You are on page 1of 20

SCIENCE CHINA

Life Sciences
•REVIEW•.
. . . . . . . . . . . . . . .. .. . .. .. .. . .. .. ... .. .. . .. .. ... .. .. . .. .. .. . .. .. ... .. .. . .. .. .. . .. .. ... .. .. . .. .. .. . .. .. ... .. .. . .. .. ... .. .. . .. .. .. . https://doi.org/10.1007/s11427-018-9371-4
............................

An overview of energy and metabolic regulation


*
Song Wen, Chaoxun Wang, Min Gong & Ligang Zhou

Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, China

Received July 5, 2018; accepted August 23, 2018; published online October 18, 2018

The physiology and behaviors related to energy balance are monitored by the nervous and humoral systems. Because of the
difficulty in treating diabetes and obesity, elucidating the energy balance mechanism and identifying critical targets for treatment
are important research goals. Therefore, the purpose of this article is to describe energy regulation by the central nervous system
(CNS) and peripheral humoral pathway. Homeostasis and rewarding are the basis of CNS regulation. Anorexigenic or orexigenic
effects reflect the activities of the POMC/CART or NPY/AgRP neurons within the hypothalamus. Neurotransmitters have roles
in food intake, and responsive brain nuclei have different functions related to food intake, glucose monitoring, reward pro-
cessing. Peripheral gut- or adipose-derived hormones are the major source of peripheral humoral regulation systems. Nutrients or
metabolites and gut microbiota affect metabolism via a discrete pathway. We also review the role of peripheral organs, the liver,
adipose tissue, and skeletal muscle in peripheral regulation. We discuss these topics and how the body regulates metabolism.
hedonic, POMC/CART, NPY/AgRP, neurotransmitter, brain nuclei, gut-derived hormone, gut microbiota, BAT

Citation: Wen, S., Wang, C., Gong, M., and Zhou, L. (2018). An overview of energy and metabolic regulation. Sci China Life Sci 61, https://doi.org/10.1007/
s11427-018-9371-4

INTRODUCTION tabolism. Specialized neurons in the hypothalamus and


Bodily functions and activities require fuel from intake of brainstem can sense the messages of energy intake and en-
food and nutrients. Food intake is influenced by conditions, ergy stores in the body. For many decades, researchers have
such as starvation, frequency of eating, and stress. Morbid focused predominantly on physical energy homeostasis, the
hyperphagia and obesity refer to the body’s inability to activities of which are governed predominantly by the nuclei
competently control energy homeostasis. An increasing within the hypothalamus, such as the arcuate nucleus (ARC)
amount of data has pointed to the role of key components in and paraventricular nucleus (PVN) (Schneeberger et al.,
both the central nervous systems (CNS) and peripheral hu- 2014). Various receptors of nutrient and hormonal signals
moral systems governing energy balance. In this review, we distributed to hypothalamic nuclei can be bound by their
discuss the roles of the CNS and humoral systems, micro- ligands derived during or immediately after a meal
biota, liver, brown adipose tissue (BAT) as well as the muscle (Adamska et al., 2014). For instance, the ARC can respond to
regulation to show how the body regulates metabolism. energy balance signals, such as leptin or GLP-1 (Baver et al.,
2014; Secher et al., 2014). However, the reward mechanisms
influencing our intake behaviors are also indispensable for
ENERGY AND METABOLIC REGULATION BY energy balance regulation. It is common that palatable food
THE CNS attracts increased intake even though an individual feels
Homeostatic and reward system sated. Thus, this hedonic behavior that overrides homeostatic
The brain has a central role in the regulation of energy me- eating if continued long term may contribute to over-
consumption and obesity (Zheng and Berthoud, 2008).
*Corresponding author (email: zhouligang@yahoo.com)
The reward system involves a hedonic and incentivized

© Science China Press and Springer-Verlag GmbH Germany, part of Springer Nature 2018 . . . . . . . . . . . . . . . . . . . . . . . life.scichina.com link.springer.com
2. . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ............................................ 2

motivational mechanism. A reward is an attractive and mo- the ventral tegmental area (VTA), nucleus accumbens
tivational property of a stimulus that induces pleasure, and in (NAc), and striatum) (Figure 1). The corticolimbic system
the case of eating, it can cause inappropriate consumption represents the “liking” zone, whereas the mesolimbic system
behavior (Schultz, 2015). The reward system wields distinct represents “wanting” areas (Berridge et al., 2010; Egecioglu
actions through dissimilar brain regions and neural path- et al., 2011). The mesolimbic dopamine system, especially
ways, such as the corticolimbic (e.g., the hippocampus, from the midbrain VTA to the forebrain NAc, is thought to
amygdala, prefrontal cortex) and mesolimbic structures (e.g., be the major rewarding pathway (Wise and Bozarth, 1987).

Figure 1 The illustration shows the reward system which contains links between midbrain and forebrain. Mesolimbic pathway contains contacts among
NAc, VTA, and Striatum whereas corticolimbic pathway contains links among the hippocampus, amygdala, and prefrontal cortex. DLPFC, dorsal lateral
prefrontal cortex; ACC, anterior cingulate cortex; Hippo, hippocampus.
3. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ............................................ 3

This pathway exerts a food intake promoting effect via do- improves insulin resistance. Glucose utilization and insulin
pamine signaling. The morbid obesity phenomenon may be sensitization will be improved in the liver and skeletal
underlain by a reduction in dopamine receptor 2 (D2r) ex- muscle. A selective 5-HT2CR agonist was shown to induce
pression due to prolonged exposure to excessive palatable significant insulin-induced protein kinase B phosphorylation
food intake (Wang et al., 2001; Volkow et al., 2008). Another and a decrease in glucose neogenesis enzymes, such as
finding suggested that hyperphagia in obese individuals may phosphoenolpyruvate carboxykinase and glucose 6-phos-
reflect an altered balance between the “liking” and “wanting” phatase peripherally (Zhou et al., 2007). Activation of the 5-
aspect of food rewards. Motivated actions to obtain palatable HT1B receptor can also mediate the role of MC4R in both
food may increase to compensate for decreased pleasure for POMC neurons and NPY/AgRP neurons, which can reduce
food, which is similar to the mechanism underlying drug production of AgRP and inhibit GABA fibers projecting to
addiction (Egecioglu et al., 2011). In addition to the genes POMC/CART neurons from NPY neurons (Heisler et al.,
that encode similar feeding-related peptides (Olszewski et 2006).
al., 2008), these two systems can respond to similar per- AgRP neurons are restricted to the ARC of the hypotha-
ipheral signals. lamus, with most (~90%) expressing NPY (Anderson et al.,
2016). Administration of NPY centrally induces increased
Anorexic and orexigenic effects food intake through binding to the Y1 or Y5 receptor (Beck,
2006; Nguyen et al., 2012) distributed within the ARC, PVN,
It is well known that food intake can be strongly driven by and ventromedial hypothalamus (VMH). NPY gene expres-
two opposite behaviors: the anorexic effect inhibits feeding, sion and the release of its product can be induced by fasting
and the orexigenic effect stimulates food intake. Anatomical condition accompanied by the observable c-fos expression
and physiological evidence supports two main distinct series (Liu et al., 2012). Central administration of AgRP promotes
of cells within the hypothalamus: orexigenic neurons that hyperphagia and weight gain, which concurrently blocks the
express neuro-peptide Y (NPY) and agouti-related peptide MC3R/MC4R (Schwartz et al., 2000).
(AgRP) and anorexigenic neurons that express proopiome- Circulating nutrients and hormones may also have con-
lanocortin (POMC) and cocaine- and amphetamine-regu- current effects on the two subtype neurons. Elevated extra-
lated transcript (CART) (Figure 2) (Sohn, 2015). cellular glucose level activates POMC neurons, whereas
POMC neurons by post-transcriptional processing of NPY/AgRP neurons are provoked in glucose-deprived con-
POMC express the peptide α-MSH, which has an anorexic ditions. Long-chain fatty acids may also induce decreased
effect on food intake. Binding to the MC3R and MC4R on hypothalamic AgRP and NPY expression (Obici et al., 2002).
second-order neurons influences metabolism and thereby Increased malonyl CoA can also inhibit intake (Lane et al.,
induces the anorexigenic effect (Fan et al., 1997). One study 2005). Leptin and CCK may enhance the action of POMC or
showed that MC4R knockout mice or human individuals CART (Cowley et al., 2001).
with acquired MC4R mutants were associated with early
onset severe obesity (Huszar et al., 1997; Srisai et al., 2011). Critical neurotransmitters or peptides related to central
CART is co-expressed with POMC within the ARC regulation
(Chronwall, 1985) and suppresses food intake and con-
currently modulates energy expenditure (Wang et al., 2000). Neurotransmitters act as central signals that coordinate the
CART can encourage satiety when presented to hypotha- activities of the homeostatic and reward systems. They act on
lamic second-order neurons (Lau and Herzog, 2014). Injec- different correspondent receptors to induce effects on food
tion of CART can reduce feeding in rodents, and treatment intake, energy expenditure, and body weight. Indeed, some
with CART antiserum reduces hypophagia (Kristensen et al., current medications available for obesity treatment are de-
1998). Similarly, mutants or polymorphisms of the CART signed to target certain neurotransmitters that effect energy
gene have been associated with an increased incidence of regulation through the CNS (Table 1).
obesity (Challis et al., 2000).
Our previous studies demonstrated that POMC neurons Dopamine
expressing the 5-HT2C receptor (5-HT2CR) can be activated Dopamine is the primary neurotransmitter related to rewards.
by serotonin (5-HT), which improves glucose metabolism. Dopamine-producing neurons in the substantia nigra (SN) of
The activation can trigger increased production of α-MSH the brainstem and the VTA in the midbrain project to the
derived from precursor POMC. α-MSH acts on MCR4 dis- brain nuclei or areas related to food reward (Levey et al.,
tributed within the intermediolateral nucleus of the spinal 1993; Björklund and Dunnett, 2007; Taber et al., 2012).
cord, which contains sympathetic preganglionic neurons. Dopamine signaling pathway activation increases the
Through this action, the function of pancreas islet endocrine “wanting” property in the reward system (Peciña et al., 2003)
or GI tract hormone secretion could be improved, which and can interact with a satiety signal, such as those associated
4. . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ............................................ 4

Figure 2 The two subtype neurons related to food intake locate in the ARC. One subtype neurons express POMC/CART. It acts on melanocortin receptor
(MCR) by α-melanocyte stimulating hormone (α-MSH), which generates anorexigenic effect. In contrast, NPY/AgRP expressed neurons may lead to
orexigenic effect via either NPY binding to Y1R and Y5R or AgRP indirectly blocking the activation of α-MSH on MCR. 3rd V, the third ventricle; VMN,
ventromedial nucleus; MC3R/MC4R, melanocortin receptor 3/4; Y1R/Y5R, neuropeptide receptor 1/5.

with leptin and GLP-1 (Hommel et al., 2006; Mietlicki- food intake. CB1 exerts its orexigenic effect mainly by ac-
Baase et al., 2013). Binding to the D1 receptor (D1r) in- tivating the cells of the NAc and interacting with the leptin,
creased palatable food intake and decreased normal chow ghrelin, DA, and opioidergic signaling pathways within or
intake, whereas activation of the D2r or D3 receptor showed beyond the hypothalamus (Pagotto et al., 2006). When in-
the opposite outcome in rodents (Cooper and Al-Naser, fused in rats, which were randomly provided a 3-component
2006). It is thought that evaluation of the reward process diet containing carbohydrates, protein, and fat, the rats in-
relies on activation of the dopamine D2r (Baik, 2013). As creased their carbohydrate intake; this suggests that en-
referred to previously, a deficit in D2r signaling may con- docannabinoids may selectively influence the appetite for
tribute to excessive intake to compensate for reduced plea- macronutrients (Escartín-Pérez et al., 2009). The CB1 an-
sure during eating (van de Giessen et al., 2014). However, tagonists showed a tendency to selectively reduce appetizing
pharmacological intervention targeting the dopaminergic food intake, such as of sucrose or chocolate-flavored pellets
(DA) system has shown various outcomes in human and (Arnone et al., 1997; Droste et al., 2010). Consistent with
animal studies (Bojanowska and Ciosek, 2016), which sug- these findings, normal human subjects who had received
gests that targeting of the DA system may be a promising CB1 antagonist therapy showed decreased responses in
approach but currently needs further investigation to unravel certain neural areas related to rewarding observed by func-
the complicated effects of the DA system on food intake. tional magnetic resonance imaging (fMRI) (Horder et al.,
2010). Therefore, utilization of antagonists of CB1 may at-
Endocannabinoids tenuate appetizing food intake and be useful for obesity and
Two major endocannabinoids affecting fat intake are ana- type 2 diabetes mellitus (T2DM) therapy.
ndamide (N-arachidonoylethanolamide) and 2-arachidonoyl
glycerol produced from cell membrane phospholipids fol- Orexin
lowing cell stimulation. Generated centrally and periph- Orexin A and B act as orexigenic neuropeptides. The neu-
erally, both eventually bind to the cannabinoid receptor 1 rons that produce these neuropeptides are located within the
(CB1) predominantly distributed in the CNS, which affects lateral hypothalamic area (LHA), perifornical area, dor-
5. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ............................................ 5

a)
Table 1 Summary on neurotransmitters in relation to energy balance regulation

Categorizes Neurotransmitter Derivations Projection Receptor Food intake Energy Body weight
expenditure
NAc, striatum, VTA,
Anandamide hypothalamus, *
Endocannabinoids (AEA) brainstem, In situ (presynaptic) CB1r Orexigenic Decrease Increase
hippocampus, medulla
NAc, striatum, basal
ganglia, amygdala,
2-Arachidonoyl hypothalamus, In situ (presynaptic)
*
CB1r Orexigenic Decrease Increase
glycerol (2-AG) brainstem,
hippocampus, medulla
Perifornical area, LH, VTA, ventromedial pre-
Orexins Orexin DMH, posterior frontal cortex, hypotha- OXR1, OXR2 Orexigenic Increase Decrease
hypothalamus lamus, brainstem

Hypothalamus, Hypothalamus, singu-


Opioids Endorphins pituitary gland, late cortex, hippocam- μ-Opioid recptor Orexigenic Decrease Increase
brainstem pus, locus coeruleus,
NAc
Hypothalamus,
Enkephalin neurohypophysis, Nac, VP, Nac, hypothalamus μ-, δ-Opioid Orexigenic Decrease Increase
receptor
amygdala
Hypothalamus, VTA, Hypothalamus, VTA,
Dynorphin Nac Nac, brainstem κ-Opioid recptor Orexigenic Decrease Increase

Biogenic amines Dopamine VTA, SN Prefrontal cortex, Nac, D1r,D2r, D3r, Orexigenic/ Decrease Increase
striatum D4r anorexigenic
Locus coeruleus, Hypothalamus,
Noradrenaline lateral tegmental prefrontal cortex, Nac, α, β receptor Orexigenic/ Decrease/
#
Decrease/#
field amygdala anorexigenic increase increase

Hypothalamus, Nac, 5-HT1A, 5-HT1B,


Caudal nuclei, rostral VTA, SN, hippocampus,
Serotonin nuclei amygdala, prefrontal 5-HT1D, 5-HT2C, Anorexigenic Increase Decrease
cortex 5-HT4, 5-HT6

Hypothalamus, NAc,
Histamine Tuberomammillary VTA, hippocampus, H1r Anorexigenic Increase Decrease
nucleus amygdala, prefrontal
cortex
Forebrain nuclei, Orexigenic/ #
Acetylcholine Acetylcholine brainstem nuclei, Hypothalamus, VTA M3, α3, α7, β4 anorexigenic Increase Decrease
hypothalamus
Hypothalamus, VTA, Hypothalamus, VTA,
Amino acids Glutamate striatum, brainstem Nac, hippocampus mGlu 5 Orexigenic Increase Decrease

Gamma-Amino- Hypothalamus, VTA, Hypothalamus, VTA, Orexigenic/ Decrease/# Decrease/#


butyric acid striatum Striatum GABA A, GABA B anorexigenic# increase increase
(GABA)
Peptides CRF PVN Neurohypophysis CRF2 receptor Anorexigenic Increase Decrease
AgRP ARC Hypothalamus MC3R/MC4R Orexigenic Decrease Increase
NPY PVN Hypothalamus Y1/Y5 receptor Orexigenic Decrease Increase

Oxytocin PVN, amygdala, Hypothalamus, VTA, Oxytocin receptor Anorexigenic Increase Decrease
BNST Nac, brainstem
a) *, This type of neurotransmitter acts as a retrograde signal released from postsynaptic and bind to receptor localized on presynaptic. #, The effect of this
neurotransmitter depends on activation on the certain subtype of receptor.

somedial hypothalamus (DMH), and posterior hypothala- energy expenditure eventually to decrease body weight.
mus. The orexins bind to orexin receptor 1 and 2 (OXR1 and
OXR2). Within the hypothalamus, OX1R is most highly Endogenous opioid peptides
expressed in NPY neurons within the VMN, whereas OXR2 There are opioid peptides (e.g., enkephalins, endorphins,
is predominantly expressed in the PVN (Trivedi et al., 1998). dynorphins) and receptors (κ (KOR), δ (DOR), μ (MOR)
Central administration of an orexin antibody or an OX1R opioid receptors) that extensively exist in the CNS. Espe-
antagonist has been shown to decrease food intake (Haynes cially, β-endorphins are derived from the POMC. Increases
et al., 2000; Yamada et al., 2000). Injection of orexin led to in hypothalamic endocannabinoid expression have been
enhanced motivation for food seeking (Borgland et al., shown to lead to orexigenic feeding accompanied by upre-
2009). Orexins can simultaneously increase food intake and gulated expression of POMC, which seems paradoxical at
6. . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ............................................ 6

first sight; however, an increase in POMC reflects the en- 2015). As previously shown, 5-HT can be involved in me-
hanced expression of β-endorphins, which can be attenuated tabolism regulation via melanocortin circuits that lead to
by opioid receptor blockade (Koch et al., 2015). These improved metabolism.
findings suggest an interaction between the opioid system
and endocannabinoid system. Previous studies support that Histamine
stimulating μ-opioid receptors by opioid agonists can cause Histamine induces an anorexigenic effect after it activates
an increased desire for appetizing foods, which suggests that the histamine receptor (H-1r) in the VMN (Teff and Kim,
use of opioid antagonists can prevent morbid energy intake 2011; Torrealba et al., 2012). Activation of H-1r has also
predisposing to obesity (Peciña and Smith, 2010). However, been determined to affect anticipatory food reward behavior.
μ-receptor antagonists in rodent studies have shown ambig- However, given that histamine is primarily released from the
uous results, whereas in human studies, treatment with μ- tuberomammillary nucleus, especially during the appetitive
receptor antagonists showed a decrease in the response to phase of feeding in rats, it only induces transient anorexia
palatable food in reward areas reflected by fMRI (Rabiner et rather than a sense of satiety (Valdés et al., 2010). Another
al., 2011; Murray et al., 2014; Yeomans and Gray, 1996; study also found that histamine in the combination of ser-
Langleben et al., 2012). otonergic and cholinergic medication may regulate food in-
take, whereas respective activation by antagonists had no
Oxytocin effect on food intake (Hartfield et al., 2003).
In addition to regulation of reproductive functions and its
effect on social behaviors, oxytocin has also been referred to Acetylcholine
as a hypothalamic neuropeptide that has an anorexigenic Acetylcholine is colocalized with POMC within the hy-
effect on palatable food intake (Onaka et al., 2012). Rodent pothalamus and can bind to muscarinic M3, nicotinic β4, and
studies have shown that endogenous oxytocin selectively probably α7 receptors to influence food intake (Yamada et
suppressed carbohydrate intake (Olszewski et al., 2010). al., 2001; Mineur et al., 2011; McFadden et al., 2014). An-
Peripherally injected oxytocin also has been shown to reduce other study demonstrated that application of α3 and β4 ni-
food intake both in humans and rodents (Morton et al., 2012; cotinic receptor antagonists reduced food intake and
Ott et al., 2013). prevented weight gain in rats (Taraschenko et al., 2011).
Studies have shown that nicotine can enhance energy ex-
Noradrenaline penditure and affect food preferences (Audrain-McGovern
Noradrenaline either enhances or inhibits food intake by and Benowitz, 2011; Geha et al., 2013), which in turn results
acting on different receptors. Activation of α1-, β2-, and β3- in sweeter flavors of ingested food than sensed by non-
adrenoceptors decreased food intake, whereas stimulation of smokers, but it also lowers body weight in active smokers
the α2-adrenoceptor increased food intake (Adan et al., and is associated with weight gain after cessation of smoke
2008). The related pathway is via the prefrontal cortex, (Allen et al., 2005; Chiolero et al., 2008; Stadler et al., 2014).
which affects decision making in rodents through dopamine
release derived from NAc in the presence of appetitive food. Glutamate
This finding suggests that the DA and noradrenergic systems As an excitatory neurotransmitter, glutamate can affect me-
share the same target for controlling food intake (Ventura et tabolism regulation via metabotropic glutamate receptor 5
al., 2007). Otherwise, noradrenaline can be involved in (mGlu 5) (Bradbury et al., 2005). The VTA can be an origin
regulation through a connection from the brainstem to the site where glutamate is produced. Peripheral glutamate in-
hypothalamus (Wellman, 2000) and amygdala (Savard et al., jection can affect feeding centers, such as the ARC or lateral
1983). hypothalamus (LH) by either direct infiltration through the
blood-brain-barrier (BBB) or indirect activation of vagal
Serotonin afferent neurons (Torii et al., 2013). Recent data has shown
Serotonin (5-HT)-producing neurons in the CNS extend that variations in the genes related to glutamate action or
projections to the area known to be involved in reward, such alteration of glutamatergic receptor expression in regions
as the NAc, VTA, SN, hippocampus, amygdala, and pre- related to rewards could affect food intake or be responsible
frontal cortex (Hensler, 2006; Lechin et al., 2006; Ikemoto, for obesity (Brownley et al., 2015; Locke et al., 2015). In-
2010). Seven receptor families and multiple subtypes are jection of mGlu 5 antagonists has been shown to suppress
distributed in the CNS (Lam et al., 2010; Hayes and food intake (Bisaga et al., 2008).
Greenshaw, 2011). The central 5-HT1A, 5-HT1B or human
counterpart 5-HT1D, 5-HT2C, 5-HT4, and 5-HT6 receptors GABA
have been determined to have distinct roles in food intake GABA acts as an inhibitory neurotransmitter and binds to
regulation (Garfield and Heisler, 2009; Voigt and Fink, GABA A and GABA B receptors to influence food intake. At
7. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ............................................ 7

the ARC level, GABA neurons receive input from AgRP presses receptors, such as leptin, GLP-1, and insulin, which
neurons and attenuate the anorexic effect induced by POMC can activate POMC neurons and inhibit NPY/AgRP neurons
neurons (Cansell et al., 2012). Similarly, GABA can act on to mediate food intake (Maejima et al., 2011; Patterson et al.,
the VTA dopamine system to affect reward feeding (van 2011; Baggio and Drucker, 2014). The AgRP neurons within
Zessen et al., 2012). When binding to the GABA A receptor the ARC are mainly distributed homogeneously throughout
within the VMN, GABA induces an orexigenic signal (Ka- the rostrocaudal axis, whereas POMC neurons are located in
matchi and Rathanaswami, 2012), whereas utilization of the anterior and medial areas (Anderson et al., 2016). Neu-
GABA A receptor antagonists leads to increased perception rons within the ARC can then be activated to influence
of sweetness in milk and selective fat intake when separately downstream second-order neurons, such as PVN or DMH.
infused into the anterolateral hypothalamus and the medial Nucleus of solitary tract (NTS). The NTS is anatomically
ventral pallidum (Kelly et al., 1977; Covelo et al., 2014). not far from the area postrema (AP). The NTS and AP as well
Peripheral administration of a GABA B receptor agonist was as the dorsal motor nucleus of the vagus constitutes the
shown to suppress food intake and to possibly selectively dorsal vagal complex. The DVC is a critical neural con-
reduce motivation or appetite for certain palatable food both nection between the gut and brain. Signals of metabolism,
in rodents and humans (Sato et al., 2007; Arima and Oiso, such as leptin, ghrelin, and glucagon-like peptide-1 (GLP-1)
2010). as well as GIGI satiation signals relayed by the vagal nerve
were found to be eventually processed by neurons within the
Corticotropin releasing factor NTS (Grill and Hayes, 2012). The NTS is also a major site
Corticotropin releasing factor (CRF) has been shown to play from which endogenous GLP-1 is derived. Preproglucagon
a pivotal role in stress and eating disorders. Food restriction, neurons located in the NTS synthesize endogenous GLP-1
commonly known to be a stressor, has been determined to that spreads throughout the CNS (Trapp and Cork, 2015).
cause altered CRF expression in the hypothalamus (Lenglos The NTS has been shown to have extensive relationships
et al., 2013). CRF can be a functional antagonist to NPY and with other nuclei related to food intake in CNS. Current
AgRP expression under stress. Substantial evidence has research found that the majority of NTS neurons are glucose
pointed to the role of CRF2 receptor in reduced feeding. responsive and can be specifically targeted by α-MSH, which
Infusion of a CRF2 receptor agonist in the lateral septum implies a link with the ARC (Mimee and Ferguson, 2015).
reduced intake and encouraged anxiety-like behavior in rats The norepinephrine neurons here were shown to link with the
(Bakshi et al., 2007). Administration of urocortin (UCN)-1, mesolimbic area, such as the VTA and NAC (Richard et al.,
an endogenous CRF2 receptor agonist, was found to induce 2015). Some studies have implicated the noradrenergic A2
anorexic behavior in rats, which was blocked by 5-HT2cR population of DBH-expressing cells in the NTS’ role in sa-
activation. This result suggests that 5-HT signaling activation tiety (Rinaman, 2003; Kreisler et al., 2014), which is in ac-
regulates the CRF system in stress-related eating disorders cordance with subsequent research results showing that
(Harada et al., 2014). These studies demonstrated that the dopamine b-hydroxylase- and cholecystokinin (CCK)-ex-
CRF system is an indispensable signal in both stress and pressing neurons within the NTS can spread to the lateral
energy balance regulation. PBN and suppress orexigenic eating (Roman et al., 2016).
Additionally, oxytocin signaling in the medial NTS leads to
Critical brain nuclei for homeostasis and food rewarding food intake inhibition, and this effect was partly mediated by
GI satiation signals within the NTS that cause an NTS in-
Compiled data have suggested that nuclei can be classified teraction with the PVN on food intake (Ong et al., 2015).
into the following three categories: (i) brain nuclei related to Parabrachial nucleus (PBN). PBN neurons within this re-
food intake, (ii) brain nuclei related to blood-glucose reg- gion mainly express the calcitonin gene-related peptide
ulation, and (iii) brain nuclei related to food-rewarding be- (CGRP), and activation of CGRP PBN neurons or their
havior. projections in the central nucleus of the amygdala (CeA) can
decrease feeding (Roman et al., 2016). Studies have con-
Brain nuclei related to food intake sistently shown that stimulation of GLP-1 receptors ex-
The ARC. The ARC is located beneath the third ventricle and pressed here reduced intake and decreased body weight in
near the median eminence. Studies have shown that the BBB rats, which suggests a role of PBN in the GLP-1 anorexic
here may not be completely impermeable. Therefore, the effect (Richard et al., 2014).
ARC may provide easy access to circulation and early de- PVN. The PVN is located next to the dorsal horn of the
tection of nutrients and hormones (Re´thelyi, 1984). Satur- third ventricle. The PVN neurons act as second-order neu-
able glucose transporter isoform-1 (GLUT1) mediates the rons in the melanocortinergic neuronal circuit innervated by
entry of glucose, which has a predominant distribution in the the POMC and NPY/AgRP neurons from the ARC (Cone,
BBB (Cardoso et al., 2010).The hypothalamic ARC ex- 2005). Several critical peptides released in the PVN, such as
8. . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ............................................ 8

oxytocin, thyrotropin-releasing hormone, corticotrophin- may be disturbed under pathological conditions. This effect
releasing hormone, and nesfatin, have roles in food intake. was demonstrated by a study that showed peripheral insulin
Additionally, the PVN is indispensable in mediating the resistance in db/db mice and fa/fa Zucker rats also led to GE
anorexic effect of GLP-1, which has a downstream effect on neurons resistant to insulin action (Cotero et al., 2010). GK is
the hypothalamic-ituitary-drenal axis or hypothalamic-itui- also a crucial component in GI sensing which is similar to
tary-hyroid axis related to energy balance (Katsurada et al., GE (Kang et al., 2006). Besides GK, GI neurons also depend
2014). on nNOS and AMPK. The VMN nNOS-GI mechanism is
clearly regulated by changes in AMPK activity, which results
Brain nuclei related to blood-glucose regulation in a series of activities, including chloride channel closure,
VMN. The VMN is located in the upper ARC, receives in- membrane depolarization, and intensified action potential
puts from the ARC, and extends its axons to the diverse frequency (Fioramonti et al., 2010). Studies have shown that
neurons related to energy metabolism, such as the PVN, LH, the role of GI cells is to counter-regulate hypoglycemia
NTS, or dorsomedial hypothalamic nucleus. through glucagon and epinephrine.
Neurons within the VMN synthesize steroidogenic factor 1 Medial amygdalar nucleus (MAN). The MAN is a limbic
and release brain-derived neurotrophic factor (BDNF) (Par- area that possesses another group of glucose-sensing neu-
ker and Schimmer, 1997; Unger et al., 2007). Selective de- rons. It is well acknowledged that hypoglycemia causes a
letion of the BDNF gene in the VMN of mice induced series of responses involving activation of the sympathetic
hyperphagia and obesity. Similarly, genetic function muta- nervous system and release of hormones, including gluca-
tion of the BDNF receptor tropomyosin receptor kinase B gon, epinephrine, and growth hormone (Cryer et al., 2003).
leads to a morbid intake effect and severe obesity both in Our study found that the glucose-sensing neurons in the
humans and rodents, which suggests that BDNF possesses a medial amygdala nucleus could be responsive to hypogly-
satiety trait (Yeo et al., 2004). cemia via UCN3 acting on the CRF2 receptor. Neurons of
Studies have shown that the VMH, which contains the MAN have a reciprocal link with the VMH in the hypotha-
ARC and VMN, contains a high density of glucose-sensing lamus, and UCN3 neuronal fibers have been shown to be the
neurons called “glucose-excited” (GE) or “glucose- main input to the VMH. One third of UCN3 neurons can be
inhibited” (GI) glucose-sensing neurons (Figure 3). GE activated in hypoglycemia to regulate blood sugar by in-
neurons depolarize in the presence of elevated plasma glu- itiating food intake. Hypoglycemic glucoprivic of MAN or
cose levels, and the activity of GI neurons is enhanced with VMH did not elicit a hypoglycemic counter-regulation hor-
decreasing glucose level (Dunn-Meynell et al., 2002). Stu- monal response immediately; however, glucoprivation
dies have shown that GE neurons express peptides that exert combined with mild to moderate systemic hypoglycemia can
an anorexic effect, whereas GI neurons in this area release cause a counter-regulation hormonal response in normal
orexic peptides, which counter-regulate hypoglycemia mice. This finding suggests that MAN or VMH has a central
(Murphy et al., 2009). role in initiation of a hypoglycemic counter-regulation re-
AMPK has been shown to involve both GE and GI activity sponse (Zhou et al., 2010).
(Routh et al., 2014). As a type of cellular fuel sensor, acti-
vated AMPK due to the elevated ratio of ADP/ATP or AMP/ Brain nuclei related to food-rewarding behavior
ATP is regarded as the key component in the process of The primary brain nuclei related to food rewarding constitute
glucose sensing. AMPK promotes feeding behavior by in- the DA system from the midbrain to the forebrain. Two
creasing energy intake and glucose production, and reduces noticeable DA pathways exist: one from the VTA to the
thermogenesis. Particular hormones, such as adiponectin, ventral striatum, termed the “Satiety” pathway related to
ghrelin, leptin, and GLP-1, have been shown to interact with satiety in food-rewarding behavior and another from the SN
AMPK to mediate energy regulation (Huynh et al., 2016). to the dorsal striatum, termed the “Hedonistic” pathway for
The mechanism underlying GE neuronal function is si- promoting rewarding behavior in addition to its previous role
milar to that of β-cells within the pancreatic islet. GK is the in the motor regulation (Figure 4). It has recently been de-
crucial component in GE sensing (De Backer et al., 2016). termined that the dorsal and ventral striatum can receive
When transported into the neurons via glucose transporter 2 discrete nutrient information from intestinal segments. Stu-
(GLUT2), glucose is phosphorylated by glucokinase and dies have found that the duodenum can convey a heightened
leads to a series of downstream effects. GLUT2 has been sense of sweet to the dorsal striatum, which leads to in-
shown to play the vital role of glucokinase in glucose sen- creased intake of sugars, whereas jejunum signals commu-
sing. If GLUT2-mediated glucose detection is inhibited, food nicate with the ventral striatum that has a role in food satiety
intake will be augmented (Stolarczyk et al., 2010). The role signaling to terminate a meal.
of GE neurons in reaction to changed glucose levels is to Projection from the VTA to the ventral striatum. The VTA
stress the hyperglycemia under regulation of insulin, which lies along the midline of the midbrain near the SN and red
9. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ............................................ 9

+
Figure 3 The activation of GE neuron involves elevated ATP/ADP ratio due to increased glucose consumption which results in the K ATP channel’s
closure and depolarization. However, the GI neurons are activated via either metabolism dependent or independent way in a low glucose background.
Metabolism dependent GI neuron is mainly a nNOS-GI neuron due to the nNOS which catalyze the NO production being detected in this type of neuron. The

activation requires AMPK and closure of Cl channel, whereas independent GI mainly express Orexin or AgRP, and may be evoked by glucose structure itself
rather than its metabolite. This process may require K2P and Twik-related acid-sensitive potassium-like (TASK) 1/3 channels. SGLT, sodium-dependent
glucose transporters; GLUT4, glucose transporter type 4; TCA, tricarboxylic acid cycle; GK, glucose kinase; AMPK, AMP-activated protein kinase; NO,
nitric oxide; nNOS, nitric oxide synthase 1 (neuronal); sGC, soluble guanylate cyclase; K2P channel, two pore potassium channel; TASK1/3 channel, TWIK-
related acid sensitive potassium channel1/3; TWIK, tandem of P domains in a weak inwardly rectifying potassium channel.

nucleus and is rich in dopamine neurons. The VTA is widely from distal intestines, project their axons to the ventral
implicated in drug and natural reward addiction. Recent striatum, and terminate at the neurons within the ventral
studies have shown that VTA neurons can receive signals striatum, which may lead to enhancement of satiety during a
10 . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ........................................... 10

centrating hormone, orexins/hypocretins, and neurotensin-


expressing cells (Brown et al., 2015). The last one possesses
anorexigenic attributes that have been shown to involve the
major neurotensin receptor (NtsR) type 1 (Feifel et al.,
2010). Binding to NtsR-1 facilitates the inhibitory effect on
feeding induced by leptin (Sahu et al., 2001); however, a
deficiency in NtsR-1 in mice may affect the response to
leptin, which suggests that leptin acts on the LH to affect
energy balance regulation (Kim et al., 2008).

Overlap and interaction between homeostatic and hedonic


nuclei
Although we classified three categories of major critical
brain nuclei and areas, these structures are not completely
independent from the others given the basis of multiple
complicated interconnections among them and simultaneous
Figure 4 There are two major DA pathways involved in food rewarding functions. Evidence suggests that these two systems, in fact,
manner, one originates from the VTA, and targets the ventral striatum,
which is related to feeding satiety to end the eating; another pathway interact with each other (Williams, 2014). First, the GLP-1
derives from the SN and terminates on the dorsal striatum, which is a projections from the NTS to the NAc and VTA have a role
hedonistic pathway promoting food intake. through which meal-related gut signals may influence pa-
latability, motivation for food, and meal size. Second, orexin
meal to terminate further intake. projections from the LHA to the NTS and AP act as a
Projection from the SN to the dorsal striatum. Recent pathway through which cues that predict food rewarding
findings showed that the dorsal striatum may play a role in may respond to increased motivation for food and suppress
food-rewarding behavior. Nutrients in the duodenum may satiation. Moreover, studies have also revealed that leptin-
convey “hedonistic or enjoyable” signals to the neurons in sensitive neurons (not the GLP-1 neurons) in the NTS in-
the SN, which interacts with the dorsal striatum to promote fluence motivation and food-seeking behavior. Infusion of a
further intake. Duodenal-jejunal bypass surgery can curb ventricle subthreshold dose of leptin into the medial NTS
sugar cravings in mice by decreasing sugar intake-induced suppressed responses to sucrose and reversed high-fat food-
dopamine release specifically in the dorsal striatum. In ad- conditioned preferences (Kanoski et al., 2014). Similar to
dition to the energy restriction foregut or hindgut hypothesis GLP-1 neurons, leptin-sensitive neurons in the NTS respond
of this surgery, Han et al. stated that recurrent exposure to to meal-related GI signals, such as gastric distention (Huo et
sugar may postpone the satiety of sugar intake, whereas al., 2007). Finally, an increasing body of knowledge suggests
bariatric surgery may perturb this effect via blocking of the that PVN oxytocin neurons simultaneously affect both
link to the dorsal striatum. They found that duodenal glucose homeostatic and hedonic systems. Oxytocin neurons can be
infusions induced significantly greater dopamine release in activated by leptin and CCK and have been proposed to
the dorsal striatum than did equivalent jejunal infusions, and mediate at least some aspects of GI nutrient-induced satia-
activation of dopamine D1r expression neurons by in- tion (Olszewski et al., 2010). These neurons also project to
tracerebral laser stimulation can mimic the glucose influx in the VTA (Sofroniew, 1983; Vaccari et al., 1998) In conclu-
the duodenum, which can reverse the duodenal-jejunal by- sion, it is suggested that the circuitry of energy homeostasis
pass surgery effect on energy metabolism (Figure 4) (Han et and that of food rewarding are, in fact, mutually affected.
al., 2016).
LH. The LH is in the hypothalamic region and serves to
incorporate and process metabolic and reward-related in- ENERGY AND METABOLISM REGULATION
formation. Some LH neurons rightly link with midbrain DA BY PERIPHERAL BIO-SUBSTANCES
neurons that act on the forebrain. The studies that adopt
lesion, stimulation, and anatomical methodologies have re- Peripheral hormone regulation
cognized that the LH has a powerful influence on the desire Peptides released from the gut send energy signals to the
for eating, drinking, and moving. brain. These signals are delivered mainly through activation
The LHA is composed of a narrow line of tissue upon the of the afferent vagus nerve, which in turn can inform the
whole length of the rostral-caudal hypothalamus. Three main brainstem and hypothalamus. On the other hand, peptides
subpopulations of LHA neurons exist that coordinate dis- from the gut or adipose tissue can travel by circulation and
crete energy cues and behavioral responses: melanin-con- infiltrate the BBB through the corresponding transporters in
11 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ........................................... 11

the hypothalamus or near the brainstem (Table 2; Figure 5). NH2, and the latter is the major active isoform in the cir-
culation. GLP-1 can suppress food intake via activation
CCK neurons within the hypothalamus, brainstem, and rewarding
CCK is a type of intestinal peptide hormone, and its structure pathways as well as via activation of the vagal afferent
is very similar to that of gastrin. It is secreted by the I-cells in neurons and other involved mechanisms, such as delaying of
the first segment of the intestine (Moran, 2000). CCK re- gastric emptying.
ceptors are G protein-coupled receptors, termed CCK-A or
CCKB. CCK can also act as a neurotransmitter in CNS and Ghrelin
mostly exerts a satiation effect via the CCK-A receptor Ghrelin is encoded by the preproghrelin gene from the GI
(Lieverse et al., 1995; Beglinger et al., 2001). Administration tract. GOAT is the enzyme responsible for ghrelin acylation
of selective agonists and antagonists has proved the anor- and has a role in ghrelin’s physiological activity (Yang et al.,
exigenic trait of CCK; however, the anorexigenic effect does 2008). Ghrelin can induce hunger and weight gain in both
not have a long-term effect on caloric intake in rodents (West rodents and humans when it acts on vagal afferent neurons,
et al., 1984). which in turn affects CNS areas related to energy home-
ostasis (Date et al., 2002). The orexigenic effect centrally
PYY involves activation of GHSR1a receptors in the ARC of the
PYY is released from L cells of the intestine. It is a member hypothalamus, which affects NPY/AgRP neuronal activity
of the pancreatic polypeptide family. PYY3–36 is the active (Kamegai et al., 2001). Ghrelin can also activate food reward
isoform in circulation postprandially (Grandt et al., 1994). areas in response to food stimuli, as reflected by fMRI,
Activation of the Y2 receptor in the CNS has an impact on which suggests a role in the reward mechanism (Kroemer et
energy regulation (Batterham et al., 2002). A defect in PYY al., 2013; Goldstone et al., 2014). The critical role of ghrelin
signaling may be implicated in the development of obesity, is to prevent hypoglycemia because ablation of ghrelin-
and its concentration in circulation may become normalized producing cells in adults induce profound hypoglycemia
after bariatric surgery (le Roux et al., 2006). The activation after prolonged caloric restriction (McFarlane et al., 2014).
site of PYY covers the ARC, brainstem, and corticolimbic
pathway, such as the orbitofrontal cortex, which can be ac- Insulin
tivated to make food less rewarding (D’Agostino and Small, Insulin enters the brain from the circulation via the BBB to
2012). activate hypothalamic neurons, which induces an anorexi-
genic effect. By binding to receptors highly expressed in
GLP-1 POMC/CART and NPY/AgRP neurons, it decreases NPY
GLP-1 is the expression product of the proglucagon gene in and stimulates POMC expression (Porte et al., 2005).
L cells of the ileum and proximal colon or hindbrain in the Otherwise, insulin was determined to affect the VTA DA
CNS (Drucker, 2006; Trapp and Cork, 2015), and gut- concentration via upregulation of DA transporters to depress
derived GLP-1 is secreted postprandially. The two biologi- DA reuptake, which suggests that insulin has a role in reward
cally active isoforms are GLP-1 (7–37) and GLP-1 (7–36) regulation (Mebel et al., 2012).

Table 2 Summary on major peripheral hormone involved in energy regulation


Categories Hormone Derivation Effective form Target Receptor Pathway action Food intake
Gut-peptide Pancreas, hypothalamus, G13-RhoA-PLD/
hormone CCK I-cell, duodenum CCK-58 vagus nerve CCK-A/B r β-arrestin Anorexigenic

PYY L-cell, Ileum, colon PYY3–36 Pancreas, Y2r PI3K/PLC-ERK Anorexigenic


gastrointestine, brain
GLP-1 (7–36) amide, Pancreas, brain, cAMP-PKA-AMPK/
GLP-1 L-cell, Ileum, colon GLP-1 (7–37) vagus nerve GLP-1r p44 42MAPK Anorexigenic

Ghrelin Ghrelinergic cells, Ghrelin Hypothalamus, vagus GHS-r1a mTORC1-S6K1 Orexigenic


gastrointestine nerve, mesolimbic
Pancreatic
hormone PP PPcell, pancreas PP Brain, vagus nerve Y4/5r PI3K/PLC-ERK Anorexigenic

IRS-PI3K-AKT-FoxO1/
Insulin β-cell, pancreas Insulin Whole body IR mTOR Anorexigenic

Adipose-peptide Leptin Adipose cell, adipose Leptin Hypothalamus, adipose OB-R PI3K-AKT-FoxO1/ Anorexigenic
hormone tissue (WAT) tissue JAK-STAT3
Hypothalamus, adipose
Adiponectin Adipose cell, WAT Adiponectin tissue, muscle Adipo-R AMPK/PPARα Anorexigenic
12 . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ........................................... 12

Figure 5 The illustration above displays the interaction between gut, adipose tissue and brain in rodents. The signals derived from adipose tissue or GI tract
either have impact upon hypothalamus via infiltrating a leaky BBB, or link the NTS in the brainstem via acting on receptors within vagal afferents. These
signals induce activities of nuclei such as ARC and NTS, which may in turn affect second order neurons within PVN or VMN; other nuclei related to reward
such as VTA or NAc may also be involved in. GLP-1, glucagon like peptide-1; PYY, peptide YY; PP, pancreatic peptide; GI tract, gastrointestinal tract.

Leptin mauchi et al., 2014). There are two types of receptors: adi-
Leptin is derived from adipose tissue. It is also known as the ponectin receptor 1 (AdipoR1) and AdipoR2 (Yamauchi et
“satiety hormone”. As an anorexigenic hormone in circula- al., 2007). The metabolic effect of liver AdipoR1 may be
tion, its concentration fluctuates with feeding and increases involved in activating AMPK, whereas AdipoR2 is involved
postprandially to suppress further intake (Leibowitz et al., in activation of PPARα, which leads to increased insulin
2006). Moreover, the leptin concentration in the body re- sensitivity (Yamauchi et al., 2002; Yamauchi et al., 2003).
flects the fuel status because a higher concentration of leptin There is evidence suggesting adiponectin activity in the
has been found under obese conditions, which suggests that brain, such as in the ARC and PVN (Hoyda et al., 2009;
leptin resistance exists. Activation of the leptin receptors Psilopanagioti et al., 2009; Sun et al., 2016).
results in increased POMC transcription and suppressed
transcription of AgRP (Villanueva and Myers, 2008; Morris Nutrient regulation
and Rui, 2009).
Nutrient-related signals may have an effect on CNS. Besides
Adiponectin glucose, lactate, referred to as a metabolic waste previously,
Adiponectin is also derived from adipose tissues; its role in has also been shown to regulate metabolism, such as by
energy regulation is to promote energy consumption and its promoting insulin secretion, and mostly acts by signaling via
signaling has been determined to improve insulin sensitivity, the CNS (Sola-Penna, 2008). It has been proposed that the
reduce insulin resistance, and reduce atherosclerosis (Ya- enhanced hypothalamic lactate metabolism alters signaling
13 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ........................................... 13

to the brain and to the whole body, which results in decreased CRITICAL ORGANS OR TISSUES IN
liver glucose production (Chari et al., 2008). Free fatty acids PERIPHERAL METABOLIC REGULATION
(FFAs) bind to FFA receptors and can affect metabolism
(Hara et al., 2014). Infusion of FFA centrally affects insulin Role of the liver in energy balance regulation
secretion and insulin sensitivity via altering the activity of With the advent of an increased incidence of nonalcoholic
GE or GI within the VMH (Le Foll et al., 2013). A recent fatty liver disease (NAFLD), it is time to re-examine the role
study determined the signaling pathway of ketone body β- of the liver in energy metabolism. Although there are cur-
hydroxybutyrate, which is involved in the starvation re- rently various attribution theories on this disease, such as
sponse during nutrient deprivation and ensures energy mitochondrial hyperfunction, it is accepted that the patho-
homeostasis (Rojas-Morales et al., 2016). The branched physiology is related to energy dysregulation in the liver. In a
chain amino acid (BCAA) L-leucine, suppresses food intake physiological state, the liver acts as a hub involved in mac-
when administered into the third ventricle. This effect may ronutrients (e.g., glucose, lipid, protein) and small molecules
involve action on hypothalamic mammalian target of rapa- (e.g., vitamin, peptide, iron) metabolism essential for
mycin (Cota et al., 2006). Studies have also shown that homeostasis and affects various metabolic tissues, including
elevated BCAA concentrations in plasma are associated with skeletal muscle and adipose tissue. Food and small mole-
obesity and insulin resistance (Newgard et al., 2009; Adams, cules can be digested and taken up by GI cells and absorbed
2011; Adeva et al., 2012). This finding suggests that BCAA into the circulation of the liver, which facilitates further
acts as a satiety signal. β-Aminoisobutyric acid, which is a processing. The liver acts as a producer of chemicals that can
small-molecule metabolite of BCAA, has been shown to convert multiple nutrients or metabolites into substances that
increase browning of white WAT in response to physical benefit the body’s discrete metabolic needs. For instance,
activity and is inversely associated with cardiometabolic glucose can be synthesized into glycogen or other substances
risks (Roberts et al., 2014). and stored by the liver in the presence of insulin after a meal,
whereas in the starvation state, it can be produced increas-
Gut microbiota ingly through the glycogenolysis or gluconeogenesis pro-
cesses de novo. These processes are affected by regulators,
such as hormone milieus (e.g., insulin, glucagon, gluco-
Multiple studies have identified the role of gut microbiota. corticoids) and metabolic states (e.g., post-meal, fasted, ex-
Short-chain fatty acid (SCFA) metabolites generated by gut ercise), which have effects on the activity of critical
microbiota can regulate gut hormones, such as CCK, GLP-1, enzymes, such as phosphoenolpyruvate carboxylase cata-
PYY, and leptin. Food rich in plant fibers can influence the lysis in gluconeogenesis.
microbiota, and their metabolites, such as acetate, propio- The liver also communicates with the CNS, skeletal
nate, and butyrate, have an effect on host energy homeostasis muscles, adipose tissue, and the gut. First, the CNS directly
(Schwiertz et al., 2010). These SCFAs act as key signaling innervates the liver and modulates energy metabolism via
molecules, with activation of both FFAR2 and FFAR3 to either the sympathetic or parasympathetic nervous system.
modulate host adiposity or GLP-1 secretion (Samuel et al., The sympathetic system generally increases hepatic glucose
2008; Tolhurst et al., 2012). Butyrate activates intestinal production by promoting glycogenolysis and mobilizing the
gluconeogenesis (IGN) gene expression through a cAMP- other forms of fuel for gluconeogenesis, whereas the para-
dependent mechanism, whereas propionate, itself a substrate sympathetic system counteracts it. Multiple hormones can
of IGN, activates IGN gene expression via the gut-brain indirectly influence liver activity via activation of the re-
neural circuit involving FFAR3 (De Vadder et al., 2014). ceptors of hypothalamus cells, such as insulin, which sti-
Studies have found a distinction between the microbiota of mulates the PI 3-kinase/Akt pathway in the brain (Ramnanan
normal individuals and obese individuals or patients with et al., 2011). Second, skeletal muscles consume energy and
T2DM (Qin et al., 2012; Karlsson et al., 2013). Additionally, produce lactate and amino acids, which are eventually de-
these pre-clinical studies also proved that using fecal trans- livered to the liver, thus providing the gluconeogenic sub-
plantation of modifying rodents’ microbiota may improve strates for hepatocytes to synthesize glucose that can be
metabolism (de Clercq et al., 2017). Transfer of intestinal utilized by muscle. A study also revealed that IL-6 can be
microbiota from obese mice resulted in significantly greater generated by skeletal muscle as a myokine that has an effect
adiposity in recipients than did transfer of microbiota from on the liver to increase hepatic glucose production during
lean donors (Bäckhed et al., 2007). Microbiota-related dys- exercise (Febbraio et al., 2004). Third, crosstalk between the
function creates a predisposition toward a range of disorders, liver and adipose tissue has been shown by the finding that
including metabolic syndrome, neurodevelopmental dis- adipocytokines, such as adiponectin generated by adipo-
orders, autoimmune diseases, and allergic diseases (Shana- cytes, can influence hepatic metabolism and fibroblast
han, 2012). growth factor 21 (a hormone mainly derived from the liver in
14 . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ........................................... 14

the fasted state), can affect the adipocyte response to lipo- ROLE OF SKELETAL MUSCLE IN PERIPH-
lysis, and release adiponectin (Xu et al., 2003; Lin et al., ERAL METABOLISM REGULATION
2013). Finally, given that the liver port vein system linked
the liver and the gut, this connection allows for nutrients, gut As the major energy-consuming motor organ in the body,
hormones, and metabolites (especially derived from gut skeletal muscle can also release hormones or small proteins
microbiota) to access the liver and thus directly regulate the that affect systemic metabolism. These substances are known
liver metabolism (Henao-Mejia et al., 2012). On the other as myokines, which are similar to but distinct from adipo-
hand, the gut-brain axis can be an indirect method of gut- cytokines from adipose tissue. These proteins exert bene-
liver cross talk that requires an intact vagal nerve pathway to ficial effects on peripheral and remote organs. The TGF-β
modulate liver metabolism. In conclusion, the liver is an superfamily and related factors released by the muscles un-
essential nutrient metabolism producer and is metabolically der exercise-like conditions are myokines, such as myostatin
linked with both the CNS and multiple insulin-sensitive and activins. Myostatin and activins act to limit muscle mass
peripheral organs or tissues, such as skeletal muscle, adipose and inhibition that can induce significant muscle growth
tissue, and gut. Therefore, in addition to pharmacological (Lee et al., 2005; Kollias and McDermott, 2008). Another
targeting of intracellular key molecules, rearrangement of the myokine, follistatin, which does not belong to the TGF-β
proper links of these organs and tissues may be important for superfamily, acts to inhibit myostatin and activins, similar to
curing obesity, insulin resistance, NAFLD, and T2DM, in irisin.
future therapeutic strategies. In general, myostatin, leukemia inhibitory factor, and IL-6
and IL-7 are involved in muscle hypertrophy and myogen-
BAT in peripheral metabolic regulation esis, whereas BDNF and IL-6 are involved in AMPK-
mediated fat oxidation. IL-6 also appears to have systemic
effects on the liver, adipose tissue, and the immune system
Peripheral BAT thermogenesis regulation is dependent on and mediates crosstalk between intestinal L cells and pan-
sympathetic outflow to it. The adipocytes accumulated in creatic islets (Pedersen and Febbraio, 2012). Chronic ex-
BAT provide the binding receptors involved in metabolism ercise as a lifestyle intervention can improve overall
(the β3-adrenergic receptors). After binding to receptors, metabolism and benefit the health of individuals for not only
adrenaline triggers cyclic adenosine monophosphate, which energy expenditure but also for adjusting systemic functions.
activates mitochondrial uncoupling protein-1 (UCP-1) re-
sulting in greater thermogenesis. The preoptic area (POA)
has been shown to directly affect sympathetic premotor CONCLUSION
neurons derived from the rostral raphe pallidus to regulate
sympathetic outflow to the BAT (Morrison, 2004). Other- In this review, we initially investigated metabolic regulation
wise, POA neurons may mediate thermoregulatory responses of CNS not only as a homeostatic system, but also as a
via the projection to DMN neurons (Yoshida et al., 2009). reward system that maintains energy balance both metabo-
Sympathetic outflow to the BAT can be adjusted by certain lically and motivationally. We described the anorexigenic
nutrient signals or hormones, such as glucose or GLP-1 and orexigenic mechanisms and regulation of food intake by
(Rahmouni et al., 2004; Lockie et al., 2012). Administration hypothalamic POMC/CART and NPY/AgRP neurons.
of an MC3R/MC4R agonist centrally evokes BAT through Moreover, we summarized the role of essential neuro-
sympathetic outflow, which suggests that the melanocortin transmitters coordinated within the CNS. Critical brain re-
system can regulate BAT thermogenesis (Brito et al., 2007). gions or nuclei operate spatially and temporally and are
Brown-like fat adipocytes, also named “beige” or “brite” responsible for the three kinds of central regulation of energy
adipocytes, are located across the inguinal subcutaneous area balance. We then reviewed energy regulation by peripheral
of rodents as well as the supraclavicular, pericardial, para- mechanisms. First, the crosstalk between the gut or adipose
aortic, and suprarenal areas as well as areas surrounding the tissue and the brain involves a variety of signals derived from
trachea, pancreas, or kidney in humans. Although UCP-1 is the GI tract and adipocytes that respectively act as humor
weakly expressed in beige adipocytes under basal conditions, signals to inform the CNS or to affect the metabolism of
higher expression can be induced under extreme conditions, peripheral organs. In addition, nutrients or metabolites act
such as cold temperatures (van Marken Lichtenbelt et al., synergistically with the humor signals to affect metabolic
2009). The browning of WAT in a rodent study was shown to control. On the other hand, gut microbiota with their SCFA
encourage energy consumption and improve diet-induced metabolites have been determined to influence intestinal
obesity (Seale et al., 2011). Therefore, regulation of WAT activity and improve metabolism, and their constitutional
browning is an ideal methodology for obesity and metabo- alteration has been associated with various diseases. Finally,
lism dysregulation treatment. regulation by the liver, BAT and skeletal muscle demon-
15 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ........................................... 15

strates their comprehensive role and interactions with other Baver, S.B., Hope, K., Guyot, S., Bjorbaek, C., Kaczorowski, C., and
O’Connell, K.M.S. (2014). Leptin modulates the intrinsic excitability of
parts of the body. Accordingly, a living organism’s energy AgRP/NPY neurons in the arcuate nucleus of the hypothalamus. J
balance requires accurate and precise control at the mole- Neurosci 34, 5486–5496.
cular, cellular, and organ levels as well as the systemic level Beck, B. (2006). Neuropeptide Y in normal eating and in genetic and
through nervous and humor system mechanisms. Drug tar- dietary-induced obesity. Philos Trans R Soc Lond B Biol Sci 361, 1159–
1185.
gets in these areas may lead to disruption of unhealthy me- Beglinger, C., Degen, L., Matzinger, D., D’Amato, M., and Drewe, J.
tabolic activity or prevent development of T2DM or morbid (2001). Loxiglumide, a CCK-A receptor antagonist, stimulates calorie
obesity, which are increasingly prevalent in modern society. intake and hunger feelings in humans. Am J Physiol Regul Integr Comp
Physiol 280, R1149–R1154.
Berridge, K.C., Ho, C.Y., Richard, J.M., and DiFeliceantonio, A.G. (2010).
Compliance and ethics The author(s) declare that they have no conflict The tempted brain eats: pleasure and desire circuits in obesity and
of interest. eating disorders. Brain Res 1350, 43–64.
Bisaga, A., Danysz, W., and Foltin, R.W. (2008). Antagonism of glutam-
atergic NMDA and mGluR5 receptors decreases consumption of food
Acknowledgements This work was supported by the National Natural
in baboon model of binge-eating disorder. Eur Neuropsychopharmacol
Science Foundation of China (81370932), the United States MERCK IISP 18, 794–802.
Fund (40313, 40309), Outstanding Leaders Training Program of Pudong Björklund, A., and Dunnett, S.B. (2007). Dopamine neuron systems in the
Health Bureau of Shanghai (PWR12014-06), Integrative Medicine special brain: an update. Trends Neurosci 30, 194–202.
fund of Shanghai Municipal Health Planning Committee (ZHYY-ZXYJHZX- Bojanowska, E., and Ciosek, J. (2016). Can we selectively reduce appetite
2-201712) and the Key Studies (Special) Department Fund of the Pudong for energy-dense foods? An overview of pharmacological strategies for
New Area Health Planning Commission (PWZzk2017-03). modification of food preference behavior. Curr Neuropharmacol 14,
118–142.
Adams, S.H. (2011). Emerging perspectives on essential amino acid met- Borgland, S.L., Chang, S.J., Bowers, M.S., Thompson, J.L., Vittoz, N.,
abolism in obesity and the insulin-resistant state. Adv Nutr 2, 445–456. Floresco, S.B., Chou, J., Chen, B.T., and Bonci, A. (2009). Orexin A/
Adamska, E., Ostrowska, L., Gorska, M., and Kretowski, A. (2014). The hypocretin-1 selectively promotes motivation for positive reinforcers. J
role of gastrointestinal hormones in the pathogenesis of obesity and type Neurosci 29, 11215–11225.
2 diabetes. Prz Gastroenterol 9, 69–76. Bradbury, M.J., Campbell, U., Giracello, D., Chapman, D., King, C., Te-
Adan, R.A.H., Vanderschuren, L.J.M.J., and la Fleur, S.E. (2008). Anti- hrani, L., Cosford, N.D.P., Anderson, J., Varney, M.A., and Strack, A.
obesity drugs and neural circuits of feeding. Trends Pharmacol Sci 29, M. (2005). Metabotropic glutamate receptor mGlu5 is a mediator of
208–217. appetite and energy balance in rats and mice. J Pharmacol Exp Thera-
Adeva, M.M., Calviño, J., Souto, G., and Donapetry, C. (2012). Insulin peut 313, 395–402.
resistance and the metabolism of branched-chain amino acids in hum- Brito, M.N., Brito, N.A., Baro, D.J., Song, C.K., and Bartness, T.J. (2007).
ans. Amino Acids 43, 171–181. Differential activation of the sympathetic innervation of adipose tissues
Allen, S.S., Hatsukami, D., Brintnell, D.M., and Bade, T. (2005). Effect of by melanocortin receptor stimulation. Endocrinology 148, 5339–5347.
nicotine replacement therapy on post-cessation weight gain and nutrient Brown, J.A., Woodworth, H.L., and Leinninger, G.M. (2015). To ingest or
intake: a randomized controlled trial of postmenopausal female smok- rest? Specialized roles of lateral hypothalamic area neurons in coordi-
ers. Addict Behav 30, 1273–1280. nating energy balance. Front Syst Neurosci 9, 9.
Anderson, E.J.P., Çakir, I., Carrington, S.J., Cone, R.D., Ghamari-Langr- Brownley, K.A., Peat, C.M., La Via, M., and Bulik, C.M. (2015). Pharm-
oudi, M., Gillyard, T., Gimenez, L.E., and Litt, M.J. (2016). 60 YEARS acological approaches to the management of binge eating disorder.
OF POMC: regulation of feeding and energy homeostasis by α-MSH. J Drugs 75, 9–32.
Mol Endocrinol 56, T157–T174. Cansell, C., Denis, R.G., Joly-Amado, A., Castel, J., and Luquet, S. (2012).
Arima, H., and Oiso, Y. (2010). Positive effect of baclofen on body weight Arcuate AgRP neurons and the regulation of energy balance. Front
reduction in obese subjects: a pilot study. Intern Med 49, 2043–2047. Endocrinol (Lausanne) 3, 169.
Arnone, M., Maruani, J., Chaperon, F., Thiébot, M.H., Poncelet, M., So- Cardoso, F.L., Brites, D., and Brito, M.A. (2010). Looking at the blood-
ubrié, P., and Fur, G.L. (1997). Selective inhibition of sucrose and brain barrier: molecular anatomy and possible investigation approaches.
ethanol intake by SR 141716, an antagonist of central cannabinoid Brain Res Rev 64, 328–363.
(CB1) receptors. Psychopharmacology 132, 104–106. Challis, B.G., Yeo, G.S., Farooqi, I.S., Luan, J., Aminian, S., Halsall, D.J.,
Audrain-McGovern, J., and Benowitz, N.L. (2011). Cigarette smoking, n- Keogh, J.M., Wareham, N.J., and O’Rahilly, S. (2000). The CART gene
icotine, and body weight. Clin Pharmacol Ther 90, 164–168. and human obesity: mutational analysis and population genetics. Dia-
Bäckhed, F., Manchester, J.K., Semenkovich, C.F., and Gordon, J.I. (2007). betes 49, 872–875.
Mechanisms underlying the resistance to diet-induced obesity in germ- Chari, M., Lam, C.K.L., Wang, P.Y.T., and Lam, T.K.T. (2008). Activation
free mice. Proc Natl Acad Sci USA 104, 979–984. of central lactate metabolism lowers glucose production in uncontrolled
Baggio, L.L., and Drucker, D.J. (2014). Glucagon-like peptide-1 receptors diabetes and diet-induced insulin resistance. Diabetes 57, 836–840.
in the brain: controlling food intake and body weight. J Clin Invest 124, Chiolero, A., Faeh, D., Paccaud, F., and Cornuz, J. (2008). Consequences
4223–4226. of smoking for body weight, body fat distribution, and insulin resista-
Baik, J.H. (2013). Dopamine signaling in food addiction: role of dopamine nce. Am J Clin Nutr 87, 801–809.
D2 receptors. BMB Rep 46, 519–526. Chronwall, B.M. (1985). Anatomy and physiology of the neuroendocrine
Bakshi, V.P., Newman, S.M., Smith-Roe, S., Jochman, K.A., and Kalin, N. arcuate nucleus. Peptides 6 Suppl 2, 1–11.
H. (2007). Stimulation of lateral septum CRF2 receptors promotes an- Cone, R.D. (2005). Anatomy and regulation of the central melanocortin
orexia and stress-like behaviors: functional homology to CRF1 recep- system. Nat Neurosci 8, 571–578.
tors in basolateral amygdala. J Neurosci 27, 10568–10577. Cooper, S.J., and Al-Naser, H.A. (2006). Dopaminergic control of food
Batterham, R.L., Cowley, M.A., Small, C.J., Herzog, H., Cohen, M.A., choice: contrasting effects of SKF 38393 and quinpirole on high-pala-
Dakin, C.L., Wren, A.M., Brynes, A.E., Low, M.J., Ghatei, M.A., et al. tability food preference in the rat. Neuropharmacology 50, 953–963.
(2002). Gut hormone PYY3-36 physiologically inhibits food intake. Cota, D., Proulx, K., Smith, K.A.B., Kozma, S.C., Thomas, G., Woods, S.
Nature 418, 650–654. C., and Seeley, R.J. (2006). Hypothalamic mTOR signaling regulates
16 . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ........................................... 16

food intake. Science 312, 927–930. Nutr 97, 15–22.


Cotero, V.E., Zhang, B.B., and Routh, V.H. (2010). The response of glu- Goldstone, A.P., Prechtl, C.G., Scholtz, S., Miras, A.D., Chhina, N., Dur-
cose-excited neurones in the ventromedial hypothalamus to decreased ighel, G., Deliran, S.S., Beckmann, C., Ghatei, M.A., Ashby, D.R., et al.
glucose is enhanced in a murine model of type 2 diabetes mellitus. J (2014). Ghrelin mimics fasting to enhance human hedonic, orbitofrontal
Neuroendocrinol 22, 65–74. cortex, and hippocampal responses to food. Am J Clin Nutr 99, 1319–
Covelo, I.R., Patel, Z.I., Luviano, J.A., Stratford, T.R., and Wirtshafter, D. 1330.
(2014). Manipulation of GABA in the ventral pallidum, but not the Grandt, D., Schimiczek, M., Beglinger, C., Layer, P., Goebell, H., Eysse-
nucleus accumbens, induces intense, preferential, fat consumption in lein, V.E., and Reeve Jr., J.R. (1994). Two molecular forms of Peptide
rats. Behav Brain Res 270, 316–325. YY (PYY) are abundant in human blood: characterization of a radioi-
Cowley, M.A., Smart, J.L., Rubinstein, M., Cerdán, M.G., Diano, S., Ho- mmunoassay recognizing PYY 1–36 and PYY 3–36. Regul Pept 51,
rvath, T.L., Cone, R.D., and Low, M.J. (2001). Leptin activates anor- 151–159.
exigenic POMC neurons through a neural network in the arcuate Grill, H.J., and Hayes, M.R. (2012). Hindbrain neurons as an essential hub
nucleus. Nature 411, 480–484. in the neuroanatomically distributed control of energy balance. Cell
Cryer, P.E., Davis, S.N., and Shamoon, H. (2003). Hypoglycemia in dia- Metab 16, 296–309.
betes. Diabetes Care 26, 1902–1912. Han, W., Tellez, L.A., Niu, J., Medina, S., Ferreira, T.L., Zhang, X., Su, J.,
D’Agostino, A.E. and Small, D.M. (2012). Neuroimaging the interaction of Tong, J., Schwartz, G.J., van den Pol, A., et al. (2016). Striatal dopa-
mind and metabolism in humans. Mol Metab 1(1–2), 10–20. mine links gastrointestinal rerouting to altered sweet appetite. Cell M-
Date, Y., Murakami, N., Toshinai, K., Matsukura, S., Niijima, A., Matsuo, etab 23, 103–112.
H., Kangawa, K., and Nakazato, M. (2002). The role of the gastric Hara, T., Kashihara, D., Ichimura, A., Kimura, I., Tsujimoto, G., and
afferent vagal nerve in ghrelin-induced feeding and growth hormone Hirasawa, A. (2014). Role of free fatty acid receptors in the regulation
secretion in rats. Gastroenterology 123, 1120–1128. of energy metabolism. Biochim Biophys Acta 1841, 1292–1300.
De Backer, I., Hussain, S.S., Bloom, S.R., and Gardiner, J.V. (2016). Harada, Y., Takayama, K., Ro, S., Ochiai, M., Noguchi, M., Iizuka, S.,
Insights into the role of neuronal glucokinase. Am J Physiol Endocrinol Hattori, T., and Yakabi, K. (2014). Urocortin1-induced anorexia is re-
Metab 311, E42–E55. gulated by activation of the serotonin 2C receptor in the brain. Peptides
de Clercq, N., Frissen, M.N., Groen, A.K., and Nieuwdorp, M. (2017). Gut 51, 139–144.
microbiota and the gut-brain axis: new insights in the pathophysiology Hartfield, A.W., Moore, N.A., and Clifton, P.G. (2003). Serotonergic and
of metabolic syndrome. Psychosom Med 79, 874–879. histaminergic mechanisms involved in intralipid drinking? Pharmacol
De Vadder, F., Kovatcheva-Datchary, P., Goncalves, D., Vinera, J., Zitoun, Biochem Behav 76, 251–258.
C., Duchampt, A., Bäckhed, F., and Mithieux, G. (2014). Microbiota- Hayes, D.J., and Greenshaw, A.J. (2011). 5-HT receptors and reward-
generated metabolites promote metabolic benefits via gut-brain neural related behaviour: a review. Neurosci Biobehav Rev 35, 1419–1449.
circuits. Cell 156, 84–96. Haynes, A.C., Jackson, B., Chapman, H., Tadayyon, M., Johns, A., Porter,
Droste, S.M., Saland, S.K., Schlitter, E.K., and Rodefer, J.S. (2010). AM R.A., and Arch, J.R.S. (2000). A selective orexin-1 receptor antagonist
251 differentially effects food-maintained responding depending on reduces food consumption in male and female rats. Regul Pept 96, 45–
food palatability. Pharmacol Biochem Behav 95, 443–448. 51.
Drucker, D.J. (2006). The biology of incretin hormones. Cell Metab 3, 153– Heisler, L.K., Jobst, E.E., Sutton, G.M., Zhou, L., Borok, E., Thornton-
165. Jones, Z., Liu, H.Y., Zigman, J.M., Balthasar, N., Kishi, T., et al. (2006).
Dunn-Meynell, A.A., Routh, V.H., Kang, L., Gaspers, L., and Levin, B.E. Serotonin reciprocally regulates melanocortin neurons to modulate food
(2002). Glucokinase is the likely mediator of glucosensing in both intake. Neuron 51, 239–249.
glucose-excited and glucose-inhibited central neurons. Diabetes 51, 2- Henao-Mejia, J., Elinav, E., Jin, C., Hao, L., Mehal, W.Z., Strowig, T.,
056–2065. Thaiss, C.A., Kau, A.L., Eisenbarth, S.C., Jurczak, M.J., et al. (2012).
Egecioglu, E., Skibicka, K.P., Hansson, C., Alvarez-Crespo, M., Friberg, P. Inflammasome-mediated dysbiosis regulates progression of NAFLD
A., Jerlhag, E., Engel, J.A., and Dickson, S.L. (2011). Hedonic and and obesity. Nature 482, 179–185.
incentive signals for body weight control. Rev Endocr Metab Disord 12, Hensler, J.G. (2006). Serotonergic modulation of the limbic system. Neu-
141–151. rosci Biobehav Rev 30, 203–214.
Escartín-Pérez, R.E., Cendejas-Trejo, N.M., Cruz-Martínez, A.M., Gonzá- Hommel, J.D., Trinko, R., Sears, R.M., Georgescu, D., Liu, Z.W., Gao, X.
lez-Hernández, B., Mancilla-Díaz, J.M., and Florán-Garduño, B. B., Thurmon, J.J., Marinelli, M., and DiLeone, R.J. (2006). Leptin re-
(2009). Role of cannabinoid CB1 receptors on macronutrient selection ceptor signaling in midbrain dopamine neurons regulates feeding. Ne-
and satiety in rats. Physiol Behav 96, 646–650. uron 51, 801–810.
Fan, W., Boston, B.A., Kesterson, R.A., Hruby, V.J., and Cone, R.D. Horder, J., Harmer, C.J., Cowen, P.J., and McCabe, C. (2010). Reduced
(1997). Role of melanocortinergic neurons in feeding and the agouti neural response to reward following 7 days treatment with the canna-
obesity syndrome. Nature 385, 165–168. binoid CB1 antagonist rimonabant in healthy volunteers. Int J Neuro-
Febbraio, M.A., Hiscock, N., Sacchetti, M., Fischer, C.P., and Pedersen, B. psychopharmacol 13, 1103–1113.
K. (2004). Interleukin-6 is a novel factor mediating glucose homeostasis Hoyda, T.D., Samson, W.K., and Ferguson, A.V. (2009). Adiponectin de-
during skeletal muscle contraction. Diabetes 53, 1643–1648. polarizes parvocellular paraventricular nucleus neurons controlling ne-
Feifel, D., Goldenberg, J., Melendez, G., and Shilling, P.D. (2010). The uroendocrine and autonomic function. Endocrinology 150, 832–840.
acute and subchronic effects of a brain-penetrating, neurotensin-1 rec- Huo, L., Maeng, L., Bjørbaek, C., and Grill, H.J. (2007). Leptin and the
eptor agonist on feeding, body weight and temperature. Neuropharma- control of food intake: neurons in the nucleus of the solitary tract are
cology 58, 195–198. activated by both gastric distension and leptin. Endocrinology 148,
Fioramonti, X., Marsollier, N., Song, Z., Fakira, K.A., Patel, R.M., Brown, 2189–2197.
S., Duparc, T., Pica-Mendez, A., Sanders, N.M., Knauf, C., et al. Huszar, D., Lynch, C.A., Fairchild-Huntress, V., Dunmore, J.H., Fang, Q.,
(2010). Ventromedial hypothalamic nitric oxide production is necessary Berkemeier, L.R., Gu, W., Kesterson, R.A., Boston, B.A., Cone, R.D.,
for hypoglycemia detection and counterregulation. Diabetes 59, 519– et al. (1997). Targeted disruption of the melanocortin-4 receptor results
528. in obesity in mice. Cell 88, 131–141.
Garfield, A.S., and Heisler, L.K. (2009). Pharmacological targeting of the Huynh, M.K.Q., Kinyua, A.W., Yang, D.J., and Kim, K.W. (2016). Hyp-
serotonergic system for the treatment of obesity. J Physiol 587, 49–60. othalamic AMPK as a regulator of energy homeostasis. Neural Plasti-
Geha, P.Y., Aschenbrenner, K., Felsted, J., O’Malley, S.S., and Small, D.M. city 2016, 1–12.
(2013). Altered hypothalamic response to food in smokers. Am J Clin Ikemoto, S. (2010). Brain reward circuitry beyond the mesolimbic dopa-
17 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ........................................... 17

mine system: a neurobiological theory. Neurosci BioBehav Rev 35, (2006). Attenuated peptide YY release in obese subjects is associated
129–150. with reduced satiety. Endocrinology 147, 3–8.
Kamatchi, G.L., and Rathanaswami, P. (2012). Inhibition of deprivation- Lechin, F., van der Dijs, B., and Hernández-Adrián, G. (2006). Dorsal
induced food intake by GABAA antagonists: roles of the hypothalamic, raphe vs. median raphe serotonergic antagonism. Anatomical, physio-
endocrine and alimentary mechanisms. J Clin Biochem Nutr 51, 19–26. logical, behavioral, neuroendocrinological, neuropharmacological and
Kamegai, J., Tamura, H., Shimizu, T., Ishii, S., Sugihara, H., and Wakab- clinical evidences: relevance for neuropharmacological therapy. Prog
ayashi, I. (2001). Chronic central infusion of ghrelin increases hypot- Neuropsychopharmacol Biol Psychiatry 30, 565–585.
halamic neuropeptide Y and Agouti-related protein mRNA levels and Lee, S.J., Reed, L.A., Davies, M.V., Girgenrath, S., Goad, M.E.P., Tomk-
body weight in rats. Diabetes 50, 2438–2443. inson, K.N., Wright, J.F., Barker, C., Ehrmantraut, G., Holmstrom, J., et
Kang, L., Dunn-Meynell, A.A., Routh, V.H., Gaspers, L.D., Nagata, Y., al. (2005). Regulation of muscle growth by multiple ligands signaling
Nishimura, T., Eiki, J., Zhang, B.B., and Levin, B.E. (2006). Glucoki- through activin type II receptors. Proc Natl Acad Sci USA 102, 18117–
nase is a critical regulator of ventromedial hypothalamic neuronal gl- 18122.
ucosensing. Diabetes 55, 412–420. Leibowitz, S.F., Chang, G.Q., Dourmashkin, J.T., Yun, R., Julien, C., and
Kanoski, S.E., Alhadeff, A.L., Fortin, S.M., Gilbert, J.R., and Grill, H.J. Pamy, P.P. (2006). Leptin secretion after a high-fat meal in normal-
(2014). Leptin signaling in the medial nucleus tractus solitarius reduces weight rats: strong predictor of long-term body fat accrual on a high-fat
food seeking and willingness to work for food. Neuropsychopharma- diet. Am J Physiol Endocrinol Metab 290, E258–E267.
cology 39, 605–613. Lenglos, C., Mitra, A., Guèvremont, G., and Timofeeva, E. (2013). Sex
Karlsson, F.H., Tremaroli, V., Nookaew, I., Bergström, G., Behre, C.J., differences in the effects of chronic stress and food restriction on body
Fagerberg, B., Nielsen, J., and Bäckhed, F. (2013). Gut metagenome in weight gain and brain expression of CRF and relaxin-3 in rats. Genes
European women with normal, impaired and diabetic glucose control. Brain Behav 12, 370–387.
Nature 498, 99–103. Levey, A.I., Hersch, S.M., Rye, D.B., Sunahara, R.K., Niznik, H.B., Kitt, C.
Katsurada, K., Maejima, Y., Nakata, M., Kodaira, M., Suyama, S., Iwasaki, A., Price, D.L., Maggio, R., Brann, M.R., and Ciliax, B.J. (1993).
Y., Kario, K., and Yada, T. (2014). Endogenous GLP-1 acts on parav- Localization of D1 and D2 dopamine receptors in brain with subtype-
entricular nucleus to suppress feeding: projection from nucleus tractus specific antibodies. Proc Natl Acad Sci USA 90, 8861–8865.
solitarius and activation of corticotropin-releasing hormone, nesfatin-1 Lieverse, R.J., Masclee, A.A.M., Jansen, J.B.M.J., Rovati, L.C., and La-
and oxytocin neurons. Biochem Biophys Res Commun 451, 276–281. mers, C.B.H.W. (1995). Satiety effects of the type A CCK receptor
Kelly, J., Alheid, G.F., Newberg, A., and Grossman, S.P. (1977). GABA antagonist loxiglumide in lean and obese women. Biol Psychiatry 37,
stimulation and blockade in the hypothalamus and midbrain: effects on 331–335.
feeding and locomotor activity. Pharmacol Biochem Behav 7, 537–541. Lin, Z., Tian, H., Lam, K.S.L., Lin, S., Hoo, R.C.L., Konishi, M., Itoh, N.,
Kim, E.R., Leckstrom, A., and Mizuno, T.M. (2008). Impaired anorectic Wang, Y., Bornstein, S.R., Xu, A., et al. (2013). Adiponectin mediates
effect of leptin in neurotensin receptor 1-deficient mice. Behav Brain the metabolic effects of FGF21 on glucose homeostasis and insulin
Res 194, 66–71. sensitivity in mice. Cell Metab 17, 779–789.
Koch, M., Varela, L., Kim, J.G., Kim, J.D., Hernández-Nuño, F., Simonds, Liu, T., Kong, D., Shah, B.P., Ye, C., Koda, S., Saunders, A., Ding, J.B.,
S.E., Castorena, C.M., Vianna, C.R., Elmquist, J.K., Morozov, Y.M., et Yang, Z., Sabatini, B.L., and Lowell, B.B. (2012). Fasting activation of
al. (2015). Hypothalamic POMC neurons promote cannabinoid-induced AgRP neurons requires NMDA receptors and involves spinogenesis and
feeding. Nature 519, 45–50. increased excitatory tone. Neuron 73, 511–522.
Kollias, H.D., and McDermott, J.C. (2008). Transforming growth factor-β Locke, A.E., Kahali, B., Berndt, S.I., Justice, A.E., Pers, T.H., Day, F.R.,
and myostatin signaling in skeletal muscle. J Appl Physiol 104, 579– Powell, C., Vedantam, S., Buchkovich, M.L., Yang, J., et al. (2015).
587. Genetic studies of body mass index yield new insights for obesity bi-
Kreisler, A.D., Davis, E.A., and Rinaman, L. (2014). Differential activation ology. Nature 518, 197–206.
of chemically identified neurons in the caudal nucleus of the solitary Lockie, S.H., Heppner, K.M., Chaudhary, N., Chabenne, J.R., Morgan, D.
tract in non-entrained rats after intake of satiating vs. non-satiating A., Veyrat-Durebex, C., Ananthakrishnan, G., Rohner-Jeanrenaud, F.,
meals. Physiol Behav 136, 47–54. Drucker, D.J., DiMarchi, R., et al. (2012). Direct control of brown
Kristensen, P., Judge, M.E., Thim, L., Ribel, U., Christjansen, K.N., Wulff, adipose tissue thermogenesis by central nervous system glucagon-like
B.S., Clausen, J.T., Jensen, P.B., Madsen, O.D., Vrang, N., et al. (1998). peptide-1 receptor signaling. Diabetes 61, 2753–2762.
Hypothalamic CART is a new anorectic peptide regulated by leptin. Maejima, Y., Kohno, D., Iwasaki, Y., and Yada, T. (2011). Insulin suppr-
Nature 393, 72–76. esses ghrelin-induced calcium signaling in neuropeptide Y neurons of
Kroemer, N.B., Krebs, L., Kobiella, A., Grimm, O., Pilhatsch, M., Bidli- the hypothalamic arcuate nucleus. Aging 3, 1092–1097.
ngmaier, M., Zimmermann, U.S., and Smolka, M.N. (2013). Fasting McFadden, K.L., Cornier, M.A., and Tregellas, J.R. (2014). The role of
levels of ghrelin covary with the brain response to food pictures. Addict alpha-7 nicotinic receptors in food intake behaviors. Front Psychol 5,
Biol 18, 855–862. 553.
Lam, D.D., Garfield, A.S., Marston, O.J., Shaw, J., and Heisler, L.K. McFarlane, M.R., Brown, M.S., Goldstein, J.L., and Zhao, T.J. (2014).
(2010). Brain serotonin system in the coordination of food intake and Induced ablation of ghrelin cells in adult mice does not decrease food
body weight. Pharmacol Biochem Behav 97, 84–91. intake, body weight, or response to high-fat diet. Cell Metab 20, 54–60.
Lane, M.D., Hu, Z., Cha, S.H., Dai, Y., Wolfgang, M., and Sidhaye, A. Mebel, D.M., Wong, J.C.Y., Dong, Y.J., and Borgland, S.L. (2012). Insulin
(2005). Role of malonyl-CoA in the hypothalamic control of food int- in the ventral tegmental area reduces hedonic feeding and suppresses
ake and energy expenditure. Biochm Soc Trans 33, 1063–1067. dopamine concentration via increased reuptake. Eur J Neurosci 36,
Langleben, D.D., Busch, E.L., O’Brien, C.P., and Elman, I. (2012). Depot 2336–2346.
naltrexone decreases rewarding properties of sugar in patients with Mietlicki-Baase, E.G., Ortinski, P.I., Rupprecht, L.E., Olivos, D.R., Alha-
opioid dependence. Psychopharmacology 220, 559–564. deff, A.L., Pierce, R.C., and Hayes, M.R. (2013). The food intake-
Lau, J., and Herzog, H. (2014). CART in the regulation of appetite and suppressive effects of glucagon-like peptide-1 receptor signaling in the
energy homeostasis. Front Neurosci 8, 313. ventral tegmental area are mediated by AMPA/kainate receptors. Am J
Le Foll, C., Dunn-Meynell, A., Musatov, S., Magnan, C., and Levin, B.E. Physiol Endocrinol Metab 305, E1367–E1374.
(2013). FAT/CD36: a major regulator of neuronal fatty acid sensing and Mimee, A., and Ferguson, A.V. (2015). Glycemic state regulates melano-
energy homeostasis in rats and mice. Diabetes 62, 2709–2716. cortin, but not nesfatin-1, responsiveness of glucose-sensing neurons in
le Roux, C.W., Batterham, R.L., Aylwin, S.J.B., Patterson, M., Borg, C.M., the nucleus of the solitary tract. Am J Physiol Regul Integrat Comp
Wynne, K.J., Kent, A., Vincent, R.P., Gardiner, J., Ghatei, M.A., et al. Physiol 308, R690–R699.
18 . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ........................................... 18

Mineur, Y.S., Abizaid, A., Rao, Y., Salas, R., DiLeone, R.J., Gündisch, D., “liking” for sweet rewards. J Neurosci 23, 9395–9402.
Diano, S., De Biasi, M., Horvath, T.L., Gao, X.B., et al. (2011). Nic- Peciña, S., and Smith, K.S. (2010). Hedonic and motivational roles of
otine decreases food intake through activation of POMC neurons. opioids in food reward: implications for overeating disorders. Pharma-
Science 332, 1330–1332. col Biochem Behav 97, 34–46.
Moran, T.H. (2000). Cholecystokinin and satiety: current perspectives. Pedersen, B.K., and Febbraio, M.A. (2012). Muscles, exercise and obesity:
Nutrition 16, 858–865. skeletal muscle as a secretory organ. Nat Rev Endocrinol 8, 457–465.
Morris, D.L., and Rui, L. (2009). Recent advances in understanding leptin Porte, D., Jr., Baskin, D.G., and Schwartz, M.W. (2005). Insulin signaling
signaling and leptin resistance. Am J Physiol Endocrinol Metab 297, in the central nervous system: a critical role in metabolic homeostasis
E1247–E1259. and disease from C. elegans to humans. Diabetes 54, 1264–1276.
Morrison, S.F. (2004). Central pathways controlling brown adipose tissue Psilopanagioti, A., Papadaki, H., Kranioti, E.F., Alexandrides, T.K., and
thermogenesis. News Physiol Sci 19, 67–74. Varakis, J.N. (2009). Expression of adiponectin and adiponectin rece-
Morton, G.J., Thatcher, B.S., Reidelberger, R.D., Ogimoto, K., Wolden- ptors in human pituitary gland and brain. Neuroendocrinology 89, 38–
Hanson, T., Baskin, D.G., Schwartz, M.W., and Blevins, J.E. (2012). 47.
Peripheral oxytocin suppresses food intake and causes weight loss in Qin, J., Li, Y., Cai, Z., Li, S., Zhu, J., Zhang, F., Liang, S., Zhang, W.,
diet-induced obese rats. Am J Physiol Endocrinol Metab 302, E134– Guan, Y., Shen, D., et al. (2012). A metagenome-wide association study
E144. of gut microbiota in type 2 diabetes. Nature 490, 55–60.
Murphy, B.A., Fakira, K.A., Song, Z., Beuve, A., and Routh, V.H. (2009). Rabiner, E.A., Beaver, J., Makwana, A., Searle, G., Long, C., Nathan, P.J.,
AMP-activated protein kinase and nitric oxide regulate the glucose Newbould, R.D., Howard, J., Miller, S.R., Bush, M.A., et al. (2011).
sensitivity of ventromedial hypothalamic glucose-inhibited neurons. Molecular and functional neuroimaging of human opioid receptor ph-
Am J Physiol Cell Physiol 297, C750–C758. armacology. Mol Psychiatry 16, 785.
Murray, E., Brouwer, S., McCutcheon, R., Harmer, C.J., Cowen, P.J., and Rahmouni, K., Morgan, D.A., Morgan, G.M., Liu, X., Sigmund, C.D.,
McCabe, C. (2014). Opposing neural effects of naltrexone on food Mark, A.L., and Haynes, W.G. (2004). Hypothalamic PI3K and MAPK
reward and aversion: implications for the treatment of obesity. Psych- differentially mediate regional sympathetic activation to insulin. J Clin
opharmacology 231, 4323–4335. Invest 114, 652–658.
Newgard, C.B., An, J., Bain, J.R., Muehlbauer, M.J., Stevens, R.D., Lien, Ramnanan, C.J., Saraswathi, V., Smith, M.S., Donahue, E.P., Farmer, B.,
L.F., Haqq, A.M., Shah, S.H., Arlotto, M., Slentz, C.A., et al. (2009). A Farmer, T.D., Neal, D., Williams, P.E., Lautz, M., Mari, A., et al.
branched-chain amino acid-related metabolic signature that differenti- (2011). Brain insulin action augments hepatic glycogen synthesis wit-
ates obese and lean humans and contributes to insulin resistance. Cell hout suppressing glucose production or gluconeogenesis in dogs. J Clin
Metab 9, 311–326. Invest 121, 3713–3723.
Nguyen, A.D., Mitchell, N.F., Lin, S., Macia, L., Yulyaningsih, E., Bald- Re´thelyi, M. (1984). Diffusional barrier around the hypothalamic arcuate
ock, P.A., Enriquez, R.F., Zhang, L., Shi, Y.C., Zolotukhin, S., et al. nucleus in the rat. Brain Res 307, 355–358.
(2012). Y1 and Y5 receptors are both required for the regulation of food Richard, J.E., Anderberg, R.H., Göteson, A., Gribble, F.M., Reimann, F.,
intake and energy homeostasis in mice. PLoS ONE 7, e40191. and Skibicka, K.P. (2015). Activation of the GLP-1 receptors in the
Obici, S., Feng, Z., Morgan, K., Stein, D., Karkanias, G., and Rossetti, L. nucleus of the solitary tract reduces food reward behavior and targets
(2002). Central administration of oleic acid inhibits glucose production the mesolimbic system. PLoS ONE 10, e0119034.
and food intake. Diabetes 51, 271–275. Richard, J.E., Farkas, I., Anesten, F., Anderberg, R.H., Dickson, S.L.,
Olszewski, P.K., Cedernaes, J., Olsson, F., Levine, A.S., and Schiöth, H.B. Gribble, F.M., Reimann, F., Jansson, J.O., Liposits, Z., and Skibicka, K.
(2008). Analysis of the network of feeding neuroregulators using the P. (2014). GLP-1 receptor stimulation of the lateral parabrachial nucleus
Allen Brain Atlas. Neurosci Biobehav Rev 32, 945–956. reduces food intake: neuroanatomical, electrophysiological, and beha-
Olszewski, P.K., Klockars, A., Olszewska, A.M., Fredriksson, R., Schiöth, vioral evidence. Endocrinology 155, 4356–4367.
H.B., and Levine, A.S. (2010). Molecular, immunohistochemical, and Rinaman, L. (2003). Hindbrain noradrenergic lesions attenuate anorexia
pharmacological evidence of oxytocin’s role as inhibitor of carbohy- and alter central cFos expression in rats after gastric viscerosensory
drate but not fat intake. Endocrinology 151, 4736–4744. stimulation. J Neurosci 23, 10084–10092.
Olszewski, P.K., Klockars, A., Schiöth, H.B., and Levine, A.S. (2010). Roberts, L.D., Boström, P., O’Sullivan, J.F., Schinzel, R.T., Lewis, G.D.,
Oxytocin as feeding inhibitor: maintaining homeostasis in consumma- Dejam, A., Lee, Y.K., Palma, M.J., Calhoun, S., Georgiadi, A., et al.
tory behavior. Pharmacol Biochem Behav 97, 47–54. (2014). β-Aminoisobutyric acid induces browning of white fat and h-
Onaka, T., Takayanagi, Y., and Yoshida, M. (2012). Roles of oxytocin epatic β-oxidation and is inversely correlated with cardiometabolic risk
neurones in the control of stress, energy metabolism, and social beha- factors. Cell Metab 19, 96–108.
viour. J Neuroendocrinol 24, 587–598. Rojas-Morales, P., Tapia, E., and Pedraza-Chaverri, J. (2016). beta-Hydr-
Ong, Z.Y., Alhadeff, A.L., and Grill, H.J. (2015). Medial nucleus tractus oxybutyrate: a signaling metabolite in starvation response? Cell Signal
solitarius oxytocin receptor signaling and food intake control: the role 28, 917–923.
of gastrointestinal satiation signal processing. Am J Physiol Regul Roman, C.W., Derkach, V.A., and Palmiter, R.D. (2016). Genetically and
Integr Comp Physiol 308, R800–R806. functionally defined NTS to PBN brain circuits mediating anorexia. Nat
Ott, V., Finlayson, G., Lehnert, H., Heitmann, B., Heinrichs, M., Born, J., Commun 7, 11905.
and Hallschmid, M. (2013). Oxytocin reduces reward-driven food Routh, V.H., Hao, L., Santiago, A.M., Sheng, Z., and Zhou, C. (2014).
intake in humans. Diabetes 62, 3418–3425. Hypothalamic glucose sensing: making ends meet. Front Syst Neurosci
Pagotto, U., Marsicano, G., Cota, D., Lutz, B., and Pasquali, R. (2006). The 8, 236.
emerging role of the endocannabinoid system in endocrine regulation Sahu, A., Carraway, R.E., and Wang, Y.P. (2001). Evidence that neurote-
and energy balance. Endocr Rev 27, 73–100. nsin mediates the central effect of leptin on food intake in rat. Brain Res
Parker, K.L., and Schimmer, B.P. (1997). Steroidogenic factor 1: a key 888, 343–347.
determinant of endocrine development and function. Endocr Rev 18, Samuel, B.S., Shaito, A., Motoike, T., Rey, F.E., Backhed, F., Manchester,
361–377. J.K., Hammer, R.E., Williams, S.C., Crowley, J., Yanagisawa, M., et al.
Patterson, C.M., Leshan, R.L., Jones, J.C., and Myers Jr., M.G. (2011). (2008). Effects of the gut microbiota on host adiposity are modulated by
Molecular mapping of mouse brain regions innervated by leptin rece- the short-chain fatty-acid binding G protein-coupled receptor, Gpr41.
ptor-expressing cells. Brain Res 1378, 18–28. Proc Natl Acad Sci USA 105, 16767–16772.
Peciña, S., Cagniard, B., Berridge, K.C., Aldridge, J.W., and Zhuang, X. Sato, I., Arima, H., Ozaki, N., Ozaki, N., Watanabe, M., Goto, M., Shimizu,
(2003). Hyperdopaminergic mutant mice have higher “wanting” but not H., Hayashi, M., Banno, R., Nagasaki, H., et al. (2007). Peripherally
19 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ........................................... 19

administered baclofen reduced food intake and body weight in db/db as Trapp, S., and Cork, S.C. (2015). PPG neurons of the lower brain stem and
well as diet-induced obese mice. FEBS Lett 581, 4857–4864. their role in brain GLP-1 receptor activation. Am J Physiol Regul Integr
Savard, P., Mérand, Y., Leblanc, J., and Dupont, A. (1983). Limitation of Comp Physiol 309, R795–R804.
access to highly palatable foods increases the norepinephrine content of Trivedi, P., Yu, H., MacNeil, D.J., Van der Ploeg, L.H.T., and Guan, X.M.
many discrete hypothalamic and amygdaloidal nuclei of rat brain. Life (1998). Distribution of orexin receptor mRNA in the rat brain. FEBS
Sci 33, 2513–2519. Lett 438, 71–75.
Schneeberger, M., Gomis, R., and Claret, M. (2014). Hypothalamic and Unger, T.J., Calderon, G.A., Bradley, L.C., Sena-Esteves, M., and Rios, M.
brainstem neuronal circuits controlling homeostatic energy balance. J (2007). Selective deletion of Bdnf in the ventromedial and dorsomedial
Endocrinol 220, T25–T46. hypothalamus of adult mice results in hyperphagic behavior and obesity.
Schultz, W. (2015). Neuronal reward and decision signals: from theories to J Neurosci 27, 14265–14274.
data. Physiol Rev 95, 853–951. Vaccari, C., Lolait, S.J., and Ostrowski, N.L. (1998). Comparative distri-
Schwartz, M.W., Woods, S.C., Porte, D., Seeley, R.J., and Baskin, D.G. bution of vasopressin V1b and oxytocin receptor messenger ribonucleic
(2000). Central nervous system control of food intake. Nature 404, 661– acids in brain. Endocrinology 139, 5015–5033.
671. Valdés, J.L., Sánchez, C., Riveros, M.E., Blandina, P., Contreras, M., Farías
Schwiertz, A., Taras, D., Schäfer, K., Beijer, S., Bos, N.A., Donus, C., and , P., and Torrealba, F. (2010). The histaminergic tuberomammillary n-
Hardt, P.D. (2010). Microbiota and SCFA in lean and overweight he- ucleus is critical for motivated arousal. Eur J Neurosci 31, 2073–2085.
althy subjects. Obesity 18, 190–195. van de Giessen, E., Celik, F., Schweitzer, D.H., van den Brink, W., and
Seale, P., Conroe, H.M., Estall, J., Kajimura, S., Frontini, A., Ishibashi, J., Booij, J. (2014). Dopamine D2/3 receptor availability and amphetamine-
Cohen, P., Cinti, S., and Spiegelman, B.M. (2011). Prdm16 determines induced dopamine release in obesity. J Psychopharmacol 28, 866–873.
the thermogenic program of subcutaneous white adipose tissue in mice. van Marken Lichtenbelt, W.D., Vanhommerig, J.W., Smulders, N.M., Dr-
J Clin Invest 121, 96–105. ossaerts, J.M.A.F.L., Kemerink, G.J., Bouvy, N.D., Schrauwen, P., and
Secher, A., Jelsing, J., Baquero, A.F., Hecksher-Sørensen, J., Cowley, M. Teule, G.J.J. (2009). Cold-activated brown adipose tissue in healthy
A., Dalbøge, L.S., Hansen, G., Grove, K.L., Pyke, C., Raun, K., et al. men. N Engl J Med 360, 1500–1508.
(2014). The arcuate nucleus mediates GLP-1 receptor agonist liraglu- van Zessen, R., Phillips, J.L., Budygin, E.A., and Stuber, G.D. (2012).
tide-dependent weight loss. J Clin Invest 124, 4473–4488. Activation of VTA GABA neurons disrupts reward consumption.
Shanahan, F. (2012). The gut microbiota—a clinical perspective on lessons Neuron 73, 1184–1194.
learned. Nat Rev Gastroenterol Hepatol 9, 609–614. Ventura, R., Morrone, C., and Puglisi-Allegra, S. (2007). Prefrontal/accu-
Sofroniew, M.V. (1983). Morphology of vasopressin and oxytocin neurones mbal catecholamine system determines motivational salience attribution
and their central and vascular projections. Prog Brain Res 60, 101–114. to both reward- and aversion-related stimuli. Proc Natl Acad Sci USA
Sohn, J.W. (2015). Network of hypothalamic neurons that control appetite. 104, 5181–5186.
BMB Rep 48, 229–233. Villanueva, E.C., and Myers, M.G. (2008). Leptin receptor signaling and
Sola-Penna, M. (2008). Metabolic regulation by lactate. IUBMB Life 60, the regulation of mammalian physiology. Int J Obes 32, S8–S12.
605–608. Voigt, J.P., and Fink, H. (2015). Serotonin controlling feeding and satiety.
Srisai, D., Gillum, M.P., Panaro, B.L., Zhang, X.M., Kotchabhakdi, N., Behav Brain Res 277, 14–31.
Shulman, G.I., Ellacott, K.L.J., and Cone, R.D. (2011). Characterization Volkow, N.D., Wang, G.J., Telang, F., Fowler, J.S., Thanos, P.K., Logan, J.,
of the hyperphagic response to dietary fat in the MC4R knockout mo- Alexoff, D., Ding, Y.S., Wong, C., Ma, Y., et al. (2008). Low dopamine
use. Endocrinology 152, 890–902. striatal D2 receptors are associated with prefrontal metabolism in obese
Stadler, M., Tomann, L., Storka, A., Wolzt, M., Peric, S., Bieglmayer, C., subjects: possible contributing factors. Neuroimage 42, 1537–1543.
Pacini, G., Dickson, S.L., Brath, H., Bech, P., et al. (2014). Effects of Wang, C.F., Billington, C.J., Levine, A.S., and Kotz, C.M. (2000). Effect of
smoking cessation on β-cell function, insulin sensitivity, body weight, CART in the hypothalamic paraventricular nucleus on feeding and
and appetite. Eur J Endocrinol 170, 219–227. uncoupling protein gene expression. Neuroreport 11, 3251–3255.
Stolarczyk, E., Guissard, C., Michau, A., Even, P.C., Grosfeld, A., Serra- Wang, G.J., Volkow, N.D., Logan, J., Pappas, N.R., Wong, C.T., Zhu, W.,
das, P., Lorsignol, A., Pénicaud, L., Brot-Laroche, E., Leturque, A., et Netusll, N., and Fowler, J.S. (2001). Brain dopamine and obesity.
al. (2010). Detection of extracellular glucose by GLUT2 contributes to Lancet 357, 354–357.
hypothalamic control of food intake. Am J Physiol Endocrinol Metab Wellman, P.J. (2000). Norepinephrine and the control of food intake.
298, E1078–E1087. Nutrition 16, 837–842.
Sun, J., Gao, Y., Yao, T., Huang, Y., He, Z., Kong, X., Yu, K.J., Wang, R.T., West, D.B., Fey D., and Woods, S.C. (1984). Cholecystokinin persistently
Guo, H., Yan, J., et al. (2016). Adiponectin potentiates the acute effects suppresses meal size but not food intake in free-feeding rats. Am J
of leptin in arcuate Pomc neurons. Mol Metab 5, 882–891. Physiol 246, R776–787.
Taber, K.H., Black, D.N., Porrino, L.J., and Hurley, R.A. (2012). Neuro- Williams, D.L. (2014). Neural integration of satiation and food reward: role
anatomy of dopamine: reward and addiction. J Neuropsychiatry Clin of GLP-1 and orexin pathways. Physiol Behav 136, 194–199.
Neurosci 24, 1–4. Wise, R.A., and Bozarth, M.A. (1987). A psychomotor stimulant theory of
Taraschenko, O.D., Maisonneuve, I.M., and Glick, S.D. (2011). Resistance addiction. Psychol Rev 94, 469–492.
of male Sprague-Dawley rats to sucrose-induced obesity: effects of 18- Xu, A., Wang, Y., Keshaw, H., Xu, L.Y., Lam, K.S.L., and Cooper, G.J.S.
methoxycoronaridine. Physiol Behav 102, 126–131. (2003). The fat-derived hormone adiponectin alleviates alcoholic and
Teff, K.L., and Kim, S.F. (2011). Atypical antipsychotics and the neural nonalcoholic fatty liver diseases in mice. J Clin Invest 112, 91–100.
regulation of food intake and peripheral metabolism. Physiol Behav Yamada, H., Okumura, T., Motomura, W., Kobayashi, Y., and Kohgo, Y.
104, 590–598. (2000). Inhibition of food intake by central injection of anti-orexin
Tolhurst, G., Heffron, H., Lam, Y.S., Parker, H.E., Habib, A.M., Diakog- antibody in fasted rats. Biochem Biophys Res Commun 267, 527–531.
iannaki, E., Cameron, J., Grosse, J., Reimann, F., and Gribble, F.M. Yamada, M., Miyakawa, T., Duttaroy, A., Yamanaka, A., Moriguchi, T.,
(2012). Short-chain fatty acids stimulate glucagon-like peptide-1 secr- Makita, R., Ogawa, M., Chou, C.J., Xia, B., Crawley, J.N., et al. (2001).
etion via the G-protein-coupled receptor FFAR2. Diabetes 61, 364–371. Mice lacking the M3 muscarinic acetylcholine receptor are hypophagic
Torii, K., Uneyama, H., and Nakamura, E. (2013). Physiological roles of and lean. Nature 410, 207–212.
dietary glutamate signaling via gut-brain axis due to efficient digestion Yamauchi, T., Iwabu, M., Okada-Iwabu, M., and Kadowaki, T. (2014).
and absorption. J Gastroenterol 48, 442–451. Adiponectin receptors: a review of their structure, function and how
Torrealba, F., Riveros, M.E., Contreras, M., and Valdes, J.L. (2012). His- they work. Best Practice Res Clin Endocrinol Metab 28, 15–23.
tamine and motivation. Front Syst Neurosci 6, 51. Yamauchi, T., Kamon, J., Minokoshi, Y., Ito, Y., Waki, H., Uchida, S.,
20 . . . . . . . . . . . . . . . . . . . . . . . . . . Wen, S., et al. Sci China Life Sci ........................................... 20

Yamashita, S., Noda, M., Kita, S., Ueki, K., et al. (2002). Adiponectin mutation affecting human TrkB associated with severe obesity and
stimulates glucose utilization and fatty-acid oxidation by activating developmental delay. Nat Neurosci 7, 1187–1189.
AMP-activated protein kinase. Nat Med 8, 1288–1295. Yeomans, M.R., and Gray, R.W. (1996). Selective effects of naltrexone on
Yamauchi, T., Kamon, J., Waki, H., Imai, Y., Shimozawa, N., Hioki, K., food pleasantness and intake. Physiol Behav 60, 439–446.
Uchida, S., Ito, Y., Takakuwa, K., Matsui, J., et al. (2003). Globular Yoshida, K., Li, X., Cano, G., Lazarus, M., and Saper, C.B. (2009). Parallel
adiponectin protected ob/ob mice from diabetes and ApoE-deficient preoptic pathways for thermoregulation. J Neurosci 29, 11954–11964.
mice from atherosclerosis. J Biol Chem 278, 2461–2468. Zheng, H., and Berthoud, H.R. (2008). Neural systems controlling the drive
Yamauchi, T., Nio, Y., Maki, T., Kobayashi, M., Takazawa, T., Iwabu, M., to eat: mind versus metabolism. Physiology (Bethesda) 23, 75–83.
Okada-Iwabu, M., Kawamoto, S., Kubota, N., Kubota, T., et al. (2007). Zhou, L., Podolsky, N., Sang, Z., Ding, Y., Fan, X., Tong, Q., Levin, B.E.,
Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of and McCrimmon, R.J. (2010). The medial amygdalar nucleus: a novel
adiponectin binding and metabolic actions. Nat Med 13, 332–339. glucose-sensing region that modulates the counterregulatory response to
Yang, J., Brown, M.S., Liang, G., Grishin, N.V., and Goldstein, J.L. (2008). hypoglycemia. Diabetes 59, 2646–2652.
Identification of the acyltransferase that octanoylates ghrelin, an app- Zhou, L., Sutton, G.M., Rochford, J.J., Semple, R.K., Lam, D.D., Oksanen,
etite-stimulating peptide hormone. Cell 132, 387–396. L.J., Thornton-Jones, Z.D., Clifton, P.G., Yueh, C.Y., Evans, M.L., et al.
Yeo, G.S.H., Connie Hung, C.C., Rochford, J., Keogh, J., Gray, J., Sivar- (2007). Serotonin 2C receptor agonists improve type 2 diabetes via
amakrishnan, S., O’Rahilly, S., and Farooqi, I.S. (2004). A de novo melanocortin-4 receptor signaling pathways. Cell Metab 6, 398–405.

You might also like