Anda di halaman 1dari 12

REVIEWS

Neurobiology of food intake in health


and disease
Gregory J.Morton, Thomas H.Meek and Michael W.Schwartz
Abstract | Under normal conditions, food intake and energy expenditure are balanced by
a homeostatic system that maintains stability of body fat content over time. However, this
homeostatic system can be overridden by the activation of emergency response circuits
that mediate feeding responses to emergent or stressful stimuli. Inhibition of these
circuits is therefore permissive for normal energy homeostasis to occur, and their chronic
activation can cause profound, even life-threatening, changes in body fat mass. This
Review highlights how the interplay between homeostatic and emergency feeding
circuits influences the biologically defended level of body weight under physiological and
pathophysiological conditions.

Whether to eat, what to eat, when to eat and how much point is that activation of these emergency circuits can
Energy homeostasis
The biological process by to eat in any one meal are decisions that each of us is potentially override normal control of energy homeo
which the body maintains body intimately familiar with and that we are capable of mak stasis irrespective of whether these circuits function
fat stores by balancing energy ing with little conscious effort. As these decisions can to increase or decrease food intake. The goals of this
intake with energy expenditure be influenced by a nearly limitless number of variables, Review are to describe how food intake is governed by
over time.
daytoday energy intake tends to vary both between the energy homeostasis system and how it is altered in
Anorexia and within individuals1. However, in normal individuals, times of stress, and to consider how pathological activa
A disorder that is characterized body weight and body fat content are typically quite stable tion of emergency response circuits can cause disorders
by a reduction in energy intake over time2,3 owing to a biological process termed energy of bodyweight.
and accompanied weight loss.
homeostasis that matches energy intake to expenditure
over long periods of time. The energy homeostasis sys The energy homeostasis system
tem comprises neurons in the mediobasal hypothalamus First proposed by Kennedy 10 some 60years ago, energy
and other brain areas4 that are a part of a neurocircuit that homeostasis is achieved by a system whereby circulat
regulates food intake in response to input from humoral ing signals inform the brain of available energy stores (a
signals that circulate at concentrations proportionate to process referred to as adiposity negative feedback) and,
body fat content46. in response, the brain makes corrective adjustments
The robust efficiency with which the energy home to food intake. (The energy homeostasis system also
ostasis system works in normal-weight humans and regulates energy expenditure, a topic that has recently
animal models seems to be at odds with the very high been reviewed in REFS11,12.) The best-studied humoral
prevalence of overweight and obesity in Westernized mediator of adiposity negative feedback is the adipo
societies7. Also common are disorders characterized cyte hormone leptin. Leptin is secreted from adipose
by anorexia and progressive loss of body mass (wasting tissue13, circulates in proportion to body fat stores14,
illness) that importantly contribute to the mortality of enters the brain in proportion to its plasma level15 and
cancer and other diseases8. Little is known regarding acts on key neurons that regulate energy balance16,17.
mechanisms underlying these disorders, but recently Moreover, leptin administration directly into the brain
Diabetes and Obesity Center
of Excellence, Department of identified neurocircuits, which are referred to here as reduces food intake and body weight18,19, although con
Medicine, University of emergency feeding circuits, may play a part. Some of versely, reduced or impaired neuronal leptin signalling
Washington, Seattle, these emergency circuits are designed to increase plasma promotes hyperphagia and weight gain13,20.
Washington 98109, USA. glucose levels (partly by increased feeding) when they The pancreatic hormone insulin is also implicated
Correspondence to M.W.S.
email: mschwart@u.
are activated9, whereas others prevent feeding when to in energy homeostasis. Like leptin, insulin circulates in
washington.edu do so is maladaptive (for example, under conditions of proportion to body fat 21 and acts in the brain to reduce
doi:10.1038/nrn3745 trauma, illness or threats from the environment)8. A key food intake22; conversely, reduced neuronal insulin

NATURE REVIEWS | NEUROSCIENCE VOLUME 15 | JUNE 2014 | 367

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

Adiposity negative feedback signalling causes a mild expansion of body fat mass23. Satiety perception. Whereas the decision to eat (meal
signals Although both hormones are implicated in this adiposity initiation) is influenced by many external factors, the
Hormones that circulate in negative feedback control system, the feeding effect of amount eaten (meal size) is primarily determined by
direct proportion to body fat leptin is quantitatively much greater than that of insulin. internal signals. Among the most important internal sig
and convey the state of total
energy stores to the CNS.
Beyond adiposity negative feedback signals, numer nals are peptides such as CCK and GLP1 that are secreted
ous hormonal and nutrient-related signals can potently from the gastrointestinal tract in response to food inges
Satiety influence feeding. Among these are gut peptides that are tion26,29. Satiety information is conveyed by these peptides,
The state of feeling full to the involved in the perception of satiety and hence participate as well as by neural signals generated by gastric distension,
point of satisfaction after the
in the termination of individual meals. Putative satiety to the CNS through afferent fibres of the vagus nerve that
consumption of food.
signals include peptide YY336 (PYY336)24, glucagon-like project from the gut to the nucleus of the solitary tract
peptide1 (GLP1)25 and cholecystokinin (CCK)26. The (NTS) in the caudal hindbrain. Satiety peptides trigger
gastric hormone ghrelin27, conversely, is secreted before food intake inhibition that is profound but short-lived,
meal onset and can stimulate feeding. Food intake can and they do not reliably cause sustained weight loss with
also be inhibited by other endogenous mediators, includ repeated dosing. Illustrating this point is a study in rats
ing pro-inflammatory cytokines (such as interleukin6 in which serial injections of CCK were administered at
and tumour necrosis factor-) and nutrients themselves the onset of each meal over a period of days30. Although
(for example, glucose and free fatty acids28) (FIG.1). CCK consistently decreased meal size, its impact on body
weight was minimal because of a compensatory increase
in meal frequency, such that the total amount of calories
consumed was not substantially affected30.
Reward Buried within this observation is a fundamental
axiom regarding how the energy homeostasis system
works: adiposity negative feedback reduces food intake
in part by increasing brain responsiveness to satiety sig
Food intake Satiety nals31, an effect that is mediated by neuronal input from
leptin and insulin32,33. Weight loss lowers the plasma lev
els of these hormones, which reduces the satiating effect
of food (by reducing the response to satiety signals) and
Vagus nerve thereby increases meal size. Consistent with this concept
Spinal nerves is evidence that leptin reduces food intake by enhancing
the response to satiety signals32,34,35 and thereby decreases
Nutrient-related Satiety signals meal size36,37, whereas deficient brain leptin signalling
(short term) reduces the responsiveness to CCK, which leads to an
increase in meal size31,38,39. This interaction between lep
tin and satiety signals seems to involve the activation of
Adiposity-related
(long term) leptin receptors in both the hindbrain (on NTS neurons
Blood
themselves40) and the forebrain (on neurons in the hypo
thalamic arcuate nucleus (ARC) that project directly or
indirectly to the NTS41). The key point is that this inter
Glucose,
action enables the amount of food consumed during indi
FFAs vidual meals to be adjusted to compensate for changes in
body fat mass35,42 (FIG.2).

Food reward. That palatability is a crucial determinant


Insulin of the decision to eat, and that highly palatable foods can
trigger eating at times when food would not otherwise
Leptin be consumed, comes as no surprise. This type of feed
CCK
GLP1
ing has been described as non-homeostatic (REF.43),
because it can occur in the absence of the need to replen
ish depleted fuel stores. However, we disagree with this
Figure 1 | CNS regulation of energy homeostasis. The Nature
CNS Reviews | Neuroscience
integrates input from characterization because regulation of food reward is in
long-term energy stores (for example, leptin) and short-term meal-related signals fact integral to how energy homeostasis is achieved44.
(nutrients and gut-derived satiety signals) to regulate food intake and energy First, reduced perception of food reward seems to be
expenditure in a manner that maintains stable body fat stores over time. Positive energy
an inherent aspect of satiety, a concept that has recently
balance induced by overfeeding inhibits the rewarding properties of food while
enhancing meal-induced satiety, thereby reducing food intake. In response to energy
been developed based on findings from rodent stud
deprivation, CNS adaptive responses are engaged that both increase the rewarding ies45 and that is supported by brain imaging studies in
properties of food and reduce the response to satiety signals, collectively resulting in humans46,47. Second, a fasting-induced increase in food
increased food consumption until deficient fat stores are replenished. CCK, intake (refeeding hyperphagia) is associated with an
cholecystokinin; FFAs, free fatty acids; GLP1, glucagon-like peptide 1. Modified from increase in both the amount of work an animal will do to
Marx,J. Cellular warriors at the battle of the bulge. Science 299, 846849 (2003)170. obtain food (a measure of the motivation to eat) and the
Reprinted with permission from AAAS. ability of food to condition place preference (a measure

368 | JUNE 2014 | VOLUME 15 www.nature.com/reviews/neuro

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

Neuropeptide of learning driven by foods reinforcing properties), and hypothalamic areas such as the ARC, as well as from
A small protein-like molecule both of these responses are blunted by intracerebro reward-processing areas such as the NAc, the lateral
that is used by neurons to ventricular administration of leptin or insulin48,49. Stated hypothalamic area (LHA) is proposed to integrate reward-
communicate with each other, more simply, weight loss induced by fasting or caloric related input with information related to energy homeo
often in a paracrine manner.
restriction stimulates a compensatory hyperphagia in stasis53. In turn, LHA neurons project to and influence the
part by increasing the rewarding properties of food, and mesolimbic dopamine system54 as well as hindbrain areas
this effect is mediated by reduced neuronal input from such as the NTS that regulate satiety. To summarize, the
adiposity negative feedback signals. Clinical evidence in energy homeostasis system functions through powerful
support of this assertion includes the finding that leptin and coordinated influences over the perception of satiety
administration reduces food intake in humans with con on the one hand and of food reward on theother.
genital leptin deficiency. This effect is associated with
decreases not only in the subjective experience of food Energy homeostasis neurocircuitry
reward but also in the activation of brain areas that are Our understanding of the neurocircuitry of energy
associated with reward (that is, the ventral striatum) in homeostasis has rapidly grown over the past decade.
response to food-related stimuli50 (FIG.3). Among the most-studied and best-understood neuronal
The homeostatic regulation of food reward is the sub subpopulations are those that coexpress neuropeptide Y
ject of recent reviews51,52. Briefly, brain reward circuits that (NPY), agouti-related protein (AGRP; an antagonist of
process information related to food reward (for example, melanocortin signalling) and GABA16,55; these neurons are
the hedonic value of and motivation to work for food) henceforth referred to as AGRP neurons. AGRP neurons
are influenced by metabolic and hormonal signals that are situated in the ARC and they stimulate feeding when
communicate information regarding the status of energy they are activated56. Consistent with a role in energy
stores to the CNS. These reward circuits include meso homeostasis, AGRP neurons are inhibited by both insu
limbic dopaminergic neurons in the ventral tegmental lin and leptin5759, whereas they are activated by ghrelin60.
area (VTA) that project to the nucleus accumbens (NAc) In leptin-deficient ob/ob mice, these neurons are strongly
and other forebrain areas53. With its supply of fibres from activated, and this effect has been functionally linked to
the pronounced hyperphagia in these mice55,61.
Adjacent to AGRP cells in the ARC are neurons that
express pro-opiomelanocortin (POMC) and release
melanocyte stimulating hormone (MSH), which
inhibits food intake by binding to and activating neuronal
melanocortin receptors. Unlike AGRP neurons, POMC
neurons are stimulated by leptin62,63 and they are inhibited
in leptin-deficient states62. However, caution is warranted
PVN
when generalizing the role of POMC neurons, as several
distinct subsets have been reported, some of which are
leptin-responsive64, whereas others are responsive to insu
Food lin. Surprisingly, whereas leptin depolarizes and increases
NTS intake
AGRP ARC
firing of POMC cells, insulin has the opposite effect 64.
Adiposity Nevertheless, insulin and leptin activate overlapping sig
signals
POMC
Satiety nal transduction and transcriptional cascades in POMC
signals neurons65, and the functional significance of the effects of
Leptin CCK
these hormones on membrane potential is unknown. Still
other POMC neuronal subsets are activated by ascending
Adipose tissue GI tract serotonergic input66 or by glutamatergic neurons located
in the ventromedial hypothalamic nucleus (VMN)67,
Figure 2 | Integration of long-term homeostatic andNature short-term satiety
Reviews signals.
| Neuroscience
whereas they are inhibited by GABAergic input from
A model describing homeostatic control of body adiposity proposes that regulation of
food intake on a mealtomeal basis is adjusted in response to changes in body fat AGRP neurons57.
content. Through actions in both the forebrain and hindbrain, the adiposity negative Weight loss activates AGRP neurons, which inhib
feedback signal leptin enhances responsiveness to gut-derived satiety signals such as its POMC neurons, and these effects are triggered at
cholecystokinin (CCK), which are released upon food ingestion. In addition to direct least in part by reduced leptin signalling. It is therefore
effects on hindbrain areas such as the nucleus of the solitary tract (NTS), leptin stimulates somewhat surprising that only a mild obesity pheno
pro-opiomelanocortin (POMC) neurons but inhibits neurons that express agouti-related type results when leptin receptors are deleted from both
protein (AGRP) and neuropeptide Y (labelled as just AGRP) in the hypothalamic arcuate POMC and AGRP neurons6769, whereas pan-hypotha
nucleus (ARC). These neurons project to second-order neurons in adjacent hypothalamic lamic leptin receptor deletion recapitulates the severe
nuclei, including the paraventricular nucleus (PVN) and lateral hypothalamic area (not obesity and hyperphagia phenotype of mice lacking
shown), which, in turn, project to the NTS, where satiety signals are processed. Satiety
leptin receptors altogether 70. These and other observa
signals activate vagal afferents that terminate in the NTS to promote the termination of a
meal. The NTS response to the satiety response is amplified both by direct input to the tions support the hypothesis that although leptin can
NTS from leptin and indirectly through the action of leptin in the hypothalamus. act directly on leptin receptors expressed by POMC and
Consequently, reduced leptin action (for example, following weight loss) increases meal AGRP neurons, these neurons can also be regulated
size by reducing the hindbrain response to satiety signals. GI, gastrointestinal. Figure through indirect effects mediated by leptin-responsive
from REF.4, Nature Publishing Group. neurons that are situated upstream.

NATURE REVIEWS | NEUROSCIENCE VOLUME 15 | JUNE 2014 | 369

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

are hypothesized to inhibit downstream neurocircuits


Food that are hardwired to drive feeding (perhaps including
intake Reward
AGRP neurons).
Recent observations point to an important and spe
cific role for GABA originating in AGRP neurons in the
Ventral control of feeding behaviour. Deletion of GABA signal
striatum
ling (by deletion of the vesicular GABA transporter)
NAc from AGRP neurons results in mice characterized by a
AMY VTA lean phenotype that are resistant to diet-induced obesity
(DIO)78. Perhaps more importantly, GABAergic projec
tions from AGRP neurons tonically inhibit a subset of
LHA neurons in the parabrachial nucleus (PBN) that, when
ARC
activated, trigger profound anorexia 79,80. These PBN
neurons, discussed in greater detail below, are marked
Leptin Ghrelin by expression of calcitonin and calcitonin gene-related
Hypothalamus peptide (CGRP), and their activation after the ablation
of AGRP neurons induces potentially fatal anorexia
Adipose tissue Stomach
inmice81.
Figure 3 | Integration of homeostatic and reward-related inputs. Homeostatic
Nature Reviews | Neuroscience Identification of neural circuits for feeding. The combi
signals modulate the perception of food reward (for example, the hedonic value and the
nation of mouse genetics with optogenetics or DREADD
motivation to work for food). Mesolimbic dopaminergic neurons in the ventral
tegmental area (VTA) project to the nucleus accumbens (NAc) and other brain areas to (designer receptor exclusively activated by a designer
heighten the reward value of palatable food. Neurons in the lateral hypothalamic area drug) technology has enabled investigation into the feed
(LHA) integrate reward-related input from the NAc with information related to energy ing effects induced by activating or inhibiting defined
homeostasis from arcuate nucleus (ARC) neurons. In turn, LHA neurons project to and neuronal subsets in live, conscious adult mice82,83. For
influence the mesolimbic dopaminergic system while also influencing satiety example, studies using optogenetic as well as pharmaco
perception through projections to the hindbrain (not shown). Weight loss lowers plasma genetic (DREADD-mediated) strategies show that AGRP
insulin and leptin levels while increasing plasma ghrelin levels. Working in concert, neuron activation is sufficient to rapidly and potently
these responses increase the rewarding properties of food and hence the motivation to stimulate feeding 8486. Moreover, this voracious feeding
eat through either direct effects in the ventral striatum or indirect effects in the
response occurs irrespective of nutritional state (that is,
hypothalamus through the LHA. Conversely, following periods of positive energy
in well-fed mice), time of day and without training, and
balance, body weight is returned to its biologically defended level through both a
decrease in the rewarding properties of food and an increased response to input from instead primarily depends on the level of AGRP neuron
satiety signals. AMY, amygdala. activity 84. Activation of these neurons also increases both
the motivation to work for food (as assessed by a progres
sive ratio test or lever pressing) and food-seeking behav
Although ARC neurons remain a major research iour 86, whereas selective inhibition of AGRP neurons
focus, leptin clearly influences energy balance through reduces feeding 84. Thus, hyperphagia associated with
effects in other brain areas both within the hypothala reduced leptin signalling (for example, in ob/ob or db/db
mus (for example, the VMN71) and outside the hypo mice, fasting and uncontrolled insulin-deficient diabe
thalamus (for example, the VTA72,73 and NTS40,74). With tes mellitus (uDM))8789 probably involves activation of
respect to neuronal subtypes that are important in lep AGRP neurons (FIG.4).
tin action, a role has been suggested for hypothalamic Recent research has shed light on the respective
GABAergic neurons (which are typically detected by roles of the three mediators contained within AGRP
the expression of the vesicular GABA transporter (also neurons: AGRP, NPY and GABA. Using DREADD
known as vesicular inhibitory amino acid transporter)). technology in three different mouse models, Lowell and
Deletion of leptin receptors from GABAergic neurons colleagues tested the individual contributions made by
causes a much more pronounced obesity phenotype75 GABA, NPY or AGRP to feeding induced by AGRP
than that induced by leptin receptor deletion from neuron activation90. They found that the presence of
AGRP and/or POMC cells69,76. A caveat to these obser either GABA or NPY is sufficient to rapidly stimu
Optogenetics
vations is that GABAergic neurons constitute a majority late feeding upon AGRP neuron activation. Although
A technique that uses light to of hypothalamic leptin-responsive cell types (including, AGRP release alone failed to increase food intake in
control the activity of specific for example, AGRP neurons), and hence leptin recep the short term, it did so effectively over a longer time
neurons in living tissue. tor deletion from these cells has a greater effect than interval90. Although the time course of their effects can
that which would be expected in a more select neu differ, release of any of the three mediators contained
DREADD
(Designer receptor exclusively ronal subpopulation. In this context, it is of interest that within AGRP neurons is sufficient to comparably and
activated by a designer drug). some GABAergic neurons also synthesize nitric oxide, potently stimulate feeding.
Gproteincoupled receptors and leptin receptor deletion from neurons that express Although stimulation and inhibition of POMC neu
that are modified for activation neuronal nitric oxide synthase (nNOS) causes severe rons using either optogenetics or DREADDs in mice
by binding to inert small
molecules that are used to
hyperphagia and obesity 77. These leptin-responsive reduces and increases food intake, respectively 84,85,
non-invasively control neuronal GABAergic neurons are found in both the ARC and dor these effects do not occur rapidly. Thus, POMC cells
signalling. somedial nucleus (DMN), and in response to leptin, they may primarily participate in long-term rather than

370 | JUNE 2014 | VOLUME 15 www.nature.com/reviews/neuro

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

short-term control of feeding 84,85. This observation fits Conversely, electrolytic lesioning of the PVN (like AGRP
with the delayed onset of feeding induced by local release neuron activation) has long been known to induce
of AGRP alone90, as AGRP acts by inhibiting neuronal hyperphagic obesity 91.
melanocortin receptors. The observation also implies Melanocortinreceptor4 (MC4R) mediates many of the
that the inhibition of POMC neurons by AGRP neu feeding effects of both AGRP and POMC neurons. The
ron activation (through type A GABA (GABAA) recep importance of this receptor in energy homeostasis was first
tors)57 does not explain the associated acute stimulation established by the obese phenotype of MC4Rknockout
of foodintake. mice92. Subsequent studies found that MC4R mutation
Studies to identify a feeding circuit activated by is the most common cause of monogenic obesity in
AGRP neurons have thus far focused on the hypotha humans93,94, accounting for up to 6% of early-onset or
lamic paraventricular nucleus (PVN) on the basis of severe adult obesity cases95. The PVN has been implicated
evidence that photoactivation of AGRP axons in this as a key site for the inhibitory effect of MC4R signalling
brain area mimics the feeding effect of activating AGRP on food intake, as MC4Rs are abundantly expressed in
neurons in the ARC85. The relevant PVN neurons (which this brain area96, the PVN is heavily innervated by both
are characterized by the expression of single-minded AGRP and POMC neurons97and food intake is reduced by
homologue 1 (SIM1)) seem to be inhibited by input administration of MC4R agonists directly into the PVN98.
from AGRP neurons, as activation of these PVN neu Furthermore, reexpression of MC4Rs in SIM1 (that is,
rons reduces food intake and reverses the hyperphagic PVN) neurons rescues hyperphagia in MC4Rnull mice99,
effect of AGRP neuron activation, including effects on whereas regulation of energy expenditure by MC4R is
both food seeking and willingness to work for food85. mediated by sympathetic preganglionic neurons100.

Reward

PVN

Satiety
POMC
NTS

nNOS
GABA Ghrelin

ARC AGRP Stomach

Adipose tissue Leptin

GABAAR LRb NPY1R or NPY5R


GHSR MC4R

Figure 4 | Neurocircuits involved in the homeostatic regulation of feeding. Neurons Naturein Reviews
the hypothalamic arcuate
| Neuroscience
nucleus (ARC) and nucleus of the solitary tract (NTS) sense and respond to peripheral energy signals to promote energy
homeostasis. Neuropeptides such as neuropeptide Y (NPY) and neurotransmitters such as GABA, among others, are
released onto downstream neurons including those in the paraventricular nucleus (PVN). In the PVN, oxytocin and other
neurons tonically inhibit feeding and, during energy deficit, are inhibited by orexigenic input from the ARC, thereby
stimulating feeding. The same agouti-related protein (AGRP) neurons (which co-express GABA and NPY) that are involved
in short-term feeding also contribute to long-term energy balance through the release of AGRP, an inverse agonist of
melanocortin receptor 4 (MC4R) and, through GABA release, inhibit neighbouring pro-opiomelanocortin (POMC)
neurons. POMC neurons are stimulated by input from leptin, and the release of melanocyte stimulating hormone
(MSH) activates MC4R, thereby inhibiting food intake. In addition, recent evidence also implicates leptin-responsive
GABAergic neurons that express neuronal nitric oxide synthase (nNOS) in the regulation of energy homeostasis. These
Neurotransmitters
Chemical messengers that are
neurons are found in the ARC and dorsomedial nucleus (not shown) and are hypothesized (dashed line) to inhibit
released by the end of a nerve downstream neurocircuits that drive feeding. Collectively, this input is relayed to the PVN and lateral hypothalamic area
fibre, causing an impulse to be (not shown) and integrated to modulate the rewarding properties of food and the response to satiety signals. GABAAR,
passed from once cell to type A GABA receptor; GHSR, growth hormone secretagogue receptor (ghrelin receptor); LRb, leptin receptor; NPY1R,
another. NPY receptor type 1.

NATURE REVIEWS | NEUROSCIENCE VOLUME 15 | JUNE 2014 | 371

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

Among several distinct subsets of PVN neurons that hypothalamus. Glucose-responsive neurons can either
are likely to participate in the control of food intake are increase or decrease their firing rate as local glucose
those that express oxytocin and seem to be components of concentrations rise, with the former termed glucose-
the pathway activated by leptin. Following either systemic excited and the latter referred to as glucose-inhibited
or central administration, oxytocin reduces food intake neurons121. These glucose-sensing neurons then pro
and body weight in obese as well as lean animals101104. ject to integrative centres that are located in the hind
This effect is mediated in part by projections to the NTS, brain and the hypothalamus, with VMN neurons being
where oxytocin release seems to enhance the hindbrain strongly implicated in efferent responses that act to raise
response to gut-derived satiety signals such as CCK, lead plasma glucose levels. Although the neurocircuitry that
ing to the consumption of smaller meals105. In support of mediates glucose counter-regulation remains poorly
this concept, leptin-responsive oxytocin neurons in the understood, both limbs of the autonomic nervous sys
PVN project to the NTS, and leptin-induced anorexia tem are potently activated in an organ-specific manner.
requires oxytocin signalling 106,107. Conversely, mice108 or Parasympathetic outflow increases through neurons in
humans109111 with genetic disruptions in SIM1 as well as the dorsal motor nucleus of the vagus that supply pan
individuals with PraderWilli syndrome112 are character creatic islets, and sympathetic outflow increases through
ized both by the loss of PVN oxytocin neurons and by the intermediolateral cell column of the spinal cord to
severe hyperphagia and obesity. This PVNNTS oxytocin stimulate glucose production by the liver and to activate
circuit therefore exemplifies how leptin-responsive hypo the adrenal medulla while also stimulating pancreatic
thalamic neurons can inhibit feeding by enhancing the islets122. Thus, glucopenia-induced glucagon secretion
hindbrain response to satiety signals, although (as noted is stimulated by the activation of both parasympathetic
above) leptin can also reduce meal size through direct and sympathetic neurons supplying theislet.
effects in the NTS40. Moreover, mice lacking either oxy A potent and sustained increase in food intake also
tocin113 or its receptor 114 exhibit only a modest, late-onset accompanies these responses (termed neuroglucopenic
obesity phenotype, and oxytocin neuron activation does feeding (also known as glucoprivic feeding)), and this
not reduce food intake after a fast85. Thus, the importance increase is so robust that it overrides control exerted by
of oxytocin neurons in energy homeostasis and obesity the energy homeostasis system. Stated differently, neuro
pathogenesis awaits furtherstudy. glucopenia stimulates feeding irrespective of body fuel
stores or plasma levels of leptin or insulin123 and, conse
Emergency circuits that stimulate feeding quently, obesity can result from repeated bouts of neuro
An emerging concept in the neurobiology of food intake glucopenia over a prolonged period. Indeed, the flawed
is that neurocircuits exist that are normally inhibited, notion that excess insulin is a cause of obesity stems in
but when activated in response to emergent or stress part from the early observation that pathological weight
ful stimuli they can override the homeostatic control of gain can be induced by repeated administration of insulin
energy balance. Understanding how these circuits inter at doses sufficient to cause hypoglycaemia124. However,
act with the energy homeostasis system is fundamental this effect results from neuroglucopenic feeding rather
to understanding the control of food intake and may than from insulinitself.
bear on the pathogenesis of disorders at both ends of From a teleological perspective, the notion that neuro
the body weight spectrum. glucopenic feeding should override the energy homeo
stasis system is logical in that maintaining stable body
Hypoglycaemia. Although reduced food intake accompa fat stores over time is of little use if plasma glucose levels
nies most stressful stimuli, inadequate delivery of glucose are too low to support brain function. Thus, hyperphagic
to the brain, termed neuroglucopenia (also known as feeding is sustained until glucopenia has resolved, at
glucoprivation), is an exception. The response to hypo which point the energy homeostasis system can reengage
glycaemia exemplifies how the emergency created by to offset any increase in body fat mass sustained in the
neuroglucopenia activates neurocircuits that drive feeding process of restoring euglycaemia.
along with wide-ranging neuroendocrine and autonomic A role for NPY in neuroglucopenic feeding has been
counter-regulatory responses that together function to suggested, as this response is blunted in NPY-deficient
raise blood glucose levels9,115. Components of the response mice125 and ARC NPY-containing neurons (AGRP neu
include: suppression of insulin secretion; secretion of rons) are activated by neuroglucopenia126. However,
counter-regulatory hormones such as glucagon and these neurons do not seem to be required for neuroglu
adrenaline that function in a coordinated manner with copenic feeding 127,128, as hypoglycaemia induces hyper
the activation of the hypothalamuspituitaryadrenal phagia in mice even after AGRP neurons have been
(HPA) axis; and increased sympathetic nervous system ablated127. Hindbrain catecholamine neurons that pro
outflow to liver, islet and adrenal tissue. The net effect of ject to the PVN may also drive neuroglucopenic feed
these responses is to raise serum glucose levels by stimu ing 119, as destruction of these neurons selectively blocks
lating glucose production while inhibiting glucose uptake this feeding response but leaves fasting-induced feeding
in peripheral tissues115118 (BOX1). responses intact 129. These and other findings suggest
Information regarding glucose availability is con that neuroglucopenic feeding can be induced by any of
veyed to the brain from multiple sources, including several components of a distributed neuronal system, at
the hepatic portal vein and glucose-sensing neurons least some of which are distinct from neurocircuits that
in both the hindbrain119 and forebrain120, including the drive hyperphagia after afast.

372 | JUNE 2014 | VOLUME 15 www.nature.com/reviews/neuro

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | Behavioural, autonomic and neuroendocrine responses to current or pending energy deficiency
In response to conditions of either
acute energy deficiency (for example, Reproduction Feeding Thermogenesis
hypoglycaemia) or chronic energy
deficiency (for example, leptin
Suppression
deficiency), the brain initiates a shared Brain of HPT axis
set of behavioural, autonomic and
neuroendocrine responses that are
designed to increase the availability of Activation
fuel to the CNS while also conserving of HPA axis
fuel stores. Among the responses are Glucagon
increases in both feeding and hepatic
glucose production (HGP), with the
Corticosterone
latter effect mediated in part by Insulin Leptin Glucose
increased plasma levels of glucagon
and corticosterone (through activation
of the hypothalamuspituitary Pancreas Adipose tissue Liver
adrenal (HPA) axis). At the same time,
the body conserves energy by
inhibiting energy-expensive processes HGP
such as growth and reproduction
while reducing metabolic rate through

the suppression of the hypothalamus Deciency of available or stored fuel
pituitarythyroid (HPT) axis. These
various responses are elicited by both Nature Reviews | Neuroscience
hypoglycaemia and uncontrolled insulin-deficient diabetes mellitus (uDM) despite the fact that blood glucose levels are
low in the former and high in the latter. Thus, one cause of hyperglycaemia in uDM is the pathological activation of the
same responses that are also observed in hypoglycaemia, presumably in response to deficient leptin input to the brain.
Consistent with this hypothesis, central leptin infusion restores euglycaemia in rats and mice with uDM135138. Similarly,
the behavioural, autonomic and neuroendocrine responses that are observed in hypoglycaemia and uDM are also elicited
in other leptin-deficient conditions (for example, fasting and in ob/ob mice); again, these responses are ameliorated by
administering leptin directly into the brain61,133. Thus, conditions associated with central leptin deficiency are characterized
by both hyperphagia and raised HGP, presumably reflecting activation of some of the same neurocircuits that are involved
in the counter-regulatory response to neuroglucopenia.
In summary, the CNS monitors signals that are pertinent to both immediately available fuel (for example, glucose) and the
status of fuel stores (for example, leptin), with deficiency of either hypothesized to activate the same neurocircuitry. The
result is a potent increase in both feeding and HGP until blood glucose (and leptin) levels have risen sufficiently to turn off
the response.

Diabetic hyperphagia. Type1 diabetes is a human disease seems to be driven by many of the same behavioural,
caused by autoimmune destruction of insulin-secreting autonomic and neuroendocrine responses that are
pancreatic -cells. In the absence of insulin treatment, elicited by neuroglucopenia9,132.
profound hyperglycaemia is accompanied by progressive Virtually all of these responses are also present in
weight loss because insulin deficiency impairs fat storage fasting 133 and ob/ob mice134, both of which, like uDM,
in adipose tissue. In the absence of insulin therapy, the are characterized by leptin deficiency. Together, these
resultant depletion of body fat causes deficiency of leptin observations suggest that the brain response to leptin
as well as insulin130. Recent experiments in rodent models deficiency recapitulates the response that is induced by
of uDM, in which hyperglycaemia is induced using the hypoglycaemia, a concept that seems paradoxical in that
-cell toxin streptozotocin (STZ), have begun to change plasma glucose levels are high in both uDM and ob/ob
our thinking about mechanisms underlying diabetic mice and low in fasting and hypoglycaemia. However,
hyperphagia. this view is consistent with a model in which the CNS
In addition to severe weight loss, hyperglycaemia response to a deficiency of immediately available fuel
and ketosis, uDM is accompanied by hyperphagia (for example, glucose) overlaps with the CNS response
and many of the same neuroendocrine and auto to depletion of stored body fuel (as reflected by deficient
nomic responses that are induced by neuroglucope leptin signalling). That is, the response to a deficiency of
nia (BOX1). These responses include increased plasma currently available fuel involves the same neurocircuits
levels of glucagon, catecholamines and corticoster that respond to a pending fuel deficiency; in each case,
one that collectively act to increase hepatic glucose the response functions to raise blood glucose levels
production 115,131. Moreover, sympathetic outflow to and thereby ensure an adequate supply of fuel to the
thermogenic brown adipose tissue is reduced in both brain. This concept is consistent with the observation
neuroglucopenia and uDM, and the reproductive, that intracerebroventricular leptin administration ame
growth and thyroid axes are inhibited in both con liorates each of these responses in rats and mice with
ditions as well. Therefore, in uDM, hyperglycaemia leptin deficiency induced by STZ-DM135138, whereas

NATURE REVIEWS | NEUROSCIENCE VOLUME 15 | JUNE 2014 | 373

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

restoring neuronal glucose availability reverses these body fat mass over time), and expansion of fat stores
conditions in neuroglucopenia. These observations sup is the bodys only way to increase plasma leptin levels.
port a model in which plasma glucose levels provide Accordingly, body fat mass will increase until input from
the brain with a crucial signal of immediately available leptin (combined with other adaptive changes, such as
fuel, whereas plasma leptin levels signal the amount the increased energy cost associated with increased body
of stored fuel (in the form of adipose tissue) and that weight) increases sufficiently to create a new steady state
deficiency of either triggers essentially the same brain in which body weight is once again stabilized, albeit at
response. This modification of the selfish brain hypo a raisedlevel.
thesis (REF.139) has interesting potential implications Consistent with this hypothesis, experimental activa
for obesity pathogenesis, and these are discussedbelow. tion of hypothalamic inflammatory pathways (such as
inhibitor of nuclear factor-B kinase-(IKK)nuclear
Implications for obesity pathogenesis. In normal-weight factor-B (NF-B) signalling) promotes hyperphagia
individuals, the energy homeostasis system robustly and weight gain, predisposes to DIO and blunts the
defends against both weight loss and weight gain140,141. anorectic effects of insulin and leptin160. Conversely,
The homeostatic response to weight loss has important interventions that reduce hypothalamic inflammation
clinical implications in that it explains how weight lost by can reduce food intake and body weight, and improve
dietary and/or lifestyle intervention tends to be regained hypothalamic insulin and leptin sensitivity 157,160.
over time in obese humans142,143. Conversely, available A recent study suggests that hypothalamic inflam
data from studies in rodents144 as well as humans145,146 mation induced by high-fat feeding involves neuronal
show that the energy homeostasis system also protects injury the basis of which remains unknown which
against pathological weight gain. Specifically, when in turn triggers an associated reactive gliosis (the recruit
weight is increased by involuntary (or forced) over ment and activation of microglia and astrocytes)159. In its
feeding, a dramatic reduction in caloric intake occurs early stages, this gliosis may be neuroprotective, but with
that is sustained until body weight returns to its pre- prolonged exposure to a high-fat diet, permanent damage
intervention level145. From this observation, we infer that to or loss of neurons involved in energy homeostasis (that
obesity does not arise simply from the passive accumu is, POMC neurons) can occur 159,161. Additional research
lation of excess body fat but rather is a state in which is needed to assess the causal nature of the relationship
the defended level of body fat has increased141. Indeed, between hypothalamic inflammation, gliosis and neuron
overweight individuals defend their increased body fat injury and the pathogenesis of DIO, and to determine
stores as robustly as lean individuals145,147149. Consistent whether these responses are reversible and/or can be
with this view, the main challenge confronting successful blocked by therapeutic intervention.
obesity treatment is that voluntary weight loss is resisted Regardless of the underlying mechanism, reduced
by homeostatic responses that eventually promote the neuronal leptin sensitivity seems to trigger adaptive
recovery of lostweight. responses that are by and large the same as those induced
One possible explanation for the biological defence by neuroglucopenia (BOX1). The predicted result is not
of increased body fat mass involves the phenomenon of only the biological defence of an increased level of body
leptin resistance. This hypothesis has its origin in the obser fat mass among obese, leptin-resistant individuals but also
vations that leptin levels are increased in obese individu the defence of increased blood glucose levels and an asso
als14 and that the ability of leptin to reduce food intake ciated impairment in glucose tolerance. Thus, if the brain
and body weight is blunted in most obese animals and leptin signal is not effectively transmitted to key hypo
humans18,150152. However, leptin resistance is a term that thalamic neurocircuits, increases in glycaemia as well as
should be used cautiously, as a reduced behavioural or food intake will occur until a new steady state is reached,
metabolic response to leptin can occur even when the one that is marked by delivery of leptin as well as glucose
response of cells and tissues to leptin is entirely normal in amounts sufficient to silence the adaptive response.
(for example, when the underlying defect lies downstream
of neurons that respond directly to leptin). Nevertheless, The neurobiology of stress-induced anorexia
evidence suggests that in rodent models of DIO, both the Reduced food intake is common to many types of stress
ability of leptin to cross the bloodbrain barrier and its whether being chased by a predator, in response to
capacity to activate neuronal leptin receptor signalling are trauma or illness, or following exposure to noxious sub
impaired153. stances. As the need to maintain body fat stores would
Although the cause of obesity-associated lep seem to be of secondary importance under such circum
tin resistance is unknown, inflammation, gliosis and stances, anorexia effectively overrides control of feeding
injury affecting hypothalamic neurons may have a by the energy homeostasis system. It is as if an off switch
role154158. Inflammation and gliosis are detected in the has been turned on, ensuring that feeding does not occur
rat or mouse ARC within the first few days of exposure until the threat haspassed.
to a high-fat diet, well before obesity develops159, and Palmiter and colleagues81 have recently identified a
these effects could favour weight gain by impairing the subset of neurons situated in the PBN that may function
Leptin resistance response of key neurons to insulin and leptin. In this as one such off switch for feeding. The PBN is a relay
A state in which the body is no
longer responsive to the
scenario, input from leptin would need to be increased station that receives both visceral and gustatory infor
anorexic effect of exogenous for normal energy homeostasis to occur (for exam mation and plays a part in protection against the con
leptin. ple, maintenance of neutral energy balance and stable sumption of unfamiliar or potentially toxic substances.

374 | JUNE 2014 | VOLUME 15 www.nature.com/reviews/neuro

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

Conditioned taste aversion In normal rodents, this protection manifests as neo Perhaps most importantly, the anorexia induced by
(CTA). A learned response of an phobia (for example, avoidance of a novel taste or food lithium chloride and LPS is blocked when PBN CGRP
animal to avoid repeated until its post-ingestive consequences are determined) neurons are inhibited81, whereas inhibition of these neu
ingestion of certain foods that and conditioned taste aversion (CTA)162. Thus, when con rons has no effect on intake in normal, unstressed ani
cause nausea or sickness.
fronted with a novel-tasting food with no previously mals; the latter finding fits with evidence that under
Cachexia known gustatory association, little is consumed until usual conditions, these neurons are already inhibited,
A condition that is the consequences of doing so are known, and foods at least in part by GABAergic input from AGRP neu
characterized by anorexia, that elicit an aversive experience will be avoided in the rons. Taken together, these findings suggest that one
weight loss and
future. These responses are presumed to have survival function of neurons in energy homeostasis circuits (for
disproportionate wasting of
muscle and adipose tissue.
value because they protect against the consumption of example, AGRP neurons) is to inhibit neurons in emer
potentially harmful foods162. gency feeding circuits (for example, CGRP neurons).
Lithium chloride and lipopolysaccharide (LPS) are However, when faced with a stress that is sufficient to
agents that, when administered to rodents, mimic the override this inhibition, feeding ceases until the stress
consumption of chemical and bacterial toxins, respec haspassed.
tively. As such, each induces anorexia and can support
CTA formation. The PBN is implicated in these responses Implications for disease states. Wasting illness is com
as both lithium chloride and LPS activate PBN neurons, mon not only in cancer but also in chronic infectious,
and lesions of the PBN prevent CTA formation163,164. inflammatory and other disorders. In these conditions,
Contained within the PBN are CGRP-expressing neu wasting is a major cause of morbidity and mortality 8, and
rons that project to the amygdala and are implicated as insight into its pathogenesis has proven elusive despite
mediators of anorexia that is induced by stressful stimuli81. decades of study. Empirically, wasting can be considered
The activity of these neurons increases in response to a state in which the energy homeostasis system does not
input from ascending projections from the caudal hind function properly, as food intake remains low irrespec
brain, although conversely, they are tonically inhibited by tive of the amount of weight lost 165. Furthermore, weight
GABAergic input from AGRP neurons7981. Using a com loss in normal individuals is typified by the preferential
bination of viral and genetic approaches, a recent study depletion of body fat, with relative sparing of lean mass,
showed that these PBN CGRP neurons are activated by whereas wasting illness is characterized as cachexia, which
both lithium chloride and LPS81, and that activation of is defined as progressive weight loss without sparing of
these neurons in unstressed mice (using either optogenet leanmass.
ics or DREADD technology) causes feeding to decrease With this background, the identification and char
or even to cease altogether. With repeated stimulation, acterization of CGRP neurons by the Palmiter group
animals continue to exhibit anorexia even when they are raises an important question: does chronic activation of
threatened with death from starvation7981 (FIG.5). this or a similar endogenous off switch for feeding drive

Anorexia

AMY
O switch

GABAAR
CGRP NTS

NMDAR
5-HT
ARC AGRP 5-HT3R
PBN
RMg and ROb
Emergency

Figure 5 | Activation of emergency neurocircuits that inhibit feeding. Neurocircuits exist that, when activated, can
override the homeostatic control of food intake. Recent work from the Palmiter laboratory suggests that calcitonin
gene-related peptide (CGRP) neurons expressed in the parabrachial nucleus (PBN) are an off switch that can trigger
Nature
anorexia in the context of emergency conditions (that is, illness, trauma or injury). The activity Reviews
of this | Neuroscience
neurocircuit is
constrained by inhibitory GABAergic input from agouti-related protein (AGRP) neurons in the arcuate nucleus (ARC), but
this inhibition can be overcome in response to trauma, illness or stress. Some PBN neurons express NMDA receptors
(NMDARs) that are activated by glutamatergic input from neurons in the rostral nucleus of the solitary tract (NTS), which in
turn are regulated by serotonergic input from neurons located in the raphe magnus (RMg) and raphe obscurus (ROb). The
net effect of activating this circuit is to activate CGRP neurons and thereby inhibit feeding. AMY, amygdala; GABAAR, type
A GABA receptor; 5HT, 5hydroxytryptamine (serotonin); 5HT3R, 5HT3 receptor.

NATURE REVIEWS | NEUROSCIENCE VOLUME 15 | JUNE 2014 | 375

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

the pathogenesis of wasting illness in diseases such as surgery can effectively induce and sustain weight loss,
cancer? Perhaps certain tumours (or the bodys response but the underlying mechanisms remain poorly under
to tumour cells) might result in the production of fac stood. Possibilities include both neural mechanisms (for
tors that activate CGRP neurons in the PBN, thereby example, activation of a gutbrain axis by rerouting or
causing inexorable, unrelenting weight loss that com accelerating the flow of intestinal nutrients) and humoral
pletely overrides the energy homeostasis system. If this mechanisms (for example, markedly increasing the secre
hypothesis is correct, silencing these neurons should tion of satiety-inducing gut peptides)166, but how these or
improve the wasting disorder or even induce its remis other such effects ultimately lower the defended level of
sion. We view testing of this hypothesis as an important body fat mass remains unclear.
scientific priority. Options for medical therapy increased following the
US Food and Drug Administration approval of both
Summary and conclusions lorcaserin (Belviq), a 5hydroxytryptamine (serotonin)
That robust and powerful systems ensure that the bodys 2C agonist, and Qsymia, a drug that combines phenter
ongoing energy demands are met comes as no surprise. mine (an adrenergic agonist) with the anticonvulsant
At the same time, carrying either too much or too little topiramate. Lorcaserin-induced weight loss is prob
body fat can be maladaptive. To address these competing ably mediated at least in part by activation of POMC
needs, the energy homeostasis system evolved to enable neurons167 and, although the drugs safety profile is
animals to match energy intake to energy expenditure favourable, efficacy is modest 168. Qsymia is a somewhat
over long time intervals and thereby ensure stability in more potent weight loss agent 169, but how topiramate
the amount of body energy stored as fat. However, under affects energy homeostasis neurocircuitry is unknown.
pathological conditions, activation of an unrelated set of Clearly, there is ample room for improvement where
neurocircuits can override control exerted by the energy treatment of obesity and other weight disorders is con
homeostasis system, leading to common and poten cerned, and efforts to delineate energy homeostasis and
tially serious disorders at either end of the body weight stress-responsive neurocircuitry have great potential to
spectrum. move the field forward. Recent technological advances
A key goal underlying efforts to delineate energy offer exciting new tools with which to accomplish these
homeostasis neurocircuits is to facilitate the discov goals and obtain information that will inform strategies
ery of effective new therapeutic modalities, but little for more effective approaches to the treatment of these
progress has been made in this effort to date. Bariatric common and debilitating disorders.

1. Edholm,O.G. Energy balance in man studies carried 13. Zhang,Y. etal. Positional cloning of the mouse obese 23. Bruning,J.C. etal. Role of brain insulin receptor in
out by the Division of Human Physiology, National gene and its human homologue. Nature 372, control of body weight and reproduction. Science 289,
Institute for Medical Research. J.Hum. Nutr. 31, 425432 (1994). 21222125 (2000).
413431 (1977). This study describes the mapping and sequencing 24. Batterham,R.L. etal. Gut hormone PYY336
2. Edholm,O.G., Fletcher,J.G., Widdowson,E.M. & of the gene that encodes the hormone leptin. physiologically inhibits food intake. Nature 418,
McCance,R.A. The energy expenditure and food 14. Considine,R.V. etal. Serum immunoreactive-leptin 650654 (2002).
intake of individual men. Br. J.Nutr. 9, 286300 concentrations in normal-weight and obese humans. 25. Turton,M.D. etal. A role for glucagon-like peptide1
(1955). N.Engl. J.Med. 334, 292295 (1996). in the central regulation of feeding. Nature 379,
3. Bray,G.A., Flatt,J.P., Volaufova,J., Delany,J.P. & 15. Schwartz,M.W., Peskind,E., Raskind,M., 6972 (1996).
Champagne,C.M. Corrective responses in human Boyko,E.J. & Porte,D.Jr. Cerebrospinal fluid 26. Gibbs,J., Young,R.C. & Smith,G.P. Cholecystokinin
food intake identified from an analysis of 7d food- leptin levels: relationship to plasma levels and to decreases food intake in rats. J.Comp. Physiol.
intake records. Am. J.Clin. Nutr. 88, 15041510 adiposity in humans. Nature Med. 2, 589593 Psychol. 84, 488495 (1973).
(2008). (1996). 27. Tschop,M., Smiley,D.L. & Heiman,M.L. Ghrelin
4. Schwartz,M.W. etal. Central nervous system control 16. Baskin,D.G., Breininger,J.F. & Schwartz,M.W. induces adiposity in rodents. Nature 407, 908913
of food intake. Nature 404, 661671 (2000). Leptin receptor mRNA identifies a subpopulation of (2000).
5. Morton,G.J., Cummings,D.E., Baskin,D.G., neuropeptide Y neurons activated by fasting in rat 28. Lam,T.K. Neuronal regulation of homeostasis by
Barsh,G.S. & Schwartz,M.W. Central nervous hypothalamus. Diabetes 48, 828833 (1999). nutrient sensing. Nature Med. 16, 392395 (2010).
system control of food intake and body weight. Nature 17. Elmquist,J.K., Bjorbaek,C., Ahima,R.S., Flier,J.S. 29. Strubbe,J.H. & Woods,S.C. The timing of meals.
443, 289295 (2006). & Saper,C.B. Distributions of leptin receptor mRNA Psychol. Rev. 111, 128141 (2004).
This comprehensive review provides a new isoforms in the rat brain. J.Comp. Neurol. 395, 30. West,D.B., Fey,D. & Woods,S.C. Cholecystokinin
conceptual understanding of how energy 535547 (1998). persistently suppresses meal size but not food intake
homeostasis is achieved. 18. Campfield,L.A., Smith,F.J., Guisez,Y., Devos,R. & in free-feeding rats. Am. J.Physiol. 246, R776R787
6. Saper,C.B., Chou,T.C. & Elmquist,J.K. The need to Burn,P. Recombinant mouse OB protein: evidence for (1984).
feed: homeostatic and hedonic control of eating. a peripheral signal linking adiposity and central neural 31. McMinn,J.E., Sindelar,D.K., Havel,P.J. &
Neuron 36, 199211 (2002). networks. Science 269, 546549 (1995). Schwartz,M.W. Leptin deficiency induced by fasting
7. Nguyen,D.M. & El-Serag,H.B. The epidemiology of 19. Halaas,J.L. etal. Physiological response to long-term impairs the satiety response to cholecystokinin.
obesity. Gastroenterol. Clin. North Am. 39, 17 (2010). peripheral and central leptin infusion in lean and Endocrinology 141, 44424448 (2000).
8. Braun,T.P. & Marks,D.L. Pathophysiology and obese mice. Proc. Natl Acad. Sci. USA 94, 32. Barrachina,M.D., Martinez,V., Wang,L., Wei,J.Y. &
treatment of inflammatory anorexia in chronic disease. 88788883 (1997). Tache,Y. Synergistic interaction between leptin and
J.Cachexia Sarcopenia Muscle 1, 135145 (2010). 20. Lee,G.H. etal. Abnormal splicing of the leptin cholecystokinin to reduce short-term food intake in
9. Ritter,R.C. & Slusser,P. 5Thio-D-glucose causes receptor in diabetic mice. Nature 379, 632635 lean mice. Proc. Natl Acad. Sci. USA 94,
increased feeding and hyperglycemia in the rat. Am. (1996). 1045510460 (1997).
J.Physiol. 238, E141E144 (1980). 21. Bagdade,J.D., Bierman,E.L. & Porte,D.Jr. The 33. Figlewicz,D.P., Stein,L.J., West,D., Porte,D.Jr &
This study provides evidence that the hindbrain is significance of basal insulin levels in the evaluation of Woods,S.C. Intracisternal insulin alters sensitivity to
important in responding to neuroglucopenia. the insulin response to glucose in diabetic and CCK-induced meal suppression in baboons. Am.
10. Kennedy,G. The role of depot fat in the hypothalamic nondiabetic subjects. J.Clin. Invest. 46, 15491557 J.Physiol. 250, R856R860 (1986).
control of food intake in the rat. Proc. R.Soc. Lond. B (1967). 34. Emond,M., Schwartz,G.J., Ladenheim,E.E. &
140, 578592 (1953). 22. Woods,S.C., Lotter,E.C., McKay,L.D. & Porte,D.Jr. Moran,T.H. Central leptin modulates behavioral and
11. Gao,Q. & Horvath,T.L. Neurobiology of feeding and Chronic intracerebroventricular infusion of insulin neural responsivity to CCK. Am. J.Physiol. 276,
energy expenditure. Annu. Rev. Neurosci. 30, reduces food intake and body weight of baboons. R1545R1549 (1999).
367398 (2007). Nature 282, 503505 (1979). 35. Williams,D.L., Baskin,D.G. & Schwartz,M.W.
12. Rosenbaum,M. & Leibel,R.L. Adaptive An early study demonstrating that insulin is an Evidence that intestinal glucagon-like peptide1 plays
thermogenesis in humans. Int. J.Obes. (Lond.) 34 adiposity signal that acts in the brain of non-human a physiological role in satiety. Endocrinology 150,
(Suppl. 1), 4755 (2010). primates to regulate food intake and body weight. 16801687 (2009).

376 | JUNE 2014 | VOLUME 15 www.nature.com/reviews/neuro

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

36. Hulsey,M.G., Lu,H., Wang,T., Martin,R.J. & 59. Spanswick,D., Smith,M.A., Mirshamsi,S., Routh,V.H. that suppresses appetite. Nature 503, 111114
Baile,C.A. Intracerebroventricular (i.c.v.) & Ashford,M.L. Insulin activates ATP-sensitive K+ (2013).
administration of mouse leptin in rats: behavioral channels in hypothalamic neurons of lean, but not In this paper, the authors identify a neural circuit
specificity and effects on meal patterns. Physiol. obese rats. Nature Neurosci. 3, 757758 (2000). that suppresses appetite.
Behav. 65, 445455 (1998). 60. Cowley,M.A. etal. The distribution and mechanism of 82. Kramer,R.H., Mourot,A. & Adesnik,H. Optogenetic
37. Kahler,A. etal. Chronic administration of OB protein action of ghrelin in the CNS demonstrates a novel pharmacology for control of native neuronal signaling
decreases food intake by selectively reducing meal size hypothalamic circuit regulating energy homeostasis. proteins. Nature Neurosci. 16, 816823 (2013).
in male rats. Am. J.Physiol. 275, R180R185 (1998). Neuron 37, 649661 (2003). 83. Packer,A.M., Roska,B. & Hausser,M. Targeting
38. McLaughlin,C.L. & Baile,C.A. Decreased sensitivity 61. Schwartz,M.W. etal. Specificity of leptin action on neurons and photons for optogenetics. Nature
of Zucker obese rats to the putative satiety agent elevated blood glucose levels and hypothalamic Neurosci. 16, 805815 (2013).
cholecystokinin. Physiol. Behav. 25, 543548 (1980). neuropeptide Y gene expression in ob/ob mice. 84. Aponte,Y., Atasoy,D. & Sternson,S.M. AGRP
39. Morton,G.J. etal. Leptin regulates insulin sensitivity Diabetes 45, 531535 (1996). neurons are sufficient to orchestrate feeding behavior
via phosphatidylinositol3OH kinase signaling in 62. Schwartz,M.W. etal. Leptin increases hypothalamic rapidly and without training. Nature Neurosci. 14,
mediobasal hypothalamic neurons. Cell. Metab. 2, pro-opiomelanocortin mRNA expression in the rostral 351355 (2011).
411420 (2005). arcuate nucleus. Diabetes 46, 21192123 (1997). The first use of optogenetics in the field of energy
40. Grill,H.J. etal. Evidence that the caudal brainstem is 63. Fan,W., Boston,B.A., Kesterson,R.A., Hruby,V.J. & balance, examining the effect of AGRP neuronal
a target for the inhibitory effect of leptin on food Cone,R.D. Role of melanocortinergic neurons in activation on feeding.
intake. Endocrinology 143, 239246 (2002). feeding and the agouti obesity syndrome. Nature 385, 85. Atasoy,D., Betley,J.N., Su,H.H. & Sternson,S.M.
41. Morton,G.J. etal. Leptin action in the forebrain 165168 (1997). Deconstruction of a neural circuit for hunger. Nature
regulates the hindbrain response to satiety signals. This paper was one of the first to identify the 488, 172177 (2012).
J.Clin. Invest. 115, 703710 (2005). melanocortin system and the MC4R in the control 86. Krashes,M.J. etal. Rapid, reversible activation of
42. Barrera,J.G., Sandoval,D.A., DAlessio,D.A. & of feeding behaviour. AgRP neurons drives feeding behavior in mice. J.Clin.
Seeley,R.J. GLP1 and energy balance: an integrated 64. Williams,K.W. etal. Segregation of acute leptin and Invest. 121, 14241428 (2011).
model of short-term and long-term control. Nature insulin effects in distinct populations of arcuate Another pioneering study using DREADD
Rev. Endocrinol. 7, 507516 (2011). proopiomelanocortin neurons. J.Neurosci. 30, technology to manipulate neurons involved in
43. Berthoud,H.R. Homeostatic and non-homeostatic 24722479 (2010). energy homeostasis.
pathways involved in the control of food intake and 65. Hill,J.W. etal. Direct insulin and leptin action on pro- 87. White,J.D. & Kershaw,M. Increased hypothalamic
energy balance. Obesity, 14, (Suppl. 5), 197S200S opiomelanocortin neurons is required for normal neuropeptide Y expression following food deprivation.
(2006). glucose homeostasis and fertility. Cell. Metab. 11, Mol. Cell Neurosci. 1, 4148 (1990).
44. Figlewicz,D.P. Adiposity signals and food reward: 286297 (2010). 88. Sipols,A.J., Baskin,D.G. & Schwartz,M.W. Effect of
expanding the CNS roles of insulin and leptin. Am. 66. Sohn,J.W. etal. Serotonin 2C receptor activates a intracerebroventricular insulin infusion on diabetic
J.Physiol. Regul. Integr. Comp. Physiol. 284, distinct population of arcuate pro-opiomelanocortin hyperphagia and hypothalamic neuropeptide gene
R882R892 (2003). neurons via TRPC channels. Neuron 71, 488497 expression. Diabetes 44, 147151 (1995).
This is one of the first reviews to describe the role (2011). 89. Wilding,J.P. etal. Increased neuropeptide-Y
of the adiposity hormones in the regulation of the 67. Sohn,J.W., Elmquist,J.K. & Williams,K.W. Neuronal messenger ribonucleic acid (mRNA) and decreased
reward system. circuits that regulate feeding behavior and neurotensin mRNA in the hypothalamus of the obese
45. Dossat,A.M., Lilly,N., Kay,K. & Williams,D.L. metabolism. Trends Neurosci. 36, 504512 (2013). (ob/ob) mouse. Endocrinology 132, 19391944
Glucagon-like peptide 1 receptors in nucleus 68. Gautron,L. & Elmquist,J.K. Sixteen years and (1993).
accumbens affect food intake. J.Neurosci. 31, counting: an update on leptin in energy balance. 90. Krashes,M.J., Shah,B.P., Koda,S. & Lowell,B.B.
1445314457 (2011). J.Clin. Invest. 121, 20872093 (2011). Rapid versus delayed stimulation of feeding by the
46. Mehta,S. etal. Regional brain response to visual food 69. van de Wall,E. etal. Collective and individual endogenously released AgRP neuron mediators GABA,
cues is a marker of satiety that predicts food choice. functions of leptin receptor modulated neurons NPY, and AgRP. Cell. Metab. 18, 588595 (2013).
Am. J.Clin. Nutr. 96, 989999 (2012). controlling metabolism and ingestion. Endocrinology 91. Leibowitz,S.F., Hammer,N.J. & Chang,K.
47. Nijs,I.M., Muris,P., Euser,A.S. & Franken,I.H. 149, 17731785 (2008). Hypothalamic paraventricular nucleus lesions produce
Differences in attention to food and food intake 70. Ring,L.E. & Zeltser,L.M. Disruption of hypothalamic overeating and obesity in the rat. Physiol. Behav. 27,
between overweight/obese and normal-weight females leptin signaling in mice leads to early-onset obesity, 10311040 (1981).
under conditions of hunger and satiety. Appetite 54, but physiological adaptations in mature animals 92. Huszar,D. etal. Targeted disruption of the
243254 (2010). stabilize adiposity levels. J.Clin. Invest. 120, melanocortin4 receptor results in obesity in mice. Cell
48. Figlewicz,D.P. & Sipols,A.J. Energy regulatory 29312941 (2010). 88, 131141 (1997).
signals and food reward. Pharmacol. Biochem. Behav. 71. Dhillon,H. etal. Leptin directly activates SF1 neurons 93. Vaisse,C., Clement,K., Guy Grand,B. & Froguel,P.
97, 1524 (2010). in the VMH, and this action by leptin is required for A frameshift mutation in human MC4R is associated
49. Fulton,S., Woodside,B. & Shizgal,P. Modulation of normal body-weight homeostasis. Neuron 49, with a dominant form of obesity. Nature Genet. 20,
brain reward circuitry by leptin. Science 287, 191203 (2006). 113114 (1998).
125128 (2000). 72. Fulton,S. etal. Leptin regulation of the 94. Yeo,G.S. etal. A frameshift mutation in MC4R
50. Farooqi,I.S. etal. Leptin regulates striatal regions mesoaccumbens dopamine pathway. Neuron 51, associated with dominantly inherited human obesity.
and human eating behavior. Science 317, 1355 811822 (2006). Nature Genet. 20, 111112 (1998).
(2007). 73. Hommel,J.D. etal. Leptin receptor signaling in 95. Farooqi,I.S. etal. Clinical spectrum of obesity and
51. Berthoud,H.R., Lenard,N.R. & Shin,A.C. Food midbrain dopamine neurons regulates feeding. mutations in the melanocortin 4 receptor gene.
reward, hyperphagia, and obesity. Am. J.Physiol. Neuron 51, 801810 (2006). N.Engl. J.Med. 348, 10851095 (2003).
Regul. Integr. Comp. Physiol. 300, R1266R1277 74. Hayes,M.R. etal. Endogenous leptin signaling in the 96. Mountjoy,K.G., Mortrud,M.T., Low,M.J.,
(2011). caudal nucleus tractus solitarius and area postrema is Simerly,R.B. & Cone,R.D. Localization of the
52. Stice,E., Figlewicz,D.P., Gosnell,B.A., Levine,A.S. & required for energy balance regulation. Cell. Metab. melanocortin4 receptor (MC4R) in neuroendocrine
Pratt,W.E. The contribution of brain reward circuits 11, 7783 (2010). and autonomic control circuits in the brain. Mol.
to the obesity epidemic. Neurosci. Biobehav. Rev. 37, 75. Vong,L. etal. Leptin action on GABAergic neurons Endocrinol. 8, 12981308 (1994).
2047 2058 (2012). prevents obesity and reduces inhibitory tone to POMC 97. Cowley,M.A. etal. Integration of NPY, AGRP, and
53. Kelley,A.E., Baldo,B.A., Pratt,W.E. & Will,M.J. neurons. Neuron 71, 142154 (2011). melanocortin signals in the hypothalamic
Corticostriatal-hypothalamic circuitry and food 76. Balthasar,N. etal. Leptin receptor signaling in POMC paraventricular nucleus: evidence of a cellular basis for
motivation: integration of energy, action and reward. neurons is required for normal body weight the adipostat. Neuron 24, 155163 (1999).
Physiol. Behav. 86, 773795 (2005). homeostasis. Neuron 42, 983991 (2004). 98. Giraudo,S.Q., Billington,C.J. & Levine,A.S. Feeding
54. Leinninger,G.M. etal. Leptin acts via leptin receptor- 77. Leshan,R.L., Greenwald-Yarnell,M., Patterson,C.M., effects of hypothalamic injection of melanocortin 4
expressing lateral hypothalamic neurons to modulate Gonzalez,I.E. & Myers,M.G.Jr. Leptin action receptor ligands. Brain Res. 809, 302306 (1998).
the mesolimbic dopamine system and suppress through hypothalamic nitric oxide 99. Balthasar,N. etal. Divergence of melanocortin
feeding. Cell. Metab. 10, 8998 (2009). synthase1expressing neurons controls energy pathways in the control of food intake and energy
55. Hahn,T.M., Breininger,J.F., Baskin,D.G. & balance. Nature Med. 18, 820823 (2012). expenditure. Cell 123, 493505 (2005).
Schwartz,M.W. Coexpression of Agrp and NPY in 78. Tong,Q., Ye,C.P., Jones,J.E., Elmquist,J.K. & 100. Rossi,J. etal. Melanocortin4 receptors expressed by
fasting-activated hypothalamic neurons. Nature Lowell,B.B. Synaptic release of GABA by AgRP cholinergic neurons regulate energy balance and
Neurosci. 1, 271272 (1998). neurons is required for normal regulation of energy glucose homeostasis. Cell. Metab. 13, 195204
56. Hagan,M.M. etal. Long-term orexigenic effects of balance. Nature Neurosci. 11, 9981000 (2008). (2011).
AgRP-(83132) involve mechanisms other than 79. Wu,Q., Boyle,M.P. & Palmiter,R.D. Loss of 101. Arletti,R., Benelli,A. & Bertolini,A. Influence of
melanocortin receptor blockade. Am. J.Physiol. Regul. GABAergic signaling by AgRP neurons to the oxytocin on feeding behavior in the rat. Peptides 10,
Integr. Comp. Physiol. 279, R47R52 (2000). parabrachial nucleus leads to starvation. Cell 137, 8993 (1989).
57. Cowley,M.A. etal. Leptin activates anorexigenic 12251234 (2009). 102. Arletti,R., Benelli,A. & Bertolini,A. Oxytocin inhibits
POMC neurons through a neural network in the 80. Wu,Q., Clark,M.S. & Palmiter,R.D. Deciphering a food and fluid intake in rats. Physiol. Behav. 48,
arcuate nucleus. Nature 411, 480484 (2001). neuronal circuit that mediates appetite. Nature 483, 825830 (1990).
A groundbreaking study identifying POMC neurons 594597 (2012). 103. Olson,B.R. etal. Oxytocin and an oxytocin agonist
in the ARC as being leptin-responsive and under An excellent paper identifying the PBN as an administered centrally decrease food intake in rats.
GABA inhibition. integration centre of visceral toxicity via hindbrain Peptides 12, 113118 (1991).
58. Spanswick,D., Smith,M.A., Groppi,V.E., glutamatergic signals and GABAergic homeostatic 104. Morton,G.J. etal. Peripheral oxytocin suppresses
Logan,S.D. & Ashford,M.L. Leptin inhibits feeding circuits. food intake and causes weight loss in diet-induced
hypothalamic neurons by activation of ATP-sensitive 81. Carter,M.E., Soden,M.E., Zweifel,L.S. & obese rats. Am. J.Physiol. Endocrinol. Metab. 302,
potassium channels. Nature 390, 521525 (1997). Palmiter,R.D. Genetic identification of a neural circuit E134E144 (2012).

NATURE REVIEWS | NEUROSCIENCE VOLUME 15 | JUNE 2014 | 377

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

105. Blevins,J.E. & Ho,J.M. Role of oxytocin signaling in 127. Luquet,S., Phillips,C.T. & Palmiter,R.D. NPY/AgRP A seminal study showing leptin replacement can
the regulation of body weight. Rev. Endocr. Metab. neurons are not essential for feeding responses to correct metabolic and endocrine abnormalities in
Disord. 14, 311329 (2013). glucoprivation. Peptides 28, 214225 (2007). humans with congenital leptin deficiency.
106. Blevins,J.E., Eakin,T.J., Murphy,J.A., 128. Corrin,S.E., McCarthy,H.D., McKibbin,P.E. & 151. El-Haschimi,K., Pierroz,D.D., Hileman,S.M.,
Schwartz,M.W. & Baskin,D.G. Oxytocin Williams,G. Unchanged hypothalamic neuropeptide Y Bjorbaek,C. & Flier,J.S. Two defects contribute to
innervation of caudal brainstem nuclei activated by concentrations in hyperphagic, hypoglycemic rats: hypothalamic leptin resistance in mice with diet-induced
cholecystokinin. Brain Res. 993, 3041 (2003). evidence for specific metabolic regulation of obesity. J.Clin. Invest. 105, 18271832 (2000).
107. Blevins,J.E., Schwartz,M.W. & Baskin,D.G. hypothalamic NPY. Peptides 12, 425430 (1991). 152. Heymsfield,S.B. etal. Recombinant leptin for weight
Evidence that paraventricular nucleus oxytocin 129. Ritter,S., Bugarith,K. & Dinh,T.T. Immunotoxic loss in obese and lean adults: a randomized,
neurons link hypothalamic leptin action to caudal destruction of distinct catecholamine subgroups controlled, dose-escalation trial. JAMA 282,
brainstem nuclei controlling meal size. Am. produces selective impairment of glucoregulatory 15681575 (1999).
J.Physiol. Regul. Integr. Comp. Physiol. 287, responses and neuronal activation. J.Comp. Neurol. 153. Myers,M.G.Jr., Leibel,R.L., Seeley,R.J. &
R87R96 (2004). 432, 197216 (2001). Schwartz,M.W. Obesity and leptin resistance:
108. Kublaoui,B.M., Gemelli,T., Tolson,K.P., Wang,Y. & 130. Havel,P.J. etal. Marked and rapid decreases of distinguishing cause from effect. Trends Endocrinol.
Zinn,A.R. Oxytocin deficiency mediates hyperphagic circulating leptin in streptozotocin diabetic rats: Metab. 21, 643651 (2010).
obesity of Sim1 haploinsufficient mice. Mol. reversal by insulin. Am. J.Physiol. 274, 154. Xu,H. etal. Chronic inflammation in fat plays a
Endocrinol. 22, 17231734 (2008). R1482R1491 (1998). crucial role in the development of obesity-related
109. Bonnefond,A. etal. Loss-of-function mutations in 131. American Diabetes Association. Standards of medical insulin resistance. J.Clin. Invest. 112, 18211830
SIM1 contribute to obesity and PraderWilli-like care in diabetes2011. Diabetes Care 34 (Suppl. 1), (2003).
features. J.Clin. Invest. 123, 30373041 (2013). 1161 (2011). 155. Weisberg,S.P. etal. Obesity is associated with
110. Holder,J.L.Jr, Butte,N.F. & Zinn,A.R. Profound 132. Kumaresan,P. & Turner,C.W. Effect of alloxan on feed macrophage accumulation in adipose tissue. J.Clin.
obesity associated with a balanced translocation that consumption and on replacement therapy with graded Invest. 112, 17961808 (2003).
disrupts the SIM1 gene. Hum. Mol. Genet. 9, levels of insulin in rats. Proc. Soc. Exp. Biol. Med. 122, 156. Hotamisligil,G.S. Inflammation and metabolic
101108 (2000). 526527 (1966). disorders. Nature 444, 860867 (2006).
111. Ramachandrappa,S. etal. Rare variants in single- 133. Ahima,R.S. etal. Role of leptin in the 157. Posey,K. etal. Hypothalamic proinflammatory lipid
minded 1 (SIM1) are associated with severe obesity. neuroendocrince response to fasting. Nature 382, accumulation, inflammation, and insulin resistance in
J.Clin. Invest. 123, 30423050 (2013). 250252 (1996). rats fed a high-fat diet. Am. J.Physiol. Endocrinol.
112. Swaab,D.F., Purba,J.S. & Hofman,M.A. Alterations 134. Bray,G.A., Fisler,J. & York,D.A. Neuroendocrine Metab. 296, E1003E1012 (2008).
in the hypothalamic paraventricular nucleus and its control of the development of obesity: understanding 158. De Souza,C.T. etal. Consumption of a fat-rich diet
oxytocin neurons (putative satiety cells) in Prader gained from studies of experimental animal models. activates a proinflammatory response and induces
Willi syndrome: a study of five cases. J.Clin. Front. Neuroendocrinol. 11, 128181 (1990). insulin resistance in the hypothalamus. Endocrinology
Endocrinol. Metab. 80, 573579 (1995). 135. Fujikawa,T., Chuang,J.C., Sakata,I., Ramadori,G. & 146, 41924199 (2005).
113. Camerino,C. Low sympathetic tone and obese Coppari,R. Leptin therapy improves insulin-deficient 159. Thaler,J.P. etal. Obesity is associated with
phenotype in oxytocin-deficient mice. Obesity 17, type 1 diabetes by CNS-dependent mechanisms in hypothalamic injury in rodents and humans. J.Clin.
980984 (2009). mice. Proc. Natl Acad. Sci. USA 107, 1739117396 Invest. 122, 153162 (2012).
114. Takayanagi,Y. etal. Oxytocin receptor-deficient mice (2010). A notable study postulating that neuronal injury
developed late-onset obesity. Neuroreport 19, 136. German,J.P. etal. Leptin activates a novel CNS due to high-fat feeding underlies the development
951955 (2008). mechanism for insulin-independent normalization of of obesity.
115. Beall,C., Ashford,M.L. & McCrimmon,R.J. The severe diabetic hyperglycemia. Endocrinology 152, 160. Zhang,X. etal. Hypothalamic IKK/NF-B and ER
physiology and pathophysiology of the neural 394404 (2011). stress link overnutrition to energy imbalance and
control of the counterregulatory response. Am. 137. Hidaka,S. etal. Chronic central leptin infusion obesity. Cell 135, 6173 (2008).
J.Physiol. Regul. Integr. Comp. Physiol. 302, restores hyperglycemia independent of food intake 161. Thaler,J.P., Guyenet,S.J., Dorfman,M.D.,
R215R223 (2012). and insulin level in streptozotocin-induced diabetic Wisse,B.E. & Schwartz,M.W. Hypothalamic
116. Gerich,J.E. etal. Prevention of human diabetic rats. FASEB J. 16, 509518 (2002). inflammation: marker or mechanism of obesity
ketoacidosis by somatostatin. Evidence for an 138. Lin,C.Y., Higginbotham,D.A., Judd,R.L. & pathogenesis? Diabetes 62, 26292634 (2013).
essential role of glucagon. N.Engl. J.Med. 292, White,B.D. Central leptin increases insulin sensitivity 162. Reilly,S. The parabrachial nucleus and conditioned
985989 (1975). in streptozotocin-induced diabetic rats. Am. J.Physiol. taste aversion. Brain Res. Bull. 48, 239254
117. Garber,A.J. etal. The role of adrenergic mechanisms Endocrinol. Metab. 282, E1084E1091 (2002). (1999).
in the substrate and hormonal response to insulin- 139. Peters,A. etal. The selfish brain: competition for 163. Reilly,S. & Trifunovic,R. Lateral parabrachial nucleus
induced hypoglycemia in man. J.Clin. Invest. 58, energy resources. Neurosci. Biobehav. Rev. 28, lesions in the rat: aversive and appetitive gustatory
715 (1976). 143180 (2004). conditioning. Brain Res. Bull. 52, 269278 (2000).
118. Rizza,R.A., Cryer,P.E. & Gerich,J.E. Role of 140. Schwartz,M.W. & Niswender,K.D. Adiposity 164. Reilly,S. & Trifunovic,R. Lateral parabrachial nucleus
glucagon, catecholamines, and growth hormone in signaling and biological defense against weight gain: lesions in the rat: neophobia and conditioned taste
human glucose counterregulation. Effects of absence of protection or central hormone resistance? aversion. Brain Res. Bull. 55, 359366 (2001).
somatostatin and combined - and -adrenergic J.Clin. Endocrinol. Metab. 89, 58895897 (2004). 165. Grossberg,A.J., Scarlett,J.M. & Marks,D.L.
blockade on plasma glucose recovery and glucose flux 141. Ryan,K.K., Woods,S.C. & Seeley,R.J. Central Hypothalamic mechanisms in cachexia. Physiol.
rates after insulin-induced hypoglycemia. J.Clin. nervous system mechanisms linking the consumption Behav. 100, 478489 (2010).
Invest. 64, 6271 (1979). of palatable high-fat diets to the defense of greater 166. Stefater,M.A., Wilson-Perez,H.E., Chambers,A.P.,
119. Ritter,S., Li,A.J., Wang,Q. & Dinh,T.T. Minireview: adiposity. Cell. Metab. 15, 137149 (2012). Sandoval,D.A. & Seeley,R.J. All bariatric surgeries
the value of looking backward: the essential role of 142. Knowler,W.C. etal. 10year follow-up of diabetes are not created equal: insights from mechanistic
the hindbrain in counterregulatory responses to incidence and weight loss in the Diabetes Prevention comparisons. Endocr. Rev. 33, 595622 (2012).
glucose deficit. Endocrinology 152, 40194032 Program Outcomes Study. Lancet 374, 16771686 167. Berglund,E.D. etal. Serotonin 2C receptors in pro-
(2011). (2009). opiomelanocortin neurons regulate energy and
120. McCrimmon,R.J. & Sherwin,R.S. Hypoglycemia in 143. Safer,D.J. Diet, behavior modification, and exercise: glucose homeostasis. J.Clin. Invest. 123, 50615070
type 1 diabetes. Diabetes 59, 23332339 (2010). a review of obesity treatments from a long-term (2013).
121. Routh,V.H. Glucose-sensing neurons: are they perspective. South Med. J. 84, 14701474 (1991). 168. Smith,S.R. etal. Multicenter, placebo-controlled trial
physiologically relevant? Physiol. Behav. 76, 144. Hagan,M.M. etal. Role of the CNS melanocortin of lorcaserin for weight management. N.Engl. J.Med.
403413 (2002). system in the response to overfeeding. J.Neurosci. 363, 245256 (2010).
122. Watts,A.G. & Donovan,C.M. Sweet talk in the brain: 19, 23622367 (1999). 169. Gadde,K.M. etal. Effects of low-dose, controlled-
glucosensing, neural networks, and hypoglycemic 145. Sims,E.A. etal. Experimental obesity in man. Trans. release, phentermine plus topiramate combination on
counterregulation. Front. Neuroendocrinol. 31, Assoc. Am. Physicians 81, 153170 (1968). weight and associated comorbidities in overweight
3243 (2010). 146. Deriaz,O., Fournier,G., Tremblay,A., Despres,J.P. & and obese adults (CONQUER): a randomised, placebo-
123. Dryden,S., Pickavance,L., Henderson,L. & Bouchard,C. Lean-body-mass composition and resting controlled, phase 3 trial. Lancet 377, 13411352
Williams,G. Hyperphagia induced by hypoglycemia in energy expenditure before and after long-term (2011).
rats is independent of leptin and hypothalamic overfeeding. Am. J.Clin. Nutr. 56, 840847 (1992). 170. Marx,J. Cellular warriors at the battle of the bulge.
neuropeptide Y. (NPY). Peptides 19, 15491555 147. Diaz,E.O., Prentice,A.M., Goldberg,G.R., Science 299, 846849 (2003).
(1998). Murgatroyd,P.R. & Coward,W.A. Metabolic
124. Russell-Jones,D. & Khan,R. Insulin-associated weight response to experimental overfeeding in lean and Acknowledgements
gain in diabetescauses, effects and coping strategies. overweight healthy volunteers. Am. J.Clin. Nutr. 56, This work was supported by a US National Institute of
Diabetes Obes. Metab. 9, 799812 (2007). 641655 (1992). Diabetes and Digestive and Kidney Diseases Grant to M.W.S
125. Sindelar,D.K. etal. Neuropeptide Y is required for 148. Leibel,R.L., Rosenbaum,M. & Hirsch,J. Changes in (DK090320, DK083042, and DK052989) and G.J.M.
hyperphagic feeding in response to neuroglucopenia. energy expenditure resulting from altered body (DK089053), the US National Institutes of Health (NIH)
Endocrinology 145, 33633368 (2004). weight. N.Engl. J.Med. 332, 621628 (1995). funded Nutrition Obesity Research Center (DK035816) and
126. Sergeyev,V., Broberger,C., Gorbatyuk,O. & 149. Sims,E.A. etal. Endocrine and metabolic effects of Diabetes Research Center (DK17047) at the University of
Hokfelt,T. Effect of 2mercaptoacetate and experimental obesity in man. Recent Prog. Horm. Res. Washington and a NIH Diabetes and Metabolism training
2deoxy-D-glucose administration on the 29, 457496 (1973). Grant (F32 DK097859; T32 DK0007247).
expression of NPY, AGRP, POMC, MCH and 150. Farooqi,I.S. etal. Effects of recombinant leptin
hypocretin/orexin in the rat hypothalamus. therapy in a child with congenital leptin deficiency. Competing interests statement
Neuroreport 11, 117121 (2000). N.Engl. J.Med. 341, 879884 (1999). The authors declare no competing interests.

378 | JUNE 2014 | VOLUME 15 www.nature.com/reviews/neuro

2014 Macmillan Publishers Limited. All rights reserved

Anda mungkin juga menyukai