLOCK single nucleotide
ability to protect their body temperature following cold exposure; this effect is polymorphism had higher plasma ghrelin concentrations.
abolished when ghrelin is instead administered during the early dark phase. 106. Spiegel, K., Tasali, E., Penev, P. & Van Cauter, E. Brief communication: sleep curtailment
78. Yi, C. X. et al. A circulating ghrelin mimetic attenuates light-induced phase delay of mice in healthy young men is associated with decreased leptin levels, elevated ghrelin levels,
and light-induced Fos expression in the suprachiasmatic nucleus of rats. Eur. J. Neurosci. and increased hunger and appetite. Ann. Intern. Med. 141, 846–850 (2004).
27, 1965–1972 (2008). 107. Desmet, L., Thijs, T., Segers, A., Verbeke, K. & Depoortere, I. Chronodisruption by chronic
79. Yannielli, P. C., Molyneux, P. C., Harrington, M. E. & Golombek, D. A. Ghrelin effects on the jetlag impacts metabolic and gastrointestinal homeostasis in male mice. Acta Physiol.
circadian system of mice. J. Neurosci. 27, 2890–2895 (2007). 233, e13703 (2021).
Slice physiology of the SCN shows a 3-h advance in the rhythm of spontaneous Mouse model of chronic jetlag displays disturbed circadian rhythmicity of ghrelin
neuronal firing when sections are treated with ghrelin at ZT6. mRNA expression in stomach mucosa.
80. Lamont, E. W., Bruton, J., Blum, I. D. & Abizaid, A. Ghrelin receptor-knockout mice 108. Luo, S. et al. Experimental dopaminergic neuron lesion at the area of the biological
display alterations in circadian rhythms of activity and feeding under constant lighting clock pacemaker, suprachiasmatic nuclei (SCN) induces metabolic syndrome in rats.
conditions. Eur. J. Neurosci. 39, 207–217 (2014). Diabetol. Metab. Syndr. 13, 11 (2021).
81. Merkestein, M. et al. GHS-R1a signaling in the DMH and VMH contributes to food 109. Bookout, A. L. et al. FGF21 regulates metabolism and circadian behavior by acting on the
anticipatory activity. Int. J. Obes. 38, 610–618 (2014). nervous system. Nat. Med. 19, 1147–1152 (2013).
82. Mistlberger, R. E. Neurobiology of food anticipatory circadian rhythms. Physiol. Behav. 110. Gupta, D. et al. Disrupting the ghrelin-growth hormone axis limits ghrelin’s orexigenic but
104, 535–545 (2011). not glucoregulatory actions. Mol. Metab. 53, 101258 (2021).
83. LeSauter, J., Hoque, N., Weintraub, M., Pfaff, D. W. & Silver, R. Stomach ghrelin-secreting 111. Nass, R. et al. Evidence for acyl-ghrelin modulation of growth hormone release in the fed
cells as food-entrainable circadian clocks. Proc. Natl Acad. Sci. USA 106, 13582–13587 state. J. Clin. Endocrinol. Metab. 93, 1988–1994 (2008).
(2009). 112. Zizzari, P., Hassouna, R., Grouselle, D., Epelbaum, J. & Tolle, V. Physiological roles of
Food anticipatory activity is diminished in GHSR-knockout mice, and in preproghrelin-derived peptides in GH secretion and feeding. Peptides 32, 2274–2282 (2011).
ghrelin-containing oxyntic cells, ghrelin express is rhythmic, anti-phase to clock 113. Serin, Y. & Acar Tek, N. Effect of circadian rhythm on metabolic processes and the
proteins, Per1 and Per2, and does not require light input. regulation of energy balance. Ann. Nutr. Metab. 74, 322–330 (2019).
84. Blum, I. D. et al. Reduced anticipatory locomotor responses to scheduled meals in 114. Wang, W. et al. Rotating day and night disturb growth hormone secretion profiles, body
ghrelin receptor deficient mice. Neuroscience 164, 351–359 (2009). energy metabolism, and insulin levels in mice. Neuroendocrinology 112, 481–492 (2022).
85. Nisembaum, L. G., de Pedro, N., Delgado, M. J. & Isorna, E. Crosstalking between the 115. Richards, J. & Gumz, M. L. Advances in understanding the peripheral circadian clocks.
“gut-brain” hormone ghrelin and the circadian system in the goldfish. Effects on clock FASEB J. 26, 3602–3613 (2012).
gene expression and food anticipatory activity. Gen. Comp. Endocrinol. 205, 287–295 116. Vitaterna, M. H., Takahashi, J. S. & Turek, F. W. Overview of circadian rhythms. Alcohol.
(2014). Res. Health 25, 85–93 (2001).
86. Gunapala, K. M., Gallardo, C. M., Hsu, C. T. & Steele, A. D. Single gene deletions of orexin, 117. Bass, J. & Takahashi, J. S. Circadian integration of metabolism and energetics. Science
leptin, neuropeptide Y, and ghrelin do not appreciably alter food anticipatory activity in 330, 1349–1354 (2010).
mice. PLoS ONE 6, e18377 (2011). 118. Heyde, I. & Oster, H. Induction of internal circadian desynchrony by misaligning
87. Gooley, J. J., Schomer, A. & Saper, C. B. The dorsomedial hypothalamic nucleus is critical zeitgebers. Sci. Rep. 12, 1601 (2022).
for the expression of food-entrainable circadian rhythms. Nat. Neurosci. 9, 398–407
(2006). Acknowledgements
88. Kanamoto, N. et al. Genomic structure and characterization of the 5′-flanking region The authors’ works is supported through research grants from the NIH (R01 DK103884,
of the human ghrelin gene. Endocrinology 145, 4144–4153 (2004). P01DK119130 and 5T32GM008014).
89. Herzog, E. D. Neurons and networks in daily rhythms. Nat. Rev. Neurosci. 8, 790–802
(2007). Author contributions
90. Foster-Schubert, K. E. et al. Acyl and total ghrelin are suppressed strongly by ingested All authors researched data for the article. S.S.K. and J.M.Z. contributed to the discussion of
proteins, weakly by lipids, and biphasically by carbohydrates. J. Clin. Endocrinol. Metab. content, wrote the article and reviewed/edited it before submission.
93, 1971–1979 (2008).
91. Bodosi, B. et al. Rhythms of ghrelin, leptin, and sleep in rats: effects of the normal diurnal Competing interests
cycle, restricted feeding, and sleep deprivation. Am. J. Physiol. Regul. Integr. Comp. J.M.Z. owns stock in Medtronic, Eli Lilly and Novo Nordisk, and received research funding from
Physiol. 287, R1071–R1079 (2004). Novo Nordisk for another project during the writing of this Review. The other authors declare
Time-restricted feeding of rats to just the light phase shifts ghrelin peak from the light no competing interests.
phase to the end of the dark phase.
92. Kumar, J. et al. Differential effects of chronic social stress and fluoxetine on meal patterns Additional information
in mice. Appetite 64, 81–88 (2013). Peer review information Nature Reviews Endocrinology thanks Yu Zhou and the other,
93. Cabral, A., López Soto, E. J., Epelbaum, J. & Perelló, M. Is ghrelin synthesized in the anonymous, reviewers for their contribution to the peer review of this work.
central nervous system? Int. J. Mol. Sci. 18, 638 (2017).
94. Manoogian, E. N. C. & Panda, S. Circadian rhythms, time-restricted feeding, and healthy Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
aging. Ageing Res. Rev. 39, 59–67 (2017). published maps and institutional affiliations.
95. Verbaeys, I. et al. Scheduled feeding results in adipogenesis and increased acylated
ghrelin. Am. J. Physiol. Endocrinol. Metab. 300, E1103–E1111 (2011). Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this
96. Oishi, K. & Hashimoto, C. Short-term time-restricted feeding during the resting phase article under a publishing agreement with the author(s) or other rightsholder(s); author
is sufficient to induce leptin resistance that contributes to development of obesity and self-archiving of the accepted manuscript version of this article is solely governed by the
metabolic disorders in mice. Chronobiol. Int. 35, 1576–1594 (2018). terms of such publishing agreement and applicable law.
97. Sorrell, J. et al. The central melanocortin system mediates the benefits of time-restricted
feeding on energy balance. Physiol. Behav. 227, 113132 (2020). © Springer Nature Limited 2023

Nature Reviews Endocrinology

You might also like