You are on page 1of 26

The circadian clock in white and brown adipose tissue: mechanistic,

endocrine and clinical aspects

Oren Froy, Marta Garaulet


ADVANCE ARTICLE: Endocrine Reviews

Endocrine Reviews
Endocrine Society

Submitted: July 24, 2017


Accepted: February 22, 2018
First Online: February 27, 2018

Advance Articles are PDF versions of manuscripts that have been peer reviewed and accepted but
not yet copyedited. The manuscripts are published online as soon as possible after acceptance and
before the copyedited, typeset articles are published. They are posted "as is" (i.e., as submitted by
the authors at the modification stage), and do not reflect editorial changes. No
corrections/changes to the PDF manuscripts are accepted. Accordingly, there likely will be
differences between the Advance Article manuscripts and the final, typeset articles. The
manuscripts remain listed on the Advance Article page until the final, typeset articles are posted.
At that point, the manuscripts are removed from the Advance Article page.

DISCLAIMER: These manuscripts are provided "as is" without warranty of any kind, either express
or particular purpose, or non-infringement. Changes will be made to these manuscripts before
publication. Review and/or use or reliance on these materials is at the discretion and risk of the
reader/user. In no event shall the Endocrine Society be liable for damages of any kind arising
references to, products or publications do not imply endorsement of that product or publication.

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

The circadian clock in adipose tissue

The circadian clock in white and brown adipose tissue: mechanistic,


endocrine and clinical aspects
Oren Froy1, Marta Garaulet2
1
Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and
Environment, The Hebrew University of Jerusalem, Rehovot, Israel.
2
Department of Physiology, University of Murcia, Murcia Spain; IMIB-Arrixaca, Murcia, Spain
Received 24 July 2017. Accepted 22 February 2018.

LE
Obesity is a major risk factor for the development of illnesses, such as insulin resistance and
hypertension and has become a serious public health problem. Mammals have developed a
circadian clock located in the hypothalamic suprachiasmatic nuclei (SCN) that responds to
ADVANCE ARTICLE: Endocrine Reviews

the environmental light-dark cycle. Clocks similar to the one located in the SCN are found in

IC
peripheral tissues, such as the kidney, liver and adipose tissue. The circadian clock regulates
metabolism and energy homeostasis in peripheral tissues by mediating activity and/or
expression of key metabolic enzymes and transport systems. Knockouts or mutations in clock
genes that lead to disruption of cellular rhythmicity have provided evidence to the tight link

T
between the circadian clock and metabolism. In addition, key proteins play a dual role in
regulating the core clock mechanism as well as adipose tissue metabolism and link circadian

R
rhythms with lipogenesis and lipolysis. Adipose tissues are distinguished as white, brown and
beige (or brite), each with unique metabolic characteristics. Recently, the role of the circadian
A
clock in regulating the differentiation into the different adipose tissues has been investigated.
In this review, the role of clock proteins and the downstream signaling pathways in white,
brown and brite adipose tissue function and differentiation will be reviewed. In addition,
E

chronodisruption and metabolic disorders and clinical aspects of circadian adiposity will be
addressed.
C

The role of the circadian clock in regulating adipose tissue function and differentiation and key
proteins that play a dual role in regulating clock mechanism and adipose tissue metabolism.
N

Essential Points:
• The circadian clock controls energy homeostasis by regulating circadian expression and/or
A

activity of enzymes, hormones, and transport systems involved in metabolism.


• Disruption of circadian rhythms leads to obesity and metabolic disorders.
V

• The circadian clock regulates the function of and differentiation into white and brown adipose
tissue.
D

• Key proteins play a dual role in regulating the core clock mechanism as well as adipose tissue
metabolism and link circadian rhythms with lipogenesis, lipolysis and differentiation.
A

1. Circadian rhythms and the circadian clock


Organisms on our planet developed an endogenous circadian clock entrained, or
synchronized, by light to exactly 24 hours (1). By predicting the day-night cycles, the clock
imparts a survival advantage as physiological processes are performed at the appropriate time
to find food, mating partners or to avoid predators (1-3). The clock is self-sustained, but in
the absence of light, the endogenous clock free-runs, generating cycles of either slightly
longer or slightly shorter than 24 hours, hence the term circadian (circadian, circa=about
dies=day). The circadian clock controls numerous cellular, physiological, metabolic,
endocrine and behavioral systems (4,5). Disruption of this control results in fatigue,
disorientation, insomnia, altered hormone profile and high morbidity, seriously influencing

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 1
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

overall health (4,6-12). Obesity, which is characterized by the excess of fat accumulation in
white adipose tissue, has been related to irregular sleep/wake schedules, high snacking
frequency, or social jet lag known to disrupt the circadian clock (13,14). Bright light at night
has also been associated with obesity, as was shown in 100,000 women of the Breakthrough
Generations Study (15). All these studies as a whole, strongly suggest that impairment of the
circadian system is involved in the etiology of several illnesses (16).
The central circadian clock is located in the anterior hypothalamus and is confined to the
suprachiasmatic nuclei (SCN). Photic information perceived by the retina and transmitted via
the retinohypothalamic tract (RHT) synchronizes SCN neurons to coordinate circadian
outputs (17-20). Clocks, similar to those found in SCN neurons, are found in peripheral
tissues, such as the liver, intestine, heart, adipose tissue, retina and in various brain regions

LE
(2,21-23). 5-20% of tissue-specific genes are estimated to exhibit an oscillatory expression
profile, emphasizing the circadian control over the function of peripheral tissues (23-32). The
SCN clock regulates peripheral rhythms by neuronal connections, secretion of humoral
ADVANCE ARTICLE: Endocrine Reviews

factors (33-35), or indirectly by driving rhythmic feeding, locomotor activity and/or body

IC
temperature, which, in turn, coordinate rhythmic gene expression.
The molecular clock in SCN neurons and peripheral cells is an intracellular mechanism
composed of transcription-translation feedback loops (3). The transcription factor CLOCK

T
(Circadian Locomotor Output Cycles Kaput) dimerizes with BMAL1 (brain and muscle
ARNT-like protein 1) and together the CLOCK:BMAL1 heterodimer activates transcription
by binding to E-box (5’- CACGTG -3’) and E-box-like promoter sequences (2). Amongst the

R
regulatory targets of CLOCK:BMAL1 are the Period (Per1, Per2) and Cryptochrome (Cry1
and Cry2) genes. Oligomerization and nuclear translocation of PERs:CRYs leads to the
A
inhibition of CLOCK:BMAL1-mediated transcription (2,36) (Fig. 1). The nuclear receptor
reverse ERBα (REV-ERBα) negatively regulates Bmal1 expression (37), whereas retinoic
acid receptor-related orphan receptor α (RORα) and RORγ (38) (Fig. 1) positively
E

regulates its expression via ROR response elements (RORE) (39). All clock genes exhibit
24-h oscillation in peripheral tissues (40).
C

2. White, brown and beige (or brite) adipose tissue


N

Classically, adipose tissue has been regarded as a passive reservoir for energy storage, but
this traditional point of view is no longer valid. In 1987, adipose tissue was identified as the
major site for metabolism of sex steroids (41). Several years later, in 1994, the discovery of
A

the cytokine-like factor leptin (42) entirely changed our perspectives on adipose tissue. This
was a revolution in the study of obesity, as for the first time it was shown that adipose tissue
V

was able to secrete hormones or “adipocytokines” capable of communicating information


from periphery to the central nervous system. Adipose tissue was no longer considered a
D

passive reservoir of energy; it was an endocrine organ. Adipocytokines or adipokines, such as


adiponectin, resistin, tumor necrosis factor α (TNFα), visfatin, obestatin have autoendocrine,
A

paracrine and endocrine functions. Thus, adipose tissue regulates energy storage and
expenditure and influences systemic metabolic homeostasis through production of adipokines
(43).
In mammals, at least two types of adipose tissue have been described, white adipose
tissue (WAT) and the brown adipose tissue (BAT). WAT and BAT originate from different
stem cells and they have different distribution, gene expression, and function (44) (Fig. 2).
Adipocytes with features similar to BAT found within white adipose depots are termed beige
or brite (brown in white) adipose tissue (44). Nowadays, it is known that the development of
obesity depends not only on the balance between food intake and energy expenditure but also
on the balance between WAT, BAT and brite adipose tissue.

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 2
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

WAT:
The most abundant type of adipose tissue in mammals and the main energy reservoir, stores
dietary energy as triacylglycerides (TAGs) in unilocular lipid droplets and secretes a huge
number of hormones and cytokines that regulate metabolism and insulin resistance (43,45).
After meals, WAT uptakes dietary fats and carbohydrates from the circulation and, through
lipogenesis, converts them to TAGs (46). During fasting or physical activity, lipolysis ensues
and TAGs are broken down into free fatty acids (FFAs) and glycerol that are released to the
circulation to supply the needs of other tissues (46).
BAT:
This tissue contains multilocular lipid droplets and high mitochondrial density and is able to
convert chemical energy into heat (47). Cold stimulation leads to the release of

LE
norepinephrine at innervated sites within the BAT, which leads to lipolysis of TAGs and
increase in cellular FFAs. FFAs are subsequently transported to the mitochondria and serve
as substrates for β-oxidation. Heat is generated using uncoupling protein-1 (UCP1), a protein
ADVANCE ARTICLE: Endocrine Reviews

that is localized on the inner membrane of mitochondria and uncouples electron transport

IC
from ATP production (47). The resulting energy derived from substrate oxidation is
dissipated as heat. In addition to its key role in maintaining body temperature under cold-
stress, the metabolic function of BAT is important for global energy balance, insulin

T
sensitivity and lipid metabolism (48-50). Developmentally, BAT is typified by the activation
of myogenic factor 5 (Myf5) and early paired box 7 (Pax7) expression (51,52), which encode

R
two transcription factors that mark myogenic precursor cells. Late Pax7 expression marks
cells that are predominantly restricted to the skeletal muscle lineage (52,53).
A
Brite (beige) adipose tissue:
Unlike WAT, which is characterized by the presence of few mitochondria that are devoid of
UCP1, beige adipocytes found within WAT express UCP1 and are mitochondrial-rich. These
E

adipocytes are distinct from BAT, but still have a thermogenic capacity by responding to cold
exposure to dissipate heat through UCP1 (54).
C

3. The circadian clock and metabolism


The robust and coordinated expression of clock genes in peripheral tissues, such as liver and
N

adipose tissue, mediates the activity of nuclear receptors, enzymes, hormones and
transporters involved in carbohydrate, lipid and protein metabolism (21,30,55-61). In turn,
A

animal studies have shown that a high-fat diet may contribute to the development of obesity
and insulin resistance via alterations in the circadian period of locomotor activity rhythms
V

and changes in the oscillation of clock genes in adipose and liver tissue (62-65). In humans,
results suggest that the modulation of the dietary fat and carbohydrate content alters the
function of the central and peripheral circadian clocks in humans (66). The mutual influence
D

of the clock and metabolic regulation is achieved due to the participation of key catabolic,
anabolic and lipid metabolic factors in the core clock mechanism (Fig. 1).
A

REV-ERBs and retinoic acid–related orphan receptors (RORs):


Two important families that link the core clock mechanism with lipid metabolism are REV-
ERBs and RORs. These factors are vital for adipocyte differentiation (67), lipogenesis and
lipid storage and exhibit striking circadian rhythms (68,69). In addition to their metabolic
role, REV-ERBs are the negative and RORs the positive regulators of Bmal1 expression
(37,38,70,71) (Fig. 1). In turn, the CLOCK:BMAL1 heterodimer regulates the
expression of Rev-erbs and Rors (37,38,72). Diet-induced obese mice treated with a REV-
ERB agonist showed reduced fat mass and improved dyslipidaemia and hyperglycaemia (73).
In addition, a REV-ERB agonist suppressed mouse orexinergic gene expression, whereas
REV-ERBβ-deficient mice had increased orexinergic transcripts (74). In this line, mice

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 3
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

deficient for Rev-erbα develop higher adiposity on regular chow and high-fat diet supposedly
due to increased fat uptake by adipose tissue (75). Nevertheless, in BAT, Rev-erbα seems to
have an opposite role on adiposity since deletion of Rev-erbα markedly improved the
thermogenic response to cold and physiological induction of UCP1 by cold temperatures is
preceded by rapid downregulation of Rev-erbα (76). As REV-ERBs negatively regulates
Bmal1 expression, the role of BMAL1 in WAT and BAT function merits further study.
Peroxisome proliferator-activated receptors (PPARs):
PPARs are a nuclear receptor family consisting of 3 isoforms in mammals, α, β/δ, and γ,
which are differentially expressed among tissues (77). PPARs plays a key role in the
transcription of genes involved in lipid and glucose metabolism upon binding of endogenous
free fatty acids (78,79). PPARα is abundant in the liver, BAT, heart and kidney, while

LE
PPARγ in expressed in the adipose tissue and PPARβ/δ throughout the body (80).
PPARα agonists, such as fibrates, are clinically proven lipid-lowering drugs, whereas
PPARγ ligands such as thiazolidinediones (TZDs), improve glycemic control via insulin
ADVANCE ARTICLE: Endocrine Reviews

sensitization in patients with type 2 diabetes (81). PPARs are connected to the core clock

IC
mechanism, as their expression is mediated by the CLOCK:BMAL heterodimer. In turn,
PPARα activates Bmal1 expression (Fig. 1) by binding to the peroxisome-proliferator
response element (PPRE) (82-84). The role of PPARs in the circadian regulation has been

T
demonstrated through experiments performed with PPARα agonists, that showed advanced
locomotor activity and feeding daily rhythms in mice (85), whereas experiments in PPARγ

R
deletion showed dampened behavioral and cellular circadian rhythms (86).
PPARγ coactivator 1 (PGC1):
A
The PPARγ coactivator 1 (PGC1) is a family of transcriptional coactivators that induce
mitochondrial oxidative metabolism. Indeed, PGC-1α is a catabolic factor, whose expression
is induced in the liver during starvation, in BAT during cold exposure, and in skeletal muscle
E

during physical exercise (87). PGC-1α null mice show abnormal diurnal rhythms of body
temperature, activity and metabolic rate, due to disrupted clock and metabolic gene
expression (88). Moreover, PGC1 family members exhibit circadian expression and, in turn,
C

PGC1α stimulates the expression of Bmal1, Clock, Per2 and Rev-erbα, through co-activation
of the ROR family (88,89).
N

AMP-activated protein kinase (AMPK):


AMPK is a sensor of the energy status within cells, whose phosphorylation activates cellular
A

catabolism (90,91). When activated, AMPK phosphorylates and hence activates casein kinase
I ε (CKIε), which leads to PERs degradation (92). AMPK also phosphorylates and, as a
V

result, destabilizes CRY1 in mouse fibroblasts (93) (Fig. 1). PERs and CRYs degradation
relieves CLOCK:BMAL1 inhibition leading to a phase advance in the circadian expression in
D

some tissues (63). Indeed, metformin, an indirect AMPK activator, leads to alterations of the
circadian phase in a tissue-specific manner (94).
A

Sirtuin 1 (SIRT1):
SIRT1, a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase, is a
catabolic factor involved in transcriptional silencing (95,96). SIRT1 is connected to the core
clock mechanism by interacting with CLOCK to deacetylate BMAL1 and PER2 (97,98).
Deacetylated PER2 is further phosphorylated and degraded relieving CLOCK:BMAL1
inhibition (Fig. 1). Hepatic circadian transcriptome analyses revealed that differently from
SIRT1, SIRT6 interacts with CLOCK:BMAL1 and controls their recruitment to gene
promoters. In addition, SIRT6 also controls circadian chromatin recruitment of sterol
regulatory element-binding protein 1 (SREBP-1), resulting in the cyclic regulation of genes
involved in fatty acid and cholesterol metabolism (99). It turns out that the CLOCK:BMAL1
heterodimer as well as SIRT1 regulate the circadian expression of NAMPT (nicotinamide

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 4
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

phosphoribosyltransferase), a rate-limiting enzyme in the NAD+ salvage pathway, therefore,


enabling the circadian synthesis of the coenzyme required for SIRT1 activity (100,101).
SIRT1 activity is also enhanced by AMPK, which increases cellular NAD+ levels (102). High
levels of NAD+ inhibit DNA binding of CLOCK:BMAL1 (103).
Mammalian target of rapamycin (mTOR):
mTOR is an anabolic factor involved in protein synthesis and integrates the input from
multiple upstream pathways, including insulin, growth factors, and mitogens, and functions
as a sensor of cellular nutrient and energy levels (104). The mTOR pathway is regulated by
light in the SCN (105). One of the key factors in the mTOR pathway, protein 70 S6 kinase 1
(P70S6K1), rhythmically phosphorylates BMAL1 allowing it to both associate with the
translational machinery and stimulate circadian oscillations of protein synthesis (106) (Fig.

LE
1). Thus, the mTOR signaling pathway, in addition to its role in protein translation, it is
linked to the circadian clock mechanism.
ADVANCE ARTICLE: Endocrine Reviews

4. Chronodisruption and metabolic disorders

IC
Chronodisruption is defined as the chronic desynchronization of the 24-h rhythms, resulting
in adverse health effects (107). More specifically, chronodisruption occurs when the
synchronization between external environmental cues and internal physiological processes is

T
lost. This can either result in a total loss of rhythmicity, a reduction in rhythm amplitudes or
in phase differences between the SCN and peripheral clocks (108). The interrelations

R
between the key metabolic factors mentioned above and the core clock mechanism may
explain why metabolic chronodisruption is detrimental.
A
Clock gene mutants and knockouts demonstrate the most compelling linkage between
metabolic disorders and the circadian clock (Table 1). Mice with a mutated Clock gene
(Clock∆19 mice) have a dampened diurnal feeding rhythm, are obese, and they develop a
E

metabolic syndrome of hyperleptinemia, hyperlipidemia, hepatic steatosis and hyperglycemia


(109). Combination of the Clock∆19 mutation with the leptin knockout (ob/ob) leads to
significantly heavier mice than the ob/ob phenotype (110), reiterating the contribution of
C

clock disruption to the obese phenotype (55,57,111). Compared with wild type mice, Per2-/-
mice fed a high-fat diet developed significant obesity (112).
N

Bmal1-/- knockout mice exhibit suppressed diurnal changes in triglycerides and glucose
levels as well as no gluconeogenesis (113). Hyperinsulinemic-euglycemic clamps showed
A

that Bmal1-/- mice exhibit no circadian rhythm in insulin action (114). Adipocyte-specific
deletion of Bmal1 resulted in obesity in mice. As adipocytes signal the levels of stored energy
to the brain, Bmal1 deletion led to changes in the expression of hypothalamic neuropeptides
V

that regulate appetite emphasizing the role the adipocyte clock in the temporal organization
of energy regulation (115).
D

In mouse studies, circadian changes in hepatic Cry1/2 expression were sufficient to


modulate gluconeogenesis (116), emphasizing the important role CRY proteins play in
A

regulating hepatic glucose output (117). In addition, Cry1/2-/- mice had an increased
vulnerability to high-fat diet-induced obesity that correlated with increased insulin secretion
and lipid storage in adipose tissue (118,119) and constitutive elevation of proinflammatory
cytokines (120). SCN-mutant mice showed a significant number of rhythmic transcripts
(121). Thus, although it is known that the SCN clock regulates lipolytic and lipogenic
processes (122), recent findings in tissue-specific knockouts indicate that peripheral clocks
play a crucial role in regulation of whole body energy homeostasis.
Consistent with these findings, genetic polymorphisms in human clock genes have been
associated with metabolic alteration (Table 1). Although mutations are rare in humans, it is

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 5
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

rather common to have a single nucleotide polymorphism (SNP) that underlies differences in
our vulnerability to disease.
CLOCK:
It was first demonstrated that several variants at CLOCK were associated with obesity,
especially abdominal obesity (123). Later that year, it was confirmed that CLOCK could play
a relevant role in the development of the metabolic syndrome (MetS), type 2 diabetes and
cardiovascular disease (124). Subsequently, we demonstrated that several genetic variants in
clock genes were related to obesity and related diseases, such as MetS. For example, CLOCK
SNPs (rs3749474, rs4580704 and rs1801260 (3111T>C)) were associated with obesity and
insulin resistance-related variables (125). Systolic and diastolic blood pressure values were
also strongly associated with SNP rs4580704 (125). Furthermore, these genetic variants were

LE
associated with energy intake (126). Minor alleles ate more, specifically fat, and were more
obese, whereas minor allele carriers (A) of CLOCK rs4580704 showed decreased risk of
developing diabetes (31% lower) and hypertension (46% lower) than non-carriers (125).
ADVANCE ARTICLE: Endocrine Reviews

Some of these associations were functionally explained by the presence of a polymorphism

IC
involving a change in the structure of the mRNA leading to a change in gene expression
(127). In addition, people carrying CLOCK 3111C (risk C carriers: including TC and CC) are
more likely to be obese, and exhibit greater difficulties in losing weight. Further studies

T
performed to understand the connections between CLOCK rs1801260 (3111T>C) and
resistance to weight loss showed that risk carriers (C) displayed significant abnormalities at

R
the daily rhythms of wrist temperature and activity. These abnormalities included a) lower
amplitude; b) greater rhythm fragmentation; c) less stable patterns; and d) significantly
A
decreased circadian function. C carriers were also less active, started their activities later in
the morning and were sleepier during the day, showing a delayed acrophase (maximum
expression) that characterizes 'evening-type' subjects (128). Moreover, C carriers have higher
parasympathetic activity during daytime (when it is supposed to be low) than TT carriers and
E

reduced daily rhythms of autonomic nervous system (ANS) function (129). These changes
correlated with weight loss resistance, as low sympathetic activity and high parasympathetic
C

activity reduce energy expenditure and predispose onset of obesity (MONA LISA theory)
(130).
N

PER2:
PER2 SNPs (rs2304672C>G and rs4663302C>T) has been associated with abdominal
A

obesity (131). In particular, PER2 rs2304672C>G minor allele carriers (6% of the
population) displayed several obesogenic behaviors, such as an increased attrition of the
V

weight loss treatment, increased frequency of snacking, stress while dieting, eating while
bored and skipping breakfast, when compared to non-carriers (131).
D

BMAL2:
BMAL2 SNPs have also been associated with high risk of developing type 2 diabetes in obese
patients. The A/G and A/A genotypes of BMAL2 rs7958822 showed significantly higher odds
A

ratios for type 2 diabetes than the G/G genotype among obese men and women. There were
no significant associations between BMAL1 rs11022775 or rs2290035 genotypes and type 2
diabetes (132).
REV-ERBα:
REVERBα rs2314339 has been associated with obesity in two independent populations:
Mediterranean and North American. However, as opposed to other clock gene variants,
which are associated with obesity through changes in dietary intake, REVERBα rs2314339 is
associated with obesity through a decrease in physical activity (133). These findings were
consistent with those obtained in experimental animals showing that Rev-erbα−/− mice
showed a significantly lower spontaneous locomotor activity than their wild-type littermates

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 6
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

(134). However, mice lacking Rev-erbα displayed marked hyperactivity and impaired
response habituation when introduced to a new environment (135).
CRY1:
Genetic variants at CRY1 have been connected to glucose metabolism. Meta-analysis
performed in two independent populations, Mediterranean and European origin North
American, indicated that an increase in carbohydrate intake was associated with a significant
increase in homeostasis model assessment of insulin resistance (HOMA-IR), fasting insulin
and a decrease in quantitative insulin sensitivity check index (QUICKI), only among
individuals homozygous for the minor C allele at CRY1 rs2287161. These findings support
the notion that there is a strong link between the circadian system and glucose metabolism
and suggests the importance this CRY1 locus for insulin resistance and diabetes risk (136).

LE
5. Meal timing, chronodisruption and metabolic alterations
Several behaviors also help to explain the connections between chronodisruption and
ADVANCE ARTICLE: Endocrine Reviews

IC
metabolic alterations. Meal timing, such as restricting food to a particular time of day,
defined as time restricting feeding (TRF) has profound effects on the behavior and
physiology of animals. 2-4 h before the meal, the animals display food anticipatory behavior,
which is demonstrated by an increase in locomotor activity, body temperature, corticosterone

T
secretion, gastrointestinal motility, and activity of digestive enzymes (137-140), all are
known output systems of the biological clock. Moreover, changing the time of food intake to

R
unusual times as happens when nocturnal mice are fed a high-fat diet only during the 12-h
sleep (light) phase, is associated with an increase in weight gain (141). In humans, unusual
A
feeding time produces a disruption of the circadian system, which might produce unhealthy
consequences (142). The timing of food intake may have a significant role in obesity (141),
as well as in weight loss and glucose metabolism (143). For example, mice with adipocyte-
E

specific deletion of Bmal1 fed a high-fat diet during the light period gained significantly more
weight compared to mice fed during the dark period. Thus, disruption of the adipocyte clock
leads to obesity without an overall increase in daily caloric intake when mice are fed during
C

the inactive phase (115). In humans, late eating has been shown to be predictive of weight
loss difficulties during a 20-week dietary intervention conducted in 420 obese and overweight
N

individuals (144). The effect was independent of the total 24-h caloric intake (144). In
addition, insulin sensitivity was lower in late eaters compared to early eaters. Moreover, a
A

twelve-week clinical study showed that those subjects assigned to high caloric intake during
dinner lost significantly less weight than those assigned to high caloric intake during
breakfast. Interestingly, there were no significant differences in the total amount of food or in
V

energy expenditure as assayed by a physical activity questionnaire (145).


The physiological explanation for the metabolic alterations related to late eating is that
D

when feeding patterns are changed, for example during shift work in humans or by means of
restricted access to chow in rodents, this has profound effects on peripheral oscillator
A

regulation, while SCN rhythms remain largely unaltered (146,147). Indeed, in animal models
it has been demonstrated that temporal feeding restriction can change the phase
of circadian gene expression in peripheral cell types by up to 12 h while leaving the phase of
cyclic gene expression in the SCN unaffected (146). Similarly, a recent in-lab study has
demonstrated that a 5-h delay in meals leads to a 1.5-h delay in the circadian rhythm of the
clock gene PER2 in adipose tissue, while no significant changes were found in the central
clock (148).

6. Circadian regulation of adipose tissue in health and disease

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 7
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

As all peripheral tissues, the adipocyte clock mechanism regulates cellular functions (30).
More importantly, these circadian rhythms persist in the absence of the SCN, for example, in
adipose tissue explant cultures (149). To ensure proper nutrient/energy flux and substrate use
by the organism, the circulating levels of secreted adipokines, such as leptin and adiponectin,
key players in the regulation of energy metabolism, display an oscillatory profile with a 24-h
period (122) (Fig. 3). Indeed, it was shown that BAT glucose uptake in mice and humans
reveals a diurnal rhythm that could in part be generated by signals from plasma hormones
(150,151). Alterations of this timing may have important detrimental metabolic
consequences. Interestingly, the circadian rhythms of adipokine expression/secretion differ
depending on the location of the adipose tissue, as significant differences were found both in
their relative acrophase and amplitude between subcutaneous and visceral adipose tissues

LE
(152). Analyses of various genes implicated in metabolic processes, such as energy intake
and expenditure, insulin resistance, adipocyte differentiation, dyslipidemia, and body fat
distribution indicated that circadian rhythmicity followed a predictable physiological pattern,
ADVANCE ARTICLE: Endocrine Reviews

particularly for subcutaneous depots (25,152). Visceral depots also show a higher amplitude

IC
for all glucocorticoid-related genes and (152) it secretes more pro-inflammatory factors than
subcutaneous adipose tissue (153). However, induction of lipolysis is easier in visceral fat
depots, due to the increased sensitivity to catecholamines (154).

T
Leptin:
The adipocyte-derived leptin, which acts at specific receptors in the hypothalamus to

R
suppress appetite and increase metabolism, is extremely important in obesity. Plasma leptin
levels are circadian with leptin peaking early in the non-active phase, that is during the early
A
dark phase in diurnal animals, such as monkeys and humans (155,156), and during the early
to mid-light phase in nocturnal animals, such as rats and mice (157,158) (Fig. 3). SCN
ablation was shown to eliminate leptin circadian rhythmicity in rodents, suggesting that the
central circadian clock regulates leptin expression (157). In obese subjects, leptin retains
E

diurnal variation in release, but with lower amplitude (159), suggesting that blunted circadian
variation may play a role in leptin resistance and obesity (160). If exogenous leptin is
C

continuously applied, desynchronized feeding during the light-phase failed to cause excessive
weight gain in ob/ob mice, but if it was applied in a timed pulse during the light phase,
N

obesity ensued (161). Thus, desynchronized feeding can affect the endogenous rhythm of
leptin to influence metabolic signals that lead to weight gain.
A

Adiponectin:
In human adipose tissue, the expression of adiponectin achieved its zenith (maximum) during
V

the morning, which could be implicated in the maximal withdrawal of fatty acids and the
improvement in glucose tolerance at that time of the day (162). As adiponectin promotes
insulin sensitivity, its acrophase should precede insulin sensitivity, which reaches its
D

maximum level at noon (152,163,164). PPARγ, which participates both in the clock
mechanism (Fig. 1) and adiponectin expression, could also be related to adiponectin circadian
A

expression. In fact, the high expression of PPARγ during the morning is consistent with
results obtained in nocturnal mammals (56) and could be influencing the further increase in
adiponectin expression and the increase in insulin sensitivity during this time of the day.
Corticosteroids:
It has been described that in all species the maximum of circulating corticosteroid rhythms
occurs just before or at the onset of activity (165,166). Over the course of the day, they fall,
reaching low or undetectable levels an hour or two before bedtime. In human adipose tissue,
the antiphase relationship between leptin and glucocorticoids receptors (Fig. 3) (152) is
reasonable considering that both hormones are strongly interrelated and they exert opposite
functions in food intake regulation. While leptin displays an anorexigenic role,

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 8
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

glucocorticoids increase appetite (orexigenic function). Indeed, plasma leptin ultradian pulses
were inversely correlated with those of adrenocorticotropic hormone (ACTH) and cortisol
(164).
Insulin:
Adipose tissue lipogenesis is highly regulated by insulin. Intake of sugars, like glucose, leads
to elevated insulin concentrations in the blood, which, in turn, increase glucose uptake by
upregulating glucose transporter 4 (GLUT4) expression on adipocyte membranes (167). This
process increases the substrate for glycolysis and, consequently, fatty acid biosynthesis in
adipocytes (168). As mentioned above, insulin sensitivity varies according to time of day,
with decreased values in the evening and at night in humans. Indeed, it was demonstrated that
insulin sensitivity in human subcutaneous adipose tissue displays a circadian rhythm and that

LE
it reaches its maximum (acrophase) around noon with sensitivity 54% higher than during
midnight (Fig. 3). In contrast, no circadian rhythms in insulin sensitivity were detected in
visceral adipose tissue (163). Mechanisms responsible for the diurnal variation in insulin
ADVANCE ARTICLE: Endocrine Reviews

sensitivity warrant further study.

IC
7. Role of the circadian clock in adipose tissue differentiation
Currently, it is known that key clock proteins, not only regulate the “time specific” functions

T
in adipose tissue, but they also play a direct role in adipogenesis in both WAT and BAT.
However, some of the reported results are still contradictory.
WAT adipogenesis:
R
PER2, a negative regulator of BMAL1 expression, was found to interact with PPARγ and
A
repress its proadipogenic activity (169) (Fig. 4). Indeed, PER2-deficient mice display altered
lipid metabolism with drastic reduction of total triacylglycerol and non-esterified fatty acids.
PER2 exerts its inhibitory function by blocking PPARγ recruitment to target promoters and
E

thereby inhibiting transcriptional activation (169). As mentioned above, PPARγ expression is


positively regulated by the CLOCK:BMAL1 heterodimer and it positively regulates Bmal1
C

gene expression. Indeed, as mentioned above, an agonist for REV-ERB, which negatively
regulates Bmal1 expression, suppressed mouse orexinergic gene expression, whereas REV-
N

ERBβ-deficient mice had increased orexinergic transcripts (74). In addition, cell-based


studies demonstrate that activation of REV-ERBα with either synthetic or natural (heme)
ligands is required for adipocyte differentiation (158). Thus, PPARγ repression by PER2 and
A

activation of REV-ERB, which lead to BMAL1 downregulation, would be expected to


attenuate adipogenesis. However, surprisingly, knockdown of BMAL1 led to increased
V

adipogenesis, adipocyte hypertrophy and obesity in mice and promoted adipogenic


differentiation in preadipocyte and mesenchymal stem cells (115,170,171) (Fig. 4). This was
D

achieved by down-regulation of genes in the canonical Wnt pathway, an evolutionarily


conserved pathway that regulates crucial aspects of cell fate determination (172). Wnt
pathway is also known to suppress adipogenesis, as BMAL1 binds to the promoter of genes
A

at Wnt pathway to increase their transcription. These contradictory results could stem from a
change in the levels of the involved proteins throughout the adipogenic process. Further
studies are needed in order to delineate the role of BMAL1 in adipogenesis and how other
clock regulators fit into this mechanism.
BAT adipogenesis:
BMAL1 inhibits adipogenesis and thermogenic capacity (173). Global ablation of BMAL1 in
mice and adipocyte-selective inactivation increases brown fat mass and cold tolerance.
BMAL1 inhibits brown adipogenesis through direct transcriptional control of key
components of the cellular differentiation signaling pathways of transforming growth factor β
(TGFβ) and bone morphogenetic protein (BMP); activation of TGFβ or blockade of BMP

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 9
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

pathways suppresses enhanced differentiation in BMAL1-deficient brown adipocytes (173).


REV-ERBα, the negative regulator of BMAL1 expression, promotes brown adipose tissue
development and adipogenesis. As opposed to BMAL1, REV-ERBα represses key
components of the TGFβ signaling pathway (174), an inhibitory pathway of brown fat
development (175,176). Thus, in WAT, BMAL1 primarily regulates the canonical Wnt
cascade, whereas in BAT, regulation of the TGFβ pathway is predominant (Fig. 4). Whether
the effects of BMAL1 and REV-ERBα are pleotropic or demonstrate the physiological
significance of the clock in BAT thermogenesis remain to be determined.

8. Future perspectives
Research during the last years has focused on characterizing the molecular mechanisms by

LE
which circadian clocks impact adipose tissue physiology. Synchronization of the molecular
clocks in the different types of adipose tissues with the rest of the body still merits further
investigation. The connections of circadian genetic variants and obesity now help to
ADVANCE ARTICLE: Endocrine Reviews

understand the importance of the circadian system in adipose-related illnesses, such as

IC
obesity. More importantly, they may help to design individual therapies to obesity based on
the circadian system in order to increase the effectiveness of the treatments. Identification of
certain compounds capable of manipulating clock function at the cellular level is critical to

T
reprogram adipose tissue physiology.
Despite the immense knowledge achieved in the relevance of the internal clock in health

R
and disease, there is still a big gap between this molecular knowledge and clinical practice.
As explained above, a big effort has been done in the last years in order to connect the results
observed in experimental models (animals, tissues, cells) to humans. Currently, large-scale
A
epidemiological studies are repeatedly demonstrating that the alteration of the circadian
system has an impact on health and is associated with several adipose tissue-related
metabolic illnesses, such as obesity, metabolic syndrome or diabetes. Nevertheless, the
E

success in translating this knowledge to the clinical practice is still limited.


To achieve this goal, it is necessary a) to increase the understanding of the importance of
C

the circadian regulation of the endocrine system in medical practice; b) to develop new tools
that help to assess the circadian system performance in clinical practice; c) to design new
N

therapies based on the regulation of the circadian system in order to increase the effectiveness
of treatments of adipose tissue-related pathologies. Salivary samples, which may be non-
invasively collected at several time-points throughout the day, are useful to determine the
A

daily pattern of hormones that connect the circadian system and adipose tissue. In addition,
chronodisruption scores to assess metabolic syndrome in the clinical practice (177) or the use
V

of devices that allow to continuously measure activity, body or wrist skin temperature or
heart rates may help to assess daily patterns in the habitual life of the patient (178,179) and,
D

as a result, the circadian system performance.


In conclusion, timing is a crucial aspect in adipose tissue metabolism in order to ensure
A

the correct accumulation of fat during periods of excess energy or TAG mobilization when
energy requirements are not achieved. Therefore, different adipokines have to be secreted at
the correct time to confer the proper endocrine, paracrine or autocrine function. The current
knowledge of the presence of an internal clock in the different adipose tissue types, white,
brown and beige, and its connection with key elements in metabolism may help us to achieve
a better understanding of adipose tissue function, and to design novel strategies to combat
obesity.
Corresponding authors: *Oren Froy: Tel: +972-8-9489746 Fax: +972-8-9363208
E-mail: oren.froy@mail.huji.ac.il, *Marta Garaulet: Tel: +34-868-88-39-30 Fax:
+34-868-88-39-63 E-mail: garaulet@um.es

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 10
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

Disclosure statement:
The authors have nothing to disclose

References
1. Panda S, Hogenesch JB, Kay SA. Circadian rhythms from flies to human. Nature
2002; 417:329-335
2. Reppert SM, Weaver DR. Coordination of circadian timing in mammals. Nature
2002; 418:935-941
3. Schibler U, Ripperger J, Brown SA. Peripheral circadian oscillators in mammals: time
and food. J Biol Rhythms 2003; 18:250-260
4. Bass J, Lazar MA. Circadian time signatures of fitness and disease. Science 2016;

LE
354:994-999
5. Takahashi JS. Transcriptional architecture of the mammalian circadian clock. Nat Rev
Genet 2017; 18:164-179
ADVANCE ARTICLE: Endocrine Reviews

6. Gibson EM, Williams WP, 3rd, Kriegsfeld LJ. Aging in the circadian system:

IC
considerations for health, disease prevention and longevity. Exp Gerontol 2009; 44:51-56
7. Fu L, Pelicano H, Liu J, Huang P, Lee C. The circadian gene Period2 plays an
important role in tumor suppression and DNA damage response in vivo. Cell 2002; 111:41-50
8. Filipski E, King VM, Li X, Granda TG, Mormont MC, Claustrat B, Hastings MH,

T
Levi F. Disruption of circadian coordination accelerates malignant growth in mice. Pathol
Biol 2003; 51:216-219
9.
R
Davis S, Mirick DK. Circadian disruption, shift work and the risk of cancer: a
summary of the evidence and studies in Seattle. Cancer Causes Control 2006; 17:539-545
A
10. Montagnana M, Salvagno GL, Lippi G. Circadian variation within hemostasis: an
underrecognized link between biology and disease? Semin Thromb Hemost 2009; 35:23-33
11. Anea CB, Zhang M, Stepp DW, Simkins GB, Reed G, Fulton DJ, Rudic RD. Vascular
E

disease in mice with a dysfunctional circadian clock. Circulation 2009; 119:1510-1517


12. Jung-Hynes B, Reiter RJ, Ahmad N. Sirtuins, melatonin and circadian rhythms:
C

building a bridge between aging and cancer. J Pineal Res 2010; 48:9-19
13. Foster RG, Peirson SN, Wulff K, Winnebeck E, Vetter C, Roenneberg T. Sleep and
circadian rhythm disruption in social jetlag and mental illness. Prog Mol Biol Transl Sci
N

2013; 119:325-346
14. Touitou Y. Adolescent sleep misalignment: a chronic jet lag and a matter of public
A

health. J Physiol Paris 2013; 107:323-326


15. McFadden E, Jones ME, Schoemaker MJ, Ashworth A, Swerdlow AJ. The
V

relationship between obesity and exposure to light at night: cross-sectional analyses of over
100,000 women in the Breakthrough Generations Study. Am J Epidemiol 2014; 180:245-250
16. Garaulet M, Ordovas JM, Madrid JA. The chronobiology, etiology and
D

pathophysiology of obesity. Int J Obes (Lond) 2010; 34:1667-1683


17. Welsh DK, Logothetis DE, Meister M, Reppert SM. Individual neurons dissociated
A

from rat suprachiasmatic nucleus express independently phased circadian firing rhythms.
Neuron 1995; 14:697-706
18. Liu C, Weaver DR, Strogatz SH, Reppert SM. Cellular construction of a circadian
clock: period determination in the suprachiasmatic nuclei. Cell 1997; 91:855-860
19. Herzog ED, Takahashi JS, Block GD. Clock controls circadian period in isolated
suprachiasmatic nucleus neurons. Nat Neurosci 1998; 1:708-713
20. Reppert SM, Weaver DR. Molecular analysis of mammalian circadian rhythms. Annu
Rev Physiol 2001; 63:647-676
21. Lee C, Etchegaray JP, Cagampang FR, Loudon AS, Reppert SM. Posttranslational
mechanisms regulate the mammalian circadian clock. Cell 2001; 107:855-867

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 11
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

22. Froy O, Chapnik N. Circadian oscillation of innate immunity components in mouse


small intestine. Mol Immunol 2007; 44:1954-1960
23. Young ME. The circadian clock within the heart: potential influence on myocardial
gene expression, metabolism, and function. Am J Physiol Heart Circ Physiol 2006; 290:H1-
H16
24. Kornmann B, Preitner N, Rifat D, Fleury-Olela F, Schibler U. Analysis of circadian
liver gene expression by ADDER, a highly sensitive method for the display of differentially
expressed mRNAs. Nucleic Acids Res 2001; 29:E51
25. Akhtar RA, Reddy AB, Maywood ES, Clayton JD, King VM, Smith AG, Gant TW,
Hastings MH, Kyriacou CP. Circadian cycling of the mouse liver transcriptome, as revealed
by cDNA microarray, is driven by the suprachiasmatic nucleus. Curr Biol 2002; 12:540-550

LE
26. Duffield GE, Best JD, Meurers BH, Bittner A, Loros JJ, Dunlap JC. Circadian
programs of transcriptional activation, signaling, and protein turnover revealed by microarray
analysis of mammalian cells. Curr Biol 2002; 12:551-557
ADVANCE ARTICLE: Endocrine Reviews

27. Panda S, Antoch MP, Miller BH, Su AI, Schook AB, Straume M, Schultz PG, Kay

IC
SA, Takahashi JS, Hogenesch JB. Coordinated transcription of key pathways in the mouse by
the circadian clock. Cell 2002; 109:307-320
28. Storch KF, Lipan O, Leykin I, Viswanathan N, Davis FC, Wong WH, Weitz CJ.

T
Extensive and divergent circadian gene expression in liver and heart. Nature 2002; 417:78-83
29. Kita Y, Shiozawa M, Jin W, Majewski RR, Besharse JC, Greene AS, Jacob HJ.
Implications of circadian gene expression in kidney, liver and the effects of fasting on

30. R
pharmacogenomic studies. Pharmacogenetics 2002; 12:55-65
Zvonic S, Ptitsyn AA, Conrad SA, Scott LK, Floyd ZE, Kilroy G, Wu X, Goh BC,
A
Mynatt RL, Gimble JM. Characterization of peripheral circadian clocks in adipose tissues.
Diabetes 2006; 55:962-970
31. Reddy AB, Karp NA, Maywood ES, Sage EA, Deery M, O'Neill JS, Wong GK,
E

Chesham J, Odell M, Lilley KS, Kyriacou CP, Hastings MH. Circadian orchestration of the
hepatic proteome. Curr Biol 2006; 16:1107-1115
C

32. McCarthy JJ, Andrews JL, McDearmon EL, Campbell KS, Barber BK, Miller BH,
Walker JR, Hogenesch JB, Takahashi JS, Esser KA. Identification of the circadian
N

transcriptome in adult mouse skeletal muscle. Physiol Genomics 2007; 31:86-95


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

Science 2001; 294:2511-2515


34. Cheng MY, Bullock CM, Li C, Lee AG, Bermak JC, Belluzzi J, Weaver DR, Leslie
V

FM, Zhou QY. Prokineticin 2 transmits the behavioural circadian rhythm of the
suprachiasmatic nucleus. Nature 2002; 417:405-410
D

35. Kraves S, Weitz CJ. A role for cardiotrophin-like cytokine in the circadian control of
mammalian locomotor activity. Nat Neurosci 2006; 9:212-219
36. Froy O, Chang DC, Reppert SM. Redox potential: differential roles in dCRY and
A

mCRY1 functions. Curr Biol 2002; 12:147-152


37. Preitner N, Damiola F, Lopez-Molina L, Zakany J, Duboule D, Albrecht U, Schibler
U. The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the
positive limb of the mammalian circadian oscillator. Cell 2002; 110:251-260
38. Sato TK, Panda S, Miraglia LJ, Reyes TM, Rudic RD, McNamara P, Naik KA,
FitzGerald GA, Kay SA, Hogenesch JB. A functional genomics strategy reveals Rora as a
component of the mammalian circadian clock. Neuron 2004; 43:527-537
39. Ueda HR, Hayashi S, Chen W, Sano M, Machida M, Shigeyoshi Y, Iino M,
Hashimoto S. System-level identification of transcriptional circuits underlying mammalian
circadian clocks. Nat Genet 2005; 37:187-192

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 12
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

40. Dunlap JC. Molecular bases for circadian clocks. Cell 1999; 96:271-290
41. Siiteri PK. Adipose tissue as a source of hormones. Am J Clin Nutr 1987; 45:277-282
42. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional
cloning of the mouse obese gene and its human homologue. Nature 1994; 372:425-432
43. Aldhahi W, Hamdy O. Adipokines, inflammation, and the endothelium in diabetes.
Curr Diab Rep 2003; 3:293-298
44. Chen Y, Pan R, Pfeifer A. Fat tissues, the brite and the dark sides. Pflugers Arch
2016; 468:1803-1807
45. Vitali A, Murano I, Zingaretti MC, Frontini A, Ricquier D, Cinti S. The adipose organ
of obesity-prone C57BL/6J mice is composed of mixed white and brown adipocytes. J Lipid
Res 2012; 53:619-629

LE
46. Lafontan M. Advances in adipose tissue metabolism. Int J Obes (Lond) 2008; 32
Suppl 7:S39-51
47. Cannon B, Nedergaard J. Brown adipose tissue: function and physiological
ADVANCE ARTICLE: Endocrine Reviews

significance. Physiol Rev 2004; 84:277-359

IC
48. Yoneshiro T, Aita S, Matsushita M, Kayahara T, Kameya T, Kawai Y, Iwanaga T,
Saito M. Recruited brown adipose tissue as an antiobesity agent in humans. J Clin Invest
2013; 123:3404-3408

T
49. Stanford KI, Middelbeek RJ, Townsend KL, An D, Nygaard EB, Hitchcox KM,
Markan KR, Nakano K, Hirshman MF, Tseng YH, Goodyear LJ. Brown adipose tissue
regulates glucose homeostasis and insulin sensitivity. J Clin Invest 2013; 123:215-223
50.
R
Bartelt A, Bruns OT, Reimer R, Hohenberg H, Ittrich H, Peldschus K, Kaul MG,
Tromsdorf UI, Weller H, Waurisch C, Eychmuller A, Gordts PL, Rinninger F, Bruegelmann
A
K, Freund B, Nielsen P, Merkel M, Heeren J. Brown adipose tissue activity controls
triglyceride clearance. Nat Med 2011; 17:200-205
51. Seale P, Bjork B, Yang W, Kajimura S, Chin S, Kuang S, Scime A, Devarakonda S,
E

Conroe HM, Erdjument-Bromage H, Tempst P, Rudnicki MA, Beier DR, Spiegelman BM.
PRDM16 controls a brown fat/skeletal muscle switch. Nature 2008; 454:961-967
C

52. Lepper C, Fan CM. Inducible lineage tracing of Pax7-descendant cells reveals
embryonic origin of adult satellite cells. Genesis 2010; 48:424-436
N

53. Wang W, Seale P. Control of brown and beige fat development. Nat Rev Mol Cell
Biol 2016; 17:691-702
54. Wu J, Bostrom P, Sparks LM, Ye L, Choi JH, Giang AH, Khandekar M, Virtanen
A

KA, Nuutila P, Schaart G, Huang K, Tu H, van Marken Lichtenbelt WD, Hoeks J, Enerback
S, Schrauwen P, Spiegelman BM. Beige adipocytes are a distinct type of thermogenic fat cell
V

in mouse and human. Cell 2012; 150:366-376


55. Froy O. Metabolism and circadian rhythms--implications for obesity. Endocr Rev
D

2010; 31:1-24
56. Yang X, Downes M, Yu RT, Bookout AL, He W, Straume M, Mangelsdorf DJ, Evans
RM. Nuclear receptor expression links the circadian clock to metabolism. Cell 2006;
A

126:801-810
57. Garaulet M, Madrid JA. Chronobiological aspects of nutrition, metabolic syndrome
and obesity. Adv Drug Deliv Rev 2010; 62:967-978
58. La Fleur SE, Kalsbeek A, Wortel J, Buijs RM. A suprachiasmatic nucleus generated
rhythm in basal glucose concentrations. J Neuroendocrinol 1999; 11:643-652
59. La Fleur SE. Daily rhythms in glucose metabolism: suprachiasmatic nucleus output to
peripheral tissue. J Neuroendocrinol 2003; 15:315-322
60. Davidson AJ, Castanon-Cervantes O, Stephan FK. Daily oscillations in liver function:
diurnal vs circadian rhythmicity. Liver Int 2004; 24:179-186

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 13
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

61. Ramsey KM, Marcheva B, Kohsaka A, Bass J. The clockwork of metabolism. Annu
Rev Nutr 2007; 27:219-240
62. Pendergast JS, Branecky KL, Yang W, Ellacott KL, Niswender KD, Yamazaki S.
High-fat diet acutely affects circadian organisation and eating behavior. Eur J Neurosci 2013;
37:1350-1356
63. Barnea M, Madar Z, Froy O. High-fat diet delays and fasting advances the circadian
expression of adiponectin signaling components in mouse liver. Endocrinology 2009;
150:161-168
64. Barnea M, Madar Z, Froy O. High-fat diet followed by fasting disrupts circadian
expression of adiponectin signaling pathway in muscle and adipose tissue. Obesity (Silver
Spring) 2010; 18:230-238

LE
65. Kohsaka A, Laposky AD, Ramsey KM, Estrada C, Joshu C, Kobayashi Y, Turek FW,
Bass J. High-fat diet disrupts behavioral and molecular circadian rhythms in mice. Cell
Metab 2007; 6:414-421
ADVANCE ARTICLE: Endocrine Reviews

66. Pivovarova O, Jurchott K, Rudovich N, Hornemann S, Ye L, Mockel S, Murahovschi

IC
V, Kessler K, Seltmann AC, Maser-Gluth C, Mazuch J, Kruse M, Busjahn A, Kramer A,
Pfeiffer AF. Changes of Dietary Fat and Carbohydrate Content Alter Central and Peripheral
Clock in Humans. J Clin Endocrinol Metab 2015; 100:2291-2302

T
67. Chawla A, Lazar MA. Induction of Rev-ErbA alpha, an orphan receptor encoded on
the opposite strand of the alpha-thyroid hormone receptor gene, during adipocyte
differentiation. J Biol Chem 1993; 268:16265-16269
68.
R
Bray MS, Young ME. Circadian rhythms in the development of obesity: potential role
for the circadian clock within the adipocyte. Obes Rev 2007; 8:169-181
A
69. Torra IP, Tsibulsky V, Delaunay F, Saladin R, Laudet V, Fruchart JC, Kosykh V,
Staels B. Circadian and glucocorticoid regulation of Rev-erbalpha expression in liver.
Endocrinology 2000; 141:3799-3806
E

70. Cho H, Zhao X, Hatori M, Yu RT, Barish GD, Lam MT, Chong LW, DiTacchio L,
Atkins AR, Glass CK, Liddle C, Auwerx J, Downes M, Panda S, Evans RM. Regulation of
C

circadian behaviour and metabolism by REV-ERB-alpha and REV-ERB-beta. Nature 2012;


485:123-127
N

71. Lau P, Nixon SJ, Parton RG, Muscat GE. RORalpha regulates the expression of genes
involved in lipid homeostasis in skeletal muscle cells: caveolin-3 and CPT-1 are direct targets
of ROR. J Biol Chem 2004; 279:36828-36840
A

72. Ueda HR, Chen W, Adachi A, Wakamatsu H, Hayashi S, Takasugi T, Nagano M,


Nakahama K, Suzuki Y, Sugano S, Iino M, Shigeyoshi Y, Hashimoto S. A transcription
V

factor response element for gene expression during circadian night. Nature 2002; 418:534-
539
D

73. Solt LA, Wang Y, Banerjee S, Hughes T, Kojetin DJ, Lundasen T, Shin Y, Liu J,
Cameron MD, Noel R, Yoo SH, Takahashi JS, Butler AA, Kamenecka TM, Burris TP.
Regulation of circadian behaviour and metabolism by synthetic REV-ERB agonists. Nature
A

2012; 485:62-68
74. Amador A, Wang Y, Banerjee S, Kameneka TM, Solt LA, Burris TP.
Pharmacological and Genetic Modulation of REV-ERB Activity and Expression Affects
Orexigenic Gene Expression. PLoS One 2016; 11:e0151014
75. Delezie J, Dumont S, Dardente H, Oudart H, Grechez-Cassiau A, Klosen P, Teboul
M, Delaunay F, Pevet P, Challet E. The nuclear receptor REV-ERBalpha is required for the
daily balance of carbohydrate and lipid metabolism. FASEB J 2012; 26:3321-3335
76. Gerhart-Hines Z, Feng D, Emmett MJ, Everett LJ, Loro E, Briggs ER, Bugge A, Hou
C, Ferrara C, Seale P, Pryma DA, Khurana TS, Lazar MA. The nuclear receptor Rev-
erbalpha controls circadian thermogenic plasticity. Nature 2013; 503:410-413

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 14
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

77. Desvergne B, Wahli W. Peroxisome proliferator-activated receptors: nuclear control


of metabolism. Endocr Rev 1999; 20:649-688
78. Kersten S, Desvergne B, Wahli W. Roles of PPARs in health and disease. Nature
2000; 405:421-424
79. Lefebvre P, Chinetti G, Fruchart JC, Staels B. Sorting out the roles of PPAR alpha in
energy metabolism and vascular homeostasis. J Clin Invest 2006; 116:571-580
80. Braissant O, Foufelle F, Scotto C, Dauca M, Wahli W. Differential expression of
peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPAR-alpha, -
beta, and -gamma in the adult rat. Endocrinology 1996; 137:354-366
81. Chen L, Yang G. PPARs Integrate the Mammalian Clock and Energy Metabolism.
PPAR Res 2014; 2014:653017

LE
82. Oishi K, Shirai H, Ishida N. CLOCK is involved in the circadian transactivation of
peroxisome-proliferator-activated receptor alpha (PPARalpha) in mice. Biochem J 2005;
386:575-581
ADVANCE ARTICLE: Endocrine Reviews

83. Inoue I, Shinoda Y, Ikeda M, Hayashi K, Kanazawa K, Nomura M, Matsunaga T, Xu

IC
H, Kawai S, Awata T, Komoda T, Katayama S. CLOCK/BMAL1 is involved in lipid
metabolism via transactivation of the peroxisome proliferator-activated receptor (PPAR)
response element. J Atheroscler Thromb 2005; 12:169-174

T
84. Canaple L, Rambaud J, Dkhissi-Benyahya O, Rayet B, Tan NS, Michalik L, Delaunay
F, Wahli W, Laudet V. Reciprocal regulation of brain and muscle Arnt-like protein 1 and
peroxisome proliferator-activated receptor alpha defines a novel positive feedback loop in the

85. R
rodent liver circadian clock. Mol Endocrinol 2006; 20:1715-1727
Gutman R, Barnea M, Haviv L, Chapnik N, Froy O. Peroxisome proliferator-activated
A
receptor alpha (PPARalpha) activation advances locomotor activity and feeding daily
rhythms in mice. Int J Obes (Lond) 2012; 36:1131-1134
86. Yang G, Jia Z, Aoyagi T, McClain D, Mortensen RM, Yang T. Systemic
E

PPARgamma deletion impairs circadian rhythms of behavior and metabolism. PLoS One
2012; 7:e38117
C

87. Li S, Lin JD. Transcriptional control of circadian metabolic rhythms in the liver.
Diabetes Obes Metab 2015; 17 Suppl 1:33-38
N

88. Liu C, Li S, Liu T, Borjigin J, Lin JD. Transcriptional coactivator PGC-1alpha


integrates the mammalian clock and energy metabolism. Nature 2007; 447:477-481
89. Grimaldi B, Sassone-Corsi P. Circadian rhythms: metabolic clockwork. Nature 2007;
A

447:386-387
90. Carling D. AMP-activated protein kinase: balancing the scales. Biochimie 2005;
V

87:87-91
91. Hardie DG, Hawley SA, Scott JW. AMP-activated protein kinase--development of the
D

energy sensor concept. J Physiol 2006; 574:7-15


92. Um JH, Yang S, Yamazaki S, Kang H, Viollet B, Foretz M, Chung JH. Activation of
5'-AMP-activated kinase with diabetes drug metformin induces casein kinase Iepsilon
A

(CKIepsilon)-dependent degradation of clock protein mPER2. J Biol Chem 2007; 282:20794-


20798
93. Lamia KA, Sachdeva UM, DiTacchio L, Williams EC, Alvarez JG, Egan DF,
Vasquez DS, Juguilon H, Panda S, Shaw RJ, Thompson CB, Evans RM. AMPK regulates the
circadian clock by cryptochrome phosphorylation and degradation. Science 2009; 326:437-
440
94. Barnea M, Haviv L, Gutman R, Chapnik N, Madar Z, Froy O. Metformin affects the
circadian clock and metabolic rhythms in a tissue-specific manner. Biochim Biophys Acta
2012; 1822:1796-1806

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 15
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

95. Haigis MC, Guarente LP. Mammalian sirtuins--emerging roles in physiology, aging,
and calorie restriction. Genes Dev 2006; 20:2913-2921
96. Canto C, Auwerx J. Caloric restriction, SIRT1 and longevity. Trends Endocrinol
Metab 2009; 20:325-331
97. Asher G, Gatfield D, Stratmann M, Reinke H, Dibner C, Kreppel F, Mostoslavsky R,
Alt FW, Schibler U. SIRT1 regulates circadian clock gene expression through PER2
deacetylation. Cell 2008; 134:317-328
98. Nakahata Y, Kaluzova M, Grimaldi B, Sahar S, Hirayama J, Chen D, Guarente LP,
Sassone-Corsi P. The NAD+-dependent deacetylase SIRT1 modulates CLOCK-mediated
chromatin remodeling and circadian control. Cell 2008; 134:329-340
99. Masri S, Rigor P, Cervantes M, Ceglia N, Sebastian C, Xiao C, Roqueta-Rivera M,

LE
Deng C, Osborne TF, Mostoslavsky R, Baldi P, Sassone-Corsi P. Partitioning circadian
transcription by SIRT6 leads to segregated control of cellular metabolism. Cell 2014;
158:659-672
ADVANCE ARTICLE: Endocrine Reviews

100. Nakahata Y, Sahar S, Astarita G, Kaluzova M, Sassone-Corsi P. Circadian control of

IC
the NAD+ salvage pathway by CLOCK-SIRT1. Science 2009; 324:654-657
101. Peek CB, Affinati AH, Ramsey KM, Kuo HY, Yu W, Sena LA, Ilkayeva O,
Marcheva B, Kobayashi Y, Omura C, Levine DC, Bacsik DJ, Gius D, Newgard CB,

T
Goetzman E, Chandel NS, Denu JM, Mrksich M, Bass J. Circadian clock NAD+ cycle drives
mitochondrial oxidative metabolism in mice. Science 2013; 342:1243417
102. Canto C, Gerhart-Hines Z, Feige JN, Lagouge M, Noriega L, Milne JC, Elliott PJ,

R
Puigserver P, Auwerx J. AMPK regulates energy expenditure by modulating NAD+
metabolism and SIRT1 activity. Nature 2009; 458:1056-1060
A
103. Rutter J, Reick M, Wu LC, McKnight SL. Regulation of clock and NPAS2 DNA
binding by the redox state of NAD cofactors. Science 2001; 293:510-514
104. Li F, Yin Y, Tan B, Kong X, Wu G. Leucine nutrition in animals and humans: mTOR
E

signaling and beyond. Amino Acids 2011; 41:1185-1193


105. Cao R, Lee B, Cho HY, Saklayen S, Obrietan K. Photic regulation of the mTOR
C

signaling pathway in the suprachiasmatic circadian clock. Mol Cell Neurosci 2008; 38:312-
324
N

106. Lipton JO, Yuan ED, Boyle LM, Ebrahimi-Fakhari D, Kwiatkowski E, Nathan A,
Guttler T, Davis F, Asara JM, Sahin M. The Circadian Protein BMAL1 Regulates Translation
in Response to S6K1-Mediated Phosphorylation. Cell 2015; 161:1138-1151
A

107. Erren TC, Reiter RJ. Defining chronodisruption. J Pineal Res 2009; 46:245-247
108. Laermans J, Depoortere I. Chronobesity: role of the circadian system in the obesity
V

epidemic. Obes Rev 2016; 17:108-125


109. Turek FW, Joshu C, Kohsaka A, Lin E, Ivanova G, McDearmon E, Laposky A,
D

Losee-Olson S, Easton A, Jensen DR, Eckel RH, Takahashi JS, Bass J. Obesity and
metabolic syndrome in circadian Clock mutant mice. Science 2005; 308:1043-1045
110. Oishi K, Ohkura N, Wakabayashi M, Shirai H, Sato K, Matsuda J, Atsumi G, Ishida
A

N. CLOCK is involved in obesity-induced disordered fibrinolysis in ob/ob mice by regulating


PAI-1 gene expression. J Thromb Haemost 2006; 4:1774-1780
111. Green CB, Takahashi JS, Bass J. The meter of metabolism. Cell 2008; 134:728-742
112. Yang S, Liu A, Weidenhammer A, Cooksey RC, McClain D, Kim MK, Aguilera G,
Abel ED, Chung JH. The role of mPer2 clock gene in glucocorticoid and feeding rhythms.
Endocrinology 2009;
113. Rudic RD, McNamara P, Curtis AM, Boston RC, Panda S, Hogenesch JB, Fitzgerald
GA. BMAL1 and CLOCK, two essential components of the circadian clock, are involved in
glucose homeostasis. PLoS Biol 2004; 2:e377

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 16
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

114. Shi SQ, Ansari TS, McGuinness OP, Wasserman DH, Johnson CH. Circadian
disruption leads to insulin resistance and obesity. Curr Biol 2013; 23:372-381
115. Paschos GK, Ibrahim S, Song WL, Kunieda T, Grant G, Reyes TM, Bradfield CA,
Vaughan CH, Eiden M, Masoodi M, Griffin JL, Wang F, Lawson JA, Fitzgerald GA. Obesity
in mice with adipocyte-specific deletion of clock component Arntl. Nat Med 2012; 18:1768-
1777
116. Zhang EE, Liu Y, Dentin R, Pongsawakul PY, Liu AC, Hirota T, Nusinow DA, Sun
X, Landais S, Kodama Y, Brenner DA, Montminy M, Kay SA. Cryptochrome mediates
circadian regulation of cAMP signaling and hepatic gluconeogenesis. Nat Med 2010;
16:1152-1156
117. Hatori M, Panda S. CRY links the circadian clock and CREB-mediated

LE
gluconeogenesis. Cell Res 2010; 20:1285-1288
118. Barclay JL, Shostak A, Leliavski A, Tsang AH, Johren O, Muller-Fielitz H, Landgraf
D, Naujokat N, van der Horst GT, Oster H. High-fat diet-induced hyperinsulinemia and
ADVANCE ARTICLE: Endocrine Reviews

tissue-specific insulin resistance in Cry-deficient mice. Am J Physiol Endocrinol Metab 2013;

IC
304:E1053-1063
119. Froy O. A CRY for help to fight fat. Am J Physiol Endocrinol Metab 2013;
304:E1129-1130

T
120. Narasimamurthy R, Hatori M, Nayak SK, Liu F, Panda S, Verma IM. Circadian clock
protein cryptochrome regulates the expression of proinflammatory cytokines. Proc Natl Acad
Sci U S A 2012; 109:12662-12667

R
121. Kolbe I, Husse J, Salinas G, Lingner T, Astiz M, Oster H. The SCN Clock Governs
Circadian Transcription Rhythms in Murine Epididymal White Adipose Tissue. J Biol
A
Rhythms 2016; 31:577-587
122. Shostak A, Husse J, Oster H. Circadian regulation of adipose function. Adipocyte
2013; 2:201-206
E

123. Sookoian S, Gemma C, Gianotti TF, Burgueno A, Castano G, Pirola CJ. Genetic
variants of Clock transcription factor are associated with individual susceptibility to obesity.
C

Am J Clin Nutr 2008; 87:1606-1615


124. Scott EM, Carter AM, Grant PJ. Association between polymorphisms in the Clock
N

gene, obesity and the metabolic syndrome in man. Int J Obes (Lond) 2008; 32:658-662
125. Garaulet M, Lee YC, Shen J, Parnell LD, Arnett DK, Tsai MY, Lai CQ, Ordovas JM.
CLOCK genetic variation and metabolic syndrome risk: modulation by monounsaturated
A

fatty acids. Am J Clin Nutr 2009; 90:1466-1475


126. Garaulet M, Corbalan MD, Madrid JA, Morales E, Baraza JC, Lee YC, Ordovas JM.
V

CLOCK gene is implicated in weight reduction in obese patients participating in a dietary


programme based on the Mediterranean diet. Int J Obes (Lond) 2010; 34:516-523
D

127. Garaulet M, Lee YC, Shen J, Parnell LD, Arnett DK, Tsai MY, Lai CQ, Ordovas JM.
Genetic variants in human CLOCK associate with total energy intake and cytokine sleep
factors in overweight subjects (GOLDN population). European journal of human genetics :
A

EJHG 2010; 18:364-369


128. Bandin C, Martinez-Nicolas A, Ordovas JM, Ros Lucas JA, Castell P, Silvente T,
Madrid JA, Garaulet M. Differences in circadian rhythmicity in CLOCK 3111T/C genetic
variants in moderate obese women as assessed by thermometry, actimetry and body position.
Int J Obes (Lond) 2013; 37:1044-1050
129. Garaulet M. CLOCK 3111T/C genetic variant influences the daily rhythm of
autonomic nervous function: relevance to body weight control. Int J Obes 2017; in press
130. Messina G, De Luca V, Viggiano A, Ascione A, Iannaccone T, Chieffi S, Monda M.
Autonomic nervous system in the control of energy balance and body weight: personal
contributions. Neurol Res Int 2013; 2013:639280

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 17
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

131. Garaulet M, Corbalan-Tutau MD, Madrid JA, Baraza JC, Parnell LD, Lee YC,
Ordovas JM. PERIOD2 variants are associated with abdominal obesity, psycho-behavioral
factors, and attrition in the dietary treatment of obesity. Journal of the American Dietetic
Association 2010; 110:917-921
132. Yamaguchi M, Uemura H, Arisawa K, Katsuura-Kamano S, Hamajima N, Hishida A,
Suma S, Oze I, Nakamura K, Takashima N, Suzuki S, Ibusuki R, Mikami H, Ohnaka K,
Kuriyama N, Kubo M, Tanaka H, Japan Multi-institutional Collaborative Cohort Study G.
Association between brain-muscle-ARNT-like protein-2 (BMAL2) gene polymorphism and
type 2 diabetes mellitus in obese Japanese individuals: A cross-sectional analysis of the Japan
Multi-institutional Collaborative Cohort Study. Diabetes Res Clin Pract 2015; 110:301-308
133. Garaulet M, Smith CE, Gomez-Abellan P, Ordovas-Montanes M, Lee YC, Parnell

LE
LD, Arnett DK, Ordovas JM. REV-ERB-ALPHA circadian gene variant associates with
obesity in two independent populations: Mediterranean and North American. Molecular
nutrition & food research 2014; 58:821-829
ADVANCE ARTICLE: Endocrine Reviews

134. Woldt E, Sebti Y, Solt LA, Duhem C, Lancel S, Eeckhoute J, Hesselink MK, Paquet

IC
C, Delhaye S, Shin Y, Kamenecka TM, Schaart G, Lefebvre P, Neviere R, Burris TP,
Schrauwen P, Staels B, Duez H. Rev-erb-alpha modulates skeletal muscle oxidative capacity
by regulating mitochondrial biogenesis and autophagy. Nat Med 2013; 19:1039-1046

T
135. Jager J, O'Brien WT, Manlove J, Krizman EN, Fang B, Gerhart-Hines Z, Robinson
MB, Klein PS, Lazar MA. Behavioral changes and dopaminergic dysregulation in mice
lacking the nuclear receptor Rev-erbalpha. Mol Endocrinol 2014; 28:490-498

R
136. Dashti HS, Smith CE, Lee YC, Parnell LD, Lai CQ, Arnett DK, Ordovas JM,
Garaulet M. CRY1 circadian gene variant interacts with carbohydrate intake for insulin
A
resistance in two independent populations: Mediterranean and North American. Chronobiol
Int 2014; 31:660-667
137. Saito M, Murakami E, Suda M. Circadian rhythms in disaccharidases of rat small
E

intestine and its relation to food intake. Biochim Biophys Acta 1976; 421:177-179
138. Honma KI, Honma S, Hiroshige T. Critical role of food amount for prefeeding
C

corticosterone peak in rats. Am J Physiol 1983; 245:R339-R344


139. Comperatore CA, Stephan FK. Entrainment of duodenal activity to periodic feeding. J
N

Biol Rhythms 1987; 2:227-242


140. Stephan FK. The "other" circadian system: food as a Zeitgeber. J Biol Rhythms 2002;
17:284-292
A

141. Arble DM, Bass J, Laposky AD, Vitaterna MH, Turek FW. Circadian timing of food
intake contributes to weight gain. Obesity (Silver Spring) 2009; 17:2100-2102
V

142. Morris CJ, Yang JN, Garcia JI, Myers S, Bozzi I, Wang W, Buxton OM, Shea SA,
Scheer FA. Endogenous circadian system and circadian misalignment impact glucose
D

tolerance via separate mechanisms in humans. Proc Natl Acad Sci U S A 2015; 112:E2225-
2234
143. Garaulet M, Gomez-Abellan P. Timing of food intake and obesity: A novel
A

association. Physiology & behavior 2014;


144. Garaulet M, Gomez-Abellan P, Alburquerque-Bejar JJ, Lee YC, Ordovas JM, Scheer
FA. Timing of food intake predicts weight loss effectiveness. Int J Obes (Lond) 2013;
37:604-611
145. Jakubowicz D, Barnea M, Wainstein J, Froy O. High Caloric intake at breakfast vs.
dinner differentially influences weight loss of overweight and obese women. Obesity (Silver
Spring) 2013;
146. Damiola F, Le Minh N, Preitner N, Kornmann B, Fleury-Olela F, Schibler U.
Restricted feeding uncouples circadian oscillators in peripheral tissues from the central
pacemaker in the suprachiasmatic nucleus. Genes Dev 2000; 14:2950-2961

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 18
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

147. Stokkan KA, Yamazaki S, Tei H, Sakaki Y, Menaker M. Entrainment of the circadian
clock in the liver by feeding. Science 2001; 291:490-493
148. Wehrens SMT, Christou S, Isherwood C, Middleton B, Gibbs MA, Archer SN, Skene
DJ, Johnston JD. Meal Timing Regulates the Human Circadian System. Curr Biol 2017;
27:1768-1775 e1763
149. Gomez-Santos C, Gomez-Abellan P, Madrid JA, Hernandez-Morante JJ, Lujan JA,
Ordovas JM, Garaulet M. Circadian rhythm of clock genes in human adipose explants.
Obesity (Silver Spring) 2009; 17:1481-1485
150. van der Veen DR, Shao J, Chapman S, Leevy WM, Duffield GE. A diurnal rhythm in
glucose uptake in brown adipose tissue revealed by in vivo PET-FDG imaging. Obesity
(Silver Spring) 2012; 20:1527-1529

LE
151. Lee P, Bova R, Schofield L, Bryant W, Dieckmann W, Slattery A, Govendir MA,
Emmett L, Greenfield JR. Brown Adipose Tissue Exhibits a Glucose-Responsive
Thermogenic Biorhythm in Humans. Cell Metab 2016; 23:602-609
ADVANCE ARTICLE: Endocrine Reviews

152. Garaulet M, Ordovas JM, Gomez-Abellan P, Martinez JA, Madrid JA. An

IC
approximation to the temporal order in endogenous circadian rhythms of genes implicated in
human adipose tissue metabolism. J Cell Physiol 2011; 226:2075-2080
153. Hamdy O, Porramatikul S, Al-Ozairi E. Metabolic obesity: the paradox between

T
visceral and subcutaneous fat. Curr Diabetes Rev 2006; 2:367-373
154. Arner P. Differences in lipolysis between human subcutaneous and omental adipose
tissues. Ann Med 1995; 27:435-438

R
155. Kalra SP, Bagnasco M, Otukonyong EE, Dube MG, Kalra PS. Rhythmic, reciprocal
ghrelin and leptin signaling: new insight in the development of obesity. Regul Pept 2003;
A
111:1-11
156. Downs JL, Urbanski HF. Aging-related sex-dependent loss of the circulating leptin
24-h rhythm in the rhesus monkey. J Endocrinol 2006; 190:117-127
E

157. Kalsbeek A, Fliers E, Romijn JA, La Fleur SE, Wortel J, Bakker O, Endert E, Buijs
RM. The suprachiasmatic nucleus generates the diurnal changes in plasma leptin levels.
C

Endocrinology 2001; 142:2677-2685


158. Sukumaran S, Almon RR, DuBois DC, Jusko WJ. Circadian rhythms in gene
N

expression: Relationship to physiology, disease, drug disposition and drug action. Adv Drug
Deliv Rev 2010; 62:904-917
159. Licinio J. Longitudinally sampled human plasma leptin and cortisol concentrations
A

are inversely correlated. J Clin Endocrinol Metab 1998; 83:1042


160. Heptulla R, Smitten A, Teague B, Tamborlane WV, Ma YZ, Caprio S. Temporal
V

patterns of circulating leptin levels in lean and obese adolescents: relationships to insulin,
growth hormone, and free fatty acids rhythmicity. J Clin Endocrinol Metab 2001; 86:90-96
D

161. Arble DM, Vitaterna MH, Turek FW. Rhythmic leptin is required for weight gain
from circadian desynchronized feeding in the mouse. PLoS One 2011; 6:e25079
162. Gomez-Abellan P, Gomez-Santos C, Madrid JA, Milagro FI, Campion J, Martinez
A

JA, Ordovas JM, Garaulet M. Circadian expression of adiponectin and its receptors in human
adipose tissue. Endocrinology 2010; 151:115-122
163. Carrasco-Benso MP, Rivero-Gutierrez B, Lopez-Minguez J, Anzola A, Diez-Noguera
A, Madrid JA, Lujan JA, Martinez-Augustin O, Scheer FA, Garaulet M. Human adipose
tissue expresses intrinsic circadian rhythm in insulin sensitivity. FASEB J 2016; 30:3117-
3123
164. Gavrila A, Peng CK, Chan JL, Mietus JE, Goldberger AL, Mantzoros CS. Diurnal
and ultradian dynamics of serum adiponectin in healthy men: comparison with leptin,
circulating soluble leptin receptor, and cortisol patterns. J Clin Endocrinol Metab 2003;
88:2838-2843

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 19
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

165. Peterson RE. Plasma corticosterone and hydrocortisone levels in man. J Clin
Endocrinol Metab 1957; 17:1150-1157
166. Garcia-Prieto MD, Tebar FJ, Nicolas F, Larque E, Zamora S, Garaulet M. Cortisol
secretary pattern and glucocorticoid feedback sensitivity in women from a Mediterranean
area: relationship with anthropometric characteristics, dietary intake and plasma fatty acid
profile. Clin Endocrinol (Oxf) 2007; 66:185-191
167. Kiehn JT, Tsang AH, Heyde I, Leinweber B, Kolbe I, Leliavski A, Oster H. Circadian
Rhythms in Adipose Tissue Physiology. Compr Physiol 2017; 7:383-427
168. Bryant NJ, Govers R, James DE. Regulated transport of the glucose transporter
GLUT4. Nat Rev Mol Cell Biol 2002; 3:267-277
169. Grimaldi B, Bellet MM, Katada S, Astarita G, Hirayama J, Amin RH, Granneman JG,

LE
Piomelli D, Leff T, Sassone-Corsi P. PER2 controls lipid metabolism by direct regulation of
PPARgamma. Cell Metab 2010; 12:509-520
170. Guo B, Chatterjee S, Li L, Kim JM, Lee J, Yechoor VK, Minze LJ, Hsueh W, Ma K.
ADVANCE ARTICLE: Endocrine Reviews

The clock gene, brain and muscle Arnt-like 1, regulates adipogenesis via Wnt signaling

IC
pathway. FASEB J 2012; 26:3453-3463
171. Shimba S, Ogawa T, Hitosugi S, Ichihashi Y, Nakadaira Y, Kobayashi M, Tezuka M,
Kosuge Y, Ishige K, Ito Y, Komiyama K, Okamatsu-Ogura Y, Kimura K, Saito M. Deficient

T
of a clock gene, brain and muscle Arnt-like protein-1 (BMAL1), induces dyslipidemia and
ectopic fat formation. PLoS One 2011; 6:e25231
172. Komiya Y, Habas R. Wnt signal transduction pathways. Organogenesis 2008; 4:68-75

R
173. Nam D, Guo B, Chatterjee S, Chen MH, Nelson D, Yechoor VK, Ma K. The
adipocyte clock controls brown adipogenesis through the TGF-beta and BMP signaling
A
pathways. J Cell Sci 2015; 128:1835-1847
174. Nam D, Chatterjee S, Yin H, Liu R, Lee J, Yechoor VK, Ma K. Novel Function of
Rev-erbalpha in Promoting Brown Adipogenesis. Sci Rep 2015; 5:11239
E

175. Koncarevic A, Kajimura S, Cornwall-Brady M, Andreucci A, Pullen A, Sako D,


Kumar R, Grinberg AV, Liharska K, Ucran JA, Howard E, Spiegelman BM, Seehra J,
C

Lachey J. A novel therapeutic approach to treating obesity through modulation of TGFbeta


signaling. Endocrinology 2012; 153:3133-3146
N

176. Yadav H, Quijano C, Kamaraju AK, Gavrilova O, Malek R, Chen W, Zerfas P,


Zhigang D, Wright EC, Stuelten C, Sun P, Lonning S, Skarulis M, Sumner AE, Finkel T,
Rane SG. Protection from obesity and diabetes by blockade of TGF-beta/Smad3 signaling.
A

Cell Metab 2011; 14:67-79


177. Corbalan-Tutau MD, Gomez-Abellan P, Madrid JA, Canteras M, Ordovas JM,
V

Garaulet M. Toward a chronobiological characterization of obesity and metabolic syndrome


in clinical practice. Clin Nutr 2015; 34:477-483
D

178. Martinez-Nicolas A, Ortiz-Tudela E, Rol MA, Madrid JA. Uncovering different


masking factors on wrist skin temperature rhythm in free-living subjects. PLoS One 2013;
8:e61142
A

179. Ortiz-Tudela E, Martinez-Nicolas A, Campos M, Rol MA, Madrid JA. A new


integrated variable based on thermometry, actimetry and body position (TAP) to evaluate
circadian system status in humans. PLoS Comput Biol 2010; 6:e1000996

Figure 1: The relationship between the core clock mechanism and metabolic factors.
CLOCK and BMAL1 mediate the expression of clock- and clock-controlled genes regulating
metabolism. PER1, PER2 and CRY1 serve as the negative feedback loop that inhibits
CLOCK:BMAL1-mediated expression. The catabolic factors SIRT1, PGC1 and AMPK,
when activated under low cellular energy levels, relieve the inhibition mediated by the
negative feedback loop. BMAL1 expression is positively regulated by RORs and PPARs and

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 20
on 17 March 2018
Endocrine Reviews; Copyright 2018 DOI: 10.1210/er.2017-00193

negatively regulated by REV-ERBs. mTOR, an anabolic factor interacts with BMAL1 and
suppresses the activity of the catabolic factors. The green and red arrows denote possible
pathways that activate or inhibit adipocyte metabolism, respectively.

Figure 2: Lineage of white, brown and “brite/beige” adipocytes from mesenchymal stem
cells and their cellular and molecular markers. UCP1 – uncoupling protein 1

Figure 3: Temporal expression of adipocytokines in rodents and humans. The peak


expression of each adipokine is depicted. Representation of the acrophase or timing of the
maximum levels of adipokines expression in human adipose tissue: comparison with human
circulating levels and with rodent adipose tissue mRNA levels. Data is represented as the five

LE
hours of active and resting phase in both humans and rodents because this is the timing when
most adipokines peak.

Figure 4: The relationship between the core clock mechanism and differentiation signaling
ADVANCE ARTICLE: Endocrine Reviews

IC
pathways in brown and white adipose tissues. BMAL1 activates the transcription of PPARγ
and PER2. PER2 inhibits BMAL1 activity and PPARγ increases BMAL1 expression. Wnt
signaling represses WAT adipogenesis, whereas TGFβ/Smad3 signaling represses BAT
formation. BMP signaling induces BAT formation. Reduced levels of BMAL1 lead to

T
increased WAT and BAT formation.

R
Table 1: Clock gene mutations in mice and genetic variants in humans and their effect on
metabolism.
A
Clock Gene Mutant mice Metabolic alterations Human genetic Metabolic alterations in humans
in mutant mice variant
CLOCK Clock∆19 (109) Hyperphagic and obese, CLOCK SNPs Obesity, metabolic syndrome, higher
develop a metabolic (rs3749474, energy and fat intake, higher risk of
E

syndrome of rs4580704, and developing diabetes and hypertension,


hyperleptinemia, rs1801260 alterations in the autonomic nervous
hyperlipidemia, hepatic (3111T>C)) (123- system
steatosis, and 129)
C

hyperglycemia
BMAL1 Bmal1-/- (113) Suppressed diurnal BMAL2 rs7958822 High risk of developing type 2 diabetes
variations in glucose and (132) in obese patients
N

triglycerides and
abolished
gluconeogenesis
Cry1/2(-/-) (116-120)
A

CRYs Increased insulin CRY1 rs2287161 With high intake of carbohydrates,


secretion and lipid (136) carriers of the risk variant have
storage in adipose tissue, increased insulin resistance.
elevation of
V

proinflammatory
cytokines
PER2 Per2-/- (112) On high-fat diet, eat as PER2 SNPs Abdominal obesity, several obesogenic
D

much during the light (rs2304672C>G and behaviors


period as the dark period rs4663302C>T) (131)
and develop
significant obesity
A

REV-ERBα Rev-erbα-/- (134) Reduced spontaneous REV-ERBα Obesity resulting from decreased
locomotor activity rs2314339 (133-135) physical activity

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user 21
on 17 March 2018
LE
Froy and Garaulet, Figure 1
Reviews

Catabolic Factors:

IC
AMPK, PGC1, SIRT1
Endocrinology

T
Clock Negative Feedback Loop
ADVANCE ARTICLE: Endocrine

R
PER1, PER2, CRY1
A
Adipose tissue Anabolic Factor:
CLOCK BMAL1
metabolism mTOR
E
C

Lipid Metabolism Factors:


REV-ERBs
N

Lipid Metabolism Factors:


A

PPARs, RORs
V
D
A

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user
on 17 March 2018
LE
Froy and Garaule
Reviews

IC
Mesenchymal Precursors
Endocrinology

Myf5- Myf5+

T
ADVANCE ARTICLE: Endocrine

WAT Precursor
R BAT Precursor
A
Mitochondrion
Nucleus
Mature Adipocytes
Nucleus
Mitochondrion
E

Lipid droplet
Lipid droplet
White Brite (Beige) Brown
C

UCP1- UCP1+ UCP1+


N
A
V
D
A

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user
on 17 March 2018
LE
Froy and Garaulet, Figure 3
Reviews

Resting Active

IC
phase
Endocrinology

phase
Rodents

T
Humans
ADVANCE ARTICLE: Endocrine

R
A
E

Adiponectin
C

Chemerin
Corticosteroids
N

IL-6
Insulin sensitivity
A

Leptin
PAI-1
V

PPARƳ
Visfatin
D
A

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user
on 17 March 2018
LE
Froy and Garaule
REV-ERBα
Reviews

IC
BMAL1
Endocrinology

PER2

T
ADVANCE ARTICLE: Endocrine

PPARγ
R
A
E

TGFβ/Smad3 signaling BMP signaling Wnt signaling


C
N
A
V

BAT WAT
D
A

Downloaded from https://academic.oup.com/edrv/advance-article-abstract/doi/10.1210/er.2017-00193/4912380


by University of California, Santa Barbara user
on 17 March 2018

You might also like