Science Abl7398

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

RES EARCH

◥ to obesity development in mouse models and


REVIEW SUMMARY humans.

NEUROSCIENCE ADVANCES: Recent developments of high-


throughput single-cell and single-nucleus RNA-
Integrative neurocircuits that control metabolism sequencing methods have enabled the definition
of cellular subpopulations at unprecedented
and food intake molecular resolution. Applying these technol-
ogies has recently led to the identification of
Jens C. Brüning* and Henning Fenselau numerous additional food intake and metab-
olism regulatory neuronal and non-neuronal
cell populations in the hypothalamus. In par-
BACKGROUND: There is an ever-increasing pop- and neuronal inputs, signaling nutrient avail- allel, functional molecular systems approaches
ulation of overweight and obese individuals ability of the organism. The core of this hypo- have allowed the delineation not only of the
who exhibit the predisposition for a plethora thalamic control system comprises two neuronal functional role of these newly identified cell
of obesity-associated disorders, such as type 2 populations, which exert almost opposite func- types in metabolism control but also defini-

Downloaded from https://www.science.org at Universidade Estadual de Campinas on October 16, 2023


diabetes mellitus, cardiovascular diseases, tions in the regulation of feeding behavior, tion of the neuronal network organization as
certain types of cancers, as well as neuro- energy expenditure, and fuel metabolism. well as assessment of their activity in freely
degenerative disorders. Because both energy Agouti-related peptide (AgRP) neurons are behaving animals. These experiments revealed
homeostasis and peripheral metabolism are activated in conditions of energy deficit, are that metabolism regulatory neurons are mod-
coordinated through the brain, defining the inhibited by the fuel communication signals ulated across different timescales, including
basic neurobiological mechanisms of meta- leptin and insulin, and promote foraging and upon sensory perception of food cues, post-
bolic regulation and defining how alterations food consumption. Pro-opiomelanocortin ingestive signals that originate from the gastro-
in these pathways promote obesity develop- (POMC) neurons are activated in states of intestinal tract, and more long-term hormonal
ment and the onset of obesity-associated meta- positive energy balance and the associated mediators. The integration of these signals
bolic disorders are urgently needed to devise hormonal changes and reduce food intake serves to fine-tune metabolic adaptation and
therapeutic interventions for these prevalent and increase energy expenditure. Intense re- associated behaviors in an allostatic manner.
diseases. The arcuate nucleus (ARC) of the search over the past 20 years has revealed that These studies have largely advanced our knowl-
hypothalamus integrates multiple hormonal alterations in this circuitry are causally linked edge of the fundamental principles of central
nervous system–dependent control of me-
Astrocyte tabolism. They also allowed the definition of
new strategies to combat metabolic diseases.
TH These recent advances are highlighted in the
SST Hunger and Satiety
Review.
Glucose tolerance
MSH Insulin sensitivity OUTLOOK: Further expanding on these devel-
PNOC POMC opments will allow a more holistic insight
Motivational behaviour
into the conserved metabolism regulatory cell
NPY Proteostasis
Oxtr types and neurocircuits not only in rodent
3V AgRP Vglut2 Lipolysis models but also in humans. This new knowl-
Tanycytes Energy expenditure edge will aid the definition of how their de-
Blood
regulation is linked to the development of
vessel Microglia metabolic disturbances. Moreover, such studies
ARCUATE NUCLEUS will help clarify the mode of action of prom-
ising new anti-obesity therapeutics. These in-
Adipose
clude glucagon-like peptide-1 (GLP-1) receptor
Stomach (fat) agonists as well as newly developed polyago-
Pancreas Sensory Gut Hormonal
cues signal regulation nists for different receptors of gut-derived pep-
Small
tides, for which clinical studies have provided
intestine
evidence for promising efficacy in body weight
reduction and metabolic improvement. Fur-
thermore, deeper insights into the molecular
signature of metabolism-regulatory cell types
and into the synaptic mechanisms underlying
CREDIT: (MOUSE IMAGE) N. BURGESS/SCIENCE

their network interaction carry the potential to


nurture the development of alternative ther-
Time apeutic strategies for metabolic diseases.

Hypothalamic integration of food-related signals in metabolic control. Key hunger- and satiety- The list of author affiliations is available in the full article online.
*Corresponding author. Email: bruening@sf.mpg.de
promoting neuronal cell types in the hypothalamus integrate nutrient-related signals across different
Cite this article as J. C. Brüning, H. Fenselau, Science 381,
timescales: (i) upon sensory perception of food, (ii) post-ingestive gut-derived signals, and (iii) hormonal eabl7398 (2023). DOI: 10.1126/science.abl7398
signals that communicate the energy state of the organism. In addition to feeding, these neurocircuits also
adapt multiple behaviors and other physiological parameters in peripheral tissues in accordance to the READ THE FULL ARTICLE AT
energy state of the organism. https://doi.org/10.1126/science.abl7398

Brüning et al., Science 381, 1426 (2023) 29 September 2023 1 of 1


RES EARCH

◥ dicted that the receptors of energy state–


REVIEW sensing hormones would be expressed on
key neurons that adapt food intake and energy
NEUROSCIENCE expenditure in a feedback regulatory mecha-
nism. Although it took another three decades
Integrative neurocircuits that control metabolism to identify the fundamental energy-sensing
signal as leptin, the breakthrough discovery of
and food intake this hormone and its receptor mainly in the
CNS has opened a new era in the science of
Jens C. Brüning1,2,3,4* and Henning Fenselau2,3,5 CNS-dependent control of energy balance and
metabolism (4). Defining the underlying neuro-
Systemic metabolism has to be constantly adjusted to the variance of food intake and even be prepared circuitry targeted by leptin and other energy-
for anticipated changes in nutrient availability. Therefore, the brain integrates multiple homeostatic communicating signals—such as insulin, ghrelin,
signals with numerous cues that predict future deviations in energy supply. Recently, our understanding and glucagon-like peptide-1 (GLP-1)—has al-
of the neural pathways underlying these regulatory principles—as well as their convergence in the lowed us to begin unraveling the central mecha-
hypothalamus as the key coordinator of food intake, energy expenditure, and glucose metabolism—have nisms that underlie not only feeding behavior

Downloaded from https://www.science.org at Universidade Estadual de Campinas on October 16, 2023


been revealed. These advances have changed our view of brain-dependent control of metabolic and energy expenditure but also metabolic sub-
physiology. In this Review, we discuss new concepts about how alterations in these pathways contribute strate utilization in peripheral tissues, which
to the development of prevalent metabolic diseases such as obesity and type 2 diabetes mellitus and ultimately control body weight as well as stable
how this emerging knowledge may provide new targets for their treatment. glucose and lipid metabolism (Fig. 1) (5, 6).
Defining the exact nature of this regulation

O
has become of utmost societal importance be-
rganismal survival and health require stasis to that of allostasis, which was formal- cause deregulated body weight control is in-
the maintenance of energy stores, water, ized more recently by the neuroscientist Peter creasingly affecting global populations. There
salt, and temperature within specific Sterling (2). is an ever-increasing population of overweight
physiological ranges. The basic princi- The maintenance of stable body weight was and obese individuals, which exhibit predis-
ples of homeostasis have been outlined mainly viewed under the assumptions of position for a plethora of obesity-associated
as early as the late 19th century, when the homeostatic regulation. Concepts such as lipo- disorders, such as type 2 diabetes mellitus,
physiologist Claude Bernard proposed that static control of body weight have been put cardiovascular diseases, certain types of can-
critical physiological parameters have to be forth as early as the 1950s, when the physiol- cers, as well as neurodegenerative disorders
regulated within a defined physiological range ogist Gilbert Kennedy proposed that energy (7). Therefore, defining the basic neurobio-
to ensure survival and organismal integrity. homeostasis is regulated through hormonal logical mechanisms of metabolic regulation
He provided evidence that the central nervous feedback (using then unidentified hormones), (Box 1 and Fig. 2) and defining how alterations
system (CNS) plays an important role in this which would communicate the energy state of in these pathways promote obesity development
process. In the 1930s, the physiologist Walter the organism to the CNS (Fig. 1) (3). He pre- and obesity-associated metabolic disorders are
Cannon further specified the formal concept
of homeostasis, which thereafter has been the
conceptual framework for numerous regula-
tory principles in physiology (1). Nevertheless, 1849 1953 1970s 1994 RECENT YEARS
the proposed concept of homeostasis—that a
variable parameter in physiology is monitored
and that deviations from its setpoint elicit PVH
counterregulatory responses in control systems— Glucose
does not provide ideal regulatory precision
to maintain a constant, steady-state level of a Blood vessel VMH
ARC
regulated parameter because it requires devia-
tions to occur before counterregulation sets in. The brain controls glucose
Thus, to optimize maintenance of a stable metabolism in the periphery Hypothalamic nuclei are GLP-1
milieu, integrating anticipated changes of the Insulin
regulated parameter provides higher physio- body weight and glucose Ghrelin
logical stability. Incorporating the parameters
of environmental influences and anticipated ? Leptin
Periphery
changes has broadened the concept of homeo-
Leptin
1
Department of Neuronal Control of Metabolism, Max Planck
Institute for Metabolism Research, 50931 Cologne, Germany. Dissection of complex
2
Policlinic for Endocrinology, Diabetes, and Preventive Hormones from the periphery neurocircuitry in feeding
Medicine (PEDP), University Hospital Cologne, 50924 control feeding states The discovery of leptin and systemic metabolism
Cologne, Germany. 3Excellence Cluster on Cellular Stress
Responses in Aging Associated Diseases (CECAD) and
Center of Molecular Medicine Cologne (CMMC), University of
Cologne, 50931 Cologne, Germany. 4National Center for Fig. 1. Timeline of discoveries detailing the brain mechanisms that underlie metabolic control. The
Diabetes Research (DZD), 85764 Neuherberg, Germany. timeline shows key conceptual advances in our understanding of CNS-dependent control of body weight and
5
Research Group Synaptic Transmission in Energy metabolism. Breakthrough discoveries, such as the identification of leptin and the melanocortin
Homeostasis, Max Planck Institute for Metabolism Research,
50931 Cologne, Germany. neurocircuitry, have had implications for many areas of investigations, including the development of
*Corresponding author. Email: bruening@sf.mpg.de therapeutics for obesity and metabolic diseases.

Brüning and Fenselau, Science 381, eabl7398 (2023) 29 September 2023 1 of 10


RES EARCH | R E V I E W

projections into hypothalamic and extrahypo-


Box 1. Technical advances that facilitate defining the neurocircuitry of metabolic control. thalamic regions, where they engage down-
stream neural pathways through fast-acting
The tremendous complexity arising from the particularly high level of cellular heterogeneity and synaptic neurotransmitters and neuropeptides for met-
connectivity in neurocircuits that underlie homeostatic control have made functional investigations inherently abolic coordination. Owing to their localiza-
difficult. Advances in Cre/loxP technologies have provided an extended toolbox of approaches that enable tion in the ventral part of the hypothalamus,
anatomical and functional interrogations of defined cell types in the CNS. Although these techniques have AgRP and POMC neurons have the remark-
propelled the identification and detailed characterization of neurocircuits, critical limitations remain; the able ability to precisely detect fluctuations in
techniques are often based on a single, previously identified marker gene. hormone and nutrient levels in the blood-
Recent developments of high-throughput single-cell and single-nucleus sequencing methods have enabled stream (13). It was assumed that this is due
the definition of cellular subpopulations at unprecedented molecular resolution (25, 154). These approaches to a more porous blood-brain barrier, but a
have revealed that numerous transgenes that were previously considered specific to a certain cell exhibit new participant in the relay of hormonal and
more widespread distribution in further types and subtypes of cells, adding caution regarding the previously nutritional signals from circulation to these
assumed specificity of Cre-mediated targeting of neurocircuits on the basis of a single marker gene (Fig. 2). neurons has been revealed: tanycytes (14).
Although these data have provided us with exciting new knowledge, a major task for future functional These are specialized radial glia cells that line
investigations will be to advance bioinformatic tools that enable integration of these multiple emerging the third ventricle and make contact with

Downloaded from https://www.science.org at Universidade Estadual de Campinas on October 16, 2023


datasets into standardized molecular atlases of brain regions to provide a unified nomenclature and blood vessels in the median eminence (ME)
assignment of cell types. Further, advances in spatial transcriptomic analyses and tools that enable the and nearby neurons in the mediobasal hypo-
integration of this spatial information with single-cell sequencing information will be required. Notably, thalamus. Tanycytes regulate GLP-1 and insu-
because single-nucleus sequencing can be performed on rapidly frozen tissue, it allows the capture of lin access to the ARC, although their role in
state-dependent changes in transcriptional regulation (56). Therefore, single-nucleus sequencing studies leptin transport is still a matter of debate
carry the potential to identify new neural cell types involved in metabolic control in an unbiased manner (15–18). Notably, deleting the insulin receptor
(Fig. 2). Last, recent developments in transgenic technologies that use combinatorial recombinase such as specifically from tanycytes in mice mimicked
Dre/rox– and Flp/frt–dependent recombination together with Cre/loxP–mediated recombination allows the insulin resistance that occurs in obesity
intersectional genetic targeting of specific subtypes of cells that are characterized by multiple molecular mouse models, which in turn altered the ac-
markers (Fig. 2) (47, 155, 156). These emerging developments will further our understanding of the tivity of AgRP neurons to regulate feeding
functional organization of metabolism regulatory neurocircuits at the level of highest cellular granularity. and glucose homeostasis (15). Thus, future re-
search should define the molecular and func-
tional organization of this cell type in humans
to characterize them as potential targets in
urgently needed to devise therapeutic interven- ral tissues in preparation for increasing nu- the treatment of metabolic diseases. In ad-
tions for these prevalent diseases. trient availability. Last, we discuss how this dition to tanycytes, a wide area of research
Additional evidence that obesity originates knowledge has led to the recent development has more recently unraveled a critical role for
through altered signaling in the CNS has of new therapeutics for obesity and metabolic other non-neuronal cell types in the hypo-
arisen from human genetics data, according to diseases. thalamus in control of energy metabolism reg-
which most gene variants in obesity are pre- ulatory neurocircuits. These include astrocytes,
dominantly expressed in the CNS (8). Further, Neural populations that control which themselves sense metabolism regula-
monogenetic defects in human obesity syn- systemic metabolism tory hormones, such as insulin and leptin
dromes also cluster in genes whose products The melanocortin circuitry is a prototypic reg- (19, 20), and their activity regulates neighbor-
act in the highly conserved hypothalamic neu- ulatory pathway in homeostatic control of sys- ing neurons through various mechanisms,
rocircuitry targeted by leptin in mice and rats temic metabolism (9). The core of this system such as neurotransmitter release as well as
(9). Last, defining the neurocircuitry-dependent comprises two neuronal populations, which ensheatment of metabolism-regulator neurons
regulatory principles underlying systemic en- reside in the arcuate nucleus (ARC) of the (21). Similarly, oligodendrocytes and microglia
ergy and glucose homeostasis as well as their hypothalamus and exert almost opposite func- have been identified as regulators of hypothal-
deregulation in obesity prevents the wide- tions in the regulation of feeding behavior, amic neuronal function and their deregulation
spread stigmatization of patients with obesity energy expenditure, and fuel metabolism. Agouti- in obesity (22, 23). However, how non-neuronal
as suffering from a simple lack of will power related peptide (AgRP) neurons are activated cells affect central metabolism-regulatory
to control their food intake and hence body in conditions of energy deficit, are inhibited by neurocircuits has been elegantly reviewed
weight (10). the fuel-communication signals leptin and in- elsewhere and is beyond the scope of this
In this Review, we discuss the recent ad- sulin, and promote foraging and food consump- Review (24).
vances in our understanding of the neurobio- tion. Pro-opiomelanocortin (POMC) neurons Several other neuronal populations in the
logy that underlies the regulation of food intake, are activated in states of positive energy bal- ARC have been identified as additional reg-
energy balance, and systemic glucose metabo- ance and the hormonal changes associated ulators of systemic metabolism. Initial ob-
lism. This includes molecularly distinct neuro- with it, reduce food intake, and increase energy servations of the ARC revealed that it is an
nal populations in energy-regulating neuronal expenditure. The tight interaction of AgRP and exceptionally heterogeneous hypothalamic
centers in mice and new developments in POMC neurons in control of feeding behavior nucleus, which contains phenotypic markers
human genetics to define conserved regula- had already been predicted on the basis of the for various neurotransmitters and neuropep-
tory pathways in metabolic control. These dis- orexigenic (feeding-promoting) effect of neuro- tides. Subsequent single-cell and single-nucleus
coveries have also broadened our understanding peptide Y (NPY) as well as the demonstration of RNA-sequencing approaches have enabled an
of CNS-dependent control of integrative physi- a dense network of NPY-immunoreactive axons in-depth cellular analysis of the ARC in mice
ology beyond the concept of homeostatic reg- and axon terminals in close apposition with and pinpointed new cellular substrates underly-
ulation, highlighted by recent findings that b endorphin–immunoreactive neurons through- ing metabolic control (Fig. 3) (25, 26).
food-predicting cues rapidly engage neurocir- out the medial basal hypothalamus (11, 12). Both g-Aminobutyric acid (GABA) is the most
cuits to adapt metabolic pathways in periphe- of these ARC neurons show a large number of abundant fast-acting neurotransmitter in the

Brüning and Fenselau, Science 381, eabl7398 (2023) 29 September 2023 2 of 10


RES EARCH | R E V I E W

jects to and synaptically inhibits PVH neurons


A SINGLE-NUCLEUS SEQUENCING
through GABA release (25). Chemogenetic
Beads Oil stimulation of SST-ARC neurons potently in-
Nuclei Library preparation creases feeding in mice. However, single-cell
capture and sequencing RNA-sequencing analyses of the ARC revealed
Hypothalamus Nuclei that SST-ARC neurons exhibit a transcriptional
profile that is very similar to that of AgRP-ARC
B CELL CLUSTERING AND IDENTIFICATION neurons (Fig. 3) (25). Thus, further experiments
will have to define the specific role of AgRP-
negative SST-ARC cells in feeding regulation.
UMAP of the hypothalamus Gene X A third GABAergic population was identi-
fied through an unbiased approach for molec-
Gene Y
ular profiling of activated neurons (34). This
study demonstrated that short-term (3 days)
C STATE-DEPENDENT SIGNATURE
exposure of mice to a high-fat diet (HFD) ac-
Energy deprived Activation signature tivates ARC neurons that express the neuro-

Downloaded from https://www.science.org at Universidade Estadual de Campinas on October 16, 2023


Steady state Obese Fos Noct Gem
peptide prepronoceptin (PNOC) (35). Selective
assessment of PNOC-ARC neurons showed
Circadian
that their activation promotes feeding, whereas
their ablation protects from overconsumption
D SPATIAL ALLOCATION (hyperphagia) and body weight gain upon HFD
Alignment and decoding feeding (35). Thus, PNOC-ARC neurons are a
Tissues key population for acutely controlling feed-
Gene X ing behavior and obesity development when
Gene Y
energy-dense, highly palatable food is ac-
Array cessible. Circuit-mapping studies established
that PNOC-ARC neurons synaptically inhibit
E MANIPULATIONS WITH NEUROSCIENCE TOOLS nearby POMC neurons through GABA (35).
Cre
X Gene X This is notable because dynamics in ARC-
Cre Multiple
X Targeted derived GABAergic synaptic input has been
Dre feature
subpopulation implicated in determining POMC neuron ac-
sa2
6 targeting Dre
Ro Gene Y
AAVs and/or Transgenics tivity in response to changes in nutrient avail-
ability (Fig. 3) (36–39). More recently, elegant
loxP loxP rox rox studies have revealed a function of non-AgRP,
Rosa26
NPY-expressing neurons in the ARC to control
promoter Stop Stop Transgene feeding under positive energy balance (40).
loxP loxP FRT FRT Important unsettled questions at present are
Intron
Virus gene Trans to which extent these ARC populations over-
INTERSECT promoter
lap, what central and circulating signals me-
diate the energy state–dependent activity
Fig. 2. Defining a cell’s function in metabolic control. (A) Workflow shows the use of droplet-based single- regulation of these GABAergic neurons, and
nucleus RNA-sequencing to capture and measure the expression levels of genes in discrete cell types. Nuclei can how dysregulation of these circuit mecha-
easily be isolated from either fresh or frozen tissue, and transcriptomic profiles of thousands to millions of nisms could relate to increased susceptibility
individual cells can be obtained. (B) Visualization of cell heterogeneity and expression of marker genes in to metabolic disorders.
annotated cell types is achieved through uniform manifold approximation and projection (UMAP) embedding. For many years, it was assumed that POMC
(C) Nuclei collection in different metabolic states provides a snapshot of transcriptomic profiles of numerous neurons promote meal termination because
genes, including cell-specific markers that reflect activity levels. (D) Measuring and mapping gene activity in the genetic deficiency of POMC or lack of POMC
spatial context with recent transcriptomic approaches allow the identification of a cell’s location. (E) A suite of neurons result in hyperphagia and severe
new viruses and recombinase driver lines expands the ability to target chemo- or optogenetic tools to identified obesity in humans and mice (41–44). How-
cell types on the basis of multiple genetic factors for manipulation studies. ever, acute opto- or chemogenetic manipu-
lations of POMC neurons have repeatedly
been found to have no or minimal effects on
ARC. Consistent with its relevance as a feeding- chronically silencing them attenuates food in- short-term feeding behavior in mice (41, 45–48),
promoting signal released from AgRP neurons take and body weight gain. These effects are pro- which indicates that POMC neurons are more
(27–29), additional orexigenic, GABA-releasing posed to be driven by GABAergic inhibition of important for long-term regulation of energy
ARC populations have been identified. A do- downstream satiety neurons, including those balance, and/or points to the possibility that
paminergic, tyrosine hydroxylase (TH)–expressing located in the paraventricular hypothalamus subsets of POMC neurons may have distinct
population (30) was originally classified as a (PVH) (30). Because GABAergic inhibition of PVH feeding regulatory functions. Given that the
purely neuroendocrine cell type whose dopa- satiety neurons by AgRP neurons also controls fast, feeding promoting action of orexigenic
mine release coordinates the secretion of the feeding behavior (27), these findings suggest ARC neurons is mediated by rapid inhibition
principal lactogenic hormone prolactin from that this inhibitory pathway is a common fea- of downstream satiety neurons (27), a rapidly
the pituitary (31–33). However, when optogenet- ture of orexigenic, GABA-releasing ARC neurons. acting satiety neuron, if it exists, would pre-
ically stimulated in mice, ARC-TH neurons evoke Another orexigenic ARC population is marked sumably release a fast-acting excitatory neu-
a rapid increase in feeding (30). Correspondingly, by somatostatin (SST) expression, which pro- rotransmitter. In this context, a glutamatergic

Brüning and Fenselau, Science 381, eabl7398 (2023) 29 September 2023 3 of 10


RES EARCH | R E V I E W

trast to what is observed upon chemogenetic


Food-predicting cues Hunger and Satiety stimulation of the entire POMC-ARC popula-
tion, selective activation of Glp-1r–expressing
Hypothalamus
Appetitive and but not Lepr-expressing POMC neurons de-
Sight, smell, and aversive behavioral creased feeding in a more rapid manner, which
taste of food adjustment is consistent with previous studies that revealed
that the food intake–suppressing effect of
ARCUATE GLP-1 is at least in part mediated by POMC
Gastric and PRE- Glucose tolerance
NUCLEUS
intestinal ABSORPTIVE neurons (47, 57). Thus, further definition of
distension functionally relevant subsets of previously con-
Astrocyte Insulin sensitivity
sidered homogenous metabolism regulatory
neurons is warranted, which could also define
Absorbed TH targets for therapeutic intervention (58).
nutrients SST GABA
POST- GABA
INGESTIVE Lipolysis Integration of sensory food cues
Gut hormones MSH
PNOC
POMC In addition to internal feedback regulatory
GABA

Downloaded from https://www.science.org at Universidade Estadual de Campinas on October 16, 2023


mechanisms communicating systemic energy
NPY Proteostasis
Adipocyte, status to the CNS, integration of signals that
AgRP Oxtr
pancreatic, LONG-TERM 3V Vglut2 predict future nutrient uptake are pivotal to
GABA
and hepatic FEEDBACK maintaining a stable metabolic state. Environ-
hormones Energy expenditure mental cues associated with food consump-
Tanycytes Microglia
tion as well as with seeing, smelling, or tasting
food elicit a myriad of physiological processes,
Fig. 3. ARC-based neurocircuits in control of metabolism. The arcuate nucleus of the hypothalamus
including saliva production, secretion of diges-
(ARC), a small nucleus located at the lower surface of the brain, has emerged as a key regulator of systemic
tive enzymes, and release of hormones such as
metabolism. Intense research over the past two decades has revealed that Agouti-related peptide (AgRP)
insulin (59, 60). These feed-forward, anticipa-
and proopiomelanocortin (POMC) neurons, which constitute key components of the melanocortin circuitry, exert
tory mechanisms, often referred to as cephalic
almost exclusive opposite regulatory functions of various physiological processes. In addition, recent studies
phase responses, ensure that consumed food
have established several additional ARC neurons as regulators of metabolism: dopaminergic, tyrosine hydroxylase
is rapidly digested and absorbed and that nu-
(TH)–expressing, somatostatin (SST)–expressing, prepronoceptin (PNOC)–expressing, and oxytocin receptor
trients are efficiently metabolized and removed
(Oxtr)–expressing ARC neurons. ARC neurons have the ability to sense alterations in circulating levels of hormonal
from circulation (61). In addition, cues that
and nutritive factors, which constantly adjust their neural activity to the internal state of the organism. Recent
anticipate food consumption are linked to ap-
activity recordings have further uncovered that cues predicting future food consumption and nutrient uptake as
petitive and aversive behavioral adjustments,
well as gut-derived, postingestive signals adapt the firing properties of discrete ARC neuron populations.
hunger and satiety regulation, and preferences
for certain food items. In contrast to the slow
population of ARC neurons, marked by ex- afferents emanating from the ARC—and po- feedback signals that primarily arise from
pression of the oxytocin receptor (Oxtr), rap- tentially also other, glutamatergic inputs— changes in circulating levels of hormones,
idly decreases feeding when stimulated (Fig. 3) plays an important role because PVH neurons transmission of feed-forward signals must
(48). Consistent with the above-mentioned sa- efficiently integrate excitatory inputs owing to be fast to initiate the appropriate responses
tiety model, these Oxtr neurons inhibit feeding their expression of the voltage-gated sodium before food consumption—hence, they must
through the PVH, where they release glutamate channel NAV1.7 (53). Notably, genetic deletion be mediated by neuronal pathways.
onto satiety neurons (48). Thus, in contrast to of NAV1.7 from PVH neurons in mice decreases Electrophysiological recordings in rodents
the hunger-promoting system in which rap- their firing owing to diminished summation of and monkeys found that sensory signals that
idly acting (GABA and NPY) and slowly acting excitatory inputs and leads to the development predict future consumption or nutrient uptake
(AgRP) signals are released by one group of of massive obesity (53). rapidly alter neuronal activity in hypothalamic
neurons—AgRP neurons (27, 29, 49, 50)—the In addition to the diversification of a paral- regions (62–64). Interpretation of these find-
satiety-promoting system is functionally diver- lel glutamatergic Oxtr and melanocortin POMC ings and conceptualizing them with the var-
sified and works through two parallel-projecting circuits subserving the PVH, diverse groups of ious, functionally distinct neuronal populations
neurons: ARC-Oxtr neurons, which release the POMC neurons have been identified through of the hypothalamus was, however, difficult
fast-acting neurotransmitter glutamate, and their distinct electrophysiological responses because the molecular identity of the recorded
POMC neurons, which work through melano- to insulin and leptin and molecular profiles neurons was unknown. Optical and electro-
cortin 4 receptor (MC4R) signaling (48). Nota- (54, 55). Acute activation of POMC neurons physiological measurements from genetically
bly, glutamatergic projections from ARC-Oxtr can also increase feeding in mice, which is pre- distinct cell types have offered new insight
neurons predominantly engage MC4R-expressing sumably mediated by preferential processing of into the dynamics of defined neuronal pop-
PVH satiety neurons (51, 52), and this circuit is the POMC precursor to b-endorphin in a subset ulations in awake, freely behaving animals
regulated in strength by MC4R signaling; spe- of these neurons (46). In addition, single-cell (65–67). This led to the discovery that AgRP
cifically, activation of MC4R by the POMC- RNA-sequencing analyses revealed that leptin neurons in energy-deprived mice rapidly re-
derived neuropeptide a-melanocyte-stimulating receptor (Lepr)–expressing and Glp-1 receptor duce their activity within seconds of the ap-
hormone (a-MSH) potentiates transmission (Glp-1r)–expressing POMC neurons represent pearance of food or presentation of sensory
across the glutamatergic ARC→PVH satiety largely distinct subgroups of cells (55, 56). They cues such as those predicting food consump-
synapse (48). These two parallel-projecting exhibit a distinct profile of neuropeptide re- tion (Fig. 4) (68–70). This unexpected rapid
satiety neurons thus interact by means of ceptors that are indicative of distinct regula- reduction in neuron activity was found to
MC4R-mediated synaptic plasticity. The con- tory principles in addition to differential Lepr begin before food ingestion and to be scaled
trol of PVH satiety neurons through excitatory and Glp-1r expression (47). Interestingly, in con- with the energy content of the presented food

Brüning and Fenselau, Science 381, eabl7398 (2023) 29 September 2023 4 of 10


RES EARCH | R E V I E W

(68–70). Consistent with the opposing activity crease in food intake requires NPY signaling thalamus, and the magnitude of these responses
regulation of POMC neurons, the appearance (50), indicating that AgRP neuron–derived NPY covaries with nutrient levels in the blood at the
of food induced a rapid increase in their ac- sustains hunger during a meal. A primary role time of the cue-nutrient conditioning (78), which
tivity (69, 70). These compelling data un- for the rapid activity changes in guiding learned provides support for the notion of an integration
covered that AgRP and POMC neurons can behavior was based on observations that AgRP between external and internal nutritional cues
anticipate the future impact of consumed neuron activation is mildly aversive, which sug- also in humans.
nutrients. gests that their acute reduction in activity Because acute stimulation of AgRP neurons
Given the wide array of physiological pro- could act as a teaching signal (68). To test this rapidly alters metabolism in peripheral tissues
cesses controlled by AgRP neurons, the dis- directly, one afferent neural pathway respon- (49, 79, 80), it is likely that their food cue–
covery of their food cue–evoked inhibition has sible for rapid inhibition of AgRP neurons was evoked inhibition also contributes to cephalic-
brought forward various hypotheses to explain recently defined (76). Through this experimen- phase responses. This has, however, not been
its function (68–74). The paradoxical observa- tal design, any slow—nutrient- or hormone- directly explored and should be the subject of
tion that their inhibition occurs before food is mediated—effects could be bypassed. This future studies. Given that AgRP neurons pro-
actually consumed, and even persists during revealed a polysynaptic circuit connecting vide strong inhibitory input to POMC neurons
food consumption, implied that increased fir- the lateral hypothalamus to Lepr-expressing (27), this local connection may underlie the
ing of AgRP neurons is not required for driv- GABAergic neurons of the dorsal medial hy- rapid activation of POMC neurons that is ob-

Downloaded from https://www.science.org at Universidade Estadual de Campinas on October 16, 2023


ing feeding behavior. Optogenetic stimulation pothalamus, which synaptically inhibit AgRP served after sensory food perception (69, 70).
of AgRP neurons in the absence of food, and neurons (Fig. 4) (76, 77). Notably, imaging the There is strong evidence that the acute in-
tens of minutes before food becomes available, different nodes of this circuit demonstrated crease in POMC neuron firing rapidly induces
is sufficient to evoke the same voracious feed- that it is rapidly engaged by sensory cues pre- metabolic changes in the liver after a meal
ing response that can be observed when stim- ceding food ingestion (77). Moreover, its selec- (postprandial) (81). Specifically, calcium sig-
ulation is performed in the presence of food tive disruption greatly impaired the ability of nal analyses showed that POMC neuron activ-
(75). Recent work has shown that release of mice to learn a specific operant task for acquir- ation upon sensory food perception correlates
the neuropeptide NPY bridges the sustained ing food (77). These findings show that the with hepatic activation of the mammalian tar-
feeding response by AgRP neurons (50). Spe- acute drop in the activity of AgRP neurons is get of rapamycin (mTOR), preparing the orga-
cifically, brief optogenetic stimulation of AgRP critical for behavioral adjustments, probably nism for the ingestion of food (81). This rapid
neurons in the absence of food drives vora- by acutely eliminating the negative feeling of signaling in the liver is recapitulated by opto-
cious feeding after AgRP neuron stimulation hunger. Interestingly, in humans learned re- genetic stimulation of POMC neurons, whereas
has been terminated (50). This long-lasting in- sponses to food cues are observed in the hypo- deficiency of the downstream melanocortin
pathways attenuates the hepatic responses to
sensory food perception (81). These findings
Sensory cues that are consistent with the emerging concept of
anticipate food POMC neurons as rapid sensors of food-related
Insulin Hunger Energy Nucleus of the cues to fine tune peripheral metabolism. Ex-
sensitivity Expenditure solitary tract tending the concept of sensory food perception–
dependent POMC neuron regulation, a study
revealed that during fasting, food-odor stimu-
GABA Glp1R lation is sufficient to increase free fatty acids in
AgRP Nodose the blood through adipose tissue lipolysis in an
ganglia olfactory memory–dependent manner in mice,
LepR
Normal feeding
which is mediated by the central melanocortin
Glutamate Food and sympathetic nervous systems (82).
CCK OxtR
Activity

Collectively, the emerging data indicate that


Glut
utt 5-HT
Hypothalamus sensory perception–dependent but also home-
PYY
ostatic hormone–dependent regulation of mela-
Time
Food Gastric EEC nocortin neurons not only control feeding
perception infusion behavior and energy expenditure but also
Food cue glucose and lipid metabolism, as well as pro-
Activity

Activity

teostasis in peripheral tissues. This supports


Lateral the overarching concept that these multimodal
Dorsal medial energy state–sensing neurons are ideally po-
Time Time
sitioned to adapt the complex regulation of
integrative physiology to the energy state of
Fig. 4. Rapid and sustained regulators of AgRP neuron activity. Energy deprivation activates AgRP the organism. Given that energy sensing, me-
neurons that, once engaged, adapt numerous physiological processes, such as systemic insulin sensitivity, tabolism, and proteostasis represent key reg-
hunger drive, and thermogenesis. The increased activity of AgRP neurons is inhibited upon food consumption, a ulators of longevity and health span, these
process that comprises two distinct components. The first is sensory detection of food, or cues associated with food neurons are also potential regulators of these
delivery. This cue-evoked inhibition of AgRP neurons is astoundingly rapid, mediated by inhibitory GABAergic processes (83). AgRP neurons are required for
neurons of the dorsal medial hypothalamus (DMH) that are marked by expression of the leptin receptors (LepR). the activation of liver autophagy, another key
Recent work uncovered that glutamate release from lateral hypothalamic neurons regulates the activity of this neural pathway in protein quality control and life
pathway, which is required for learning food-acquisition tasks. The second component, which is slow and longer span, upon short-term food restriction in mice.
lasting, is triggered by nutrients that reach the gut. Factors secreted from enteroendocrine cells of the intestine— Of note, AgRP neuron–dependent control of
including cholecystokinin (CCK), peptide YY (PYY), and serotonin (5-HT), as well as gut-innervating, stretch-sensing liver autophagy declines upon aging, thus
vagal afferents—mediate the sustained inhibition of AgRP neurons. opening interesting new avenues for further

Brüning and Fenselau, Science 381, eabl7398 (2023) 29 September 2023 5 of 10


RES EARCH | R E V I E W

investigation. This points to the possibility that of EEC hormones (94), as well as pharmaco- tional consequences and explains why gastric
melanocortin neurons represent targets to im- logical administration of CCK, PYY, and 5-HT and intestinal distension by non-nutritive sub-
prove life span and health span (84). Given the all potently reduce AgRP neuron activity in stance reduces feeding owing to their ability to
emerging concept of sensory food perception– the ARC (87–89, 95). Moreover, surgical re- fill GI tract organs correlates with AgRP neuron
dependent regulation of these key metabolism moval of the vagus nerve blocks the ability of inhibition (98). Moreover, acute chemo- and
regulatory neurons, primarily through olfactory gut-delivered fat and exogenous CCK to inhibit optogenetic stimulation of the same mechano-
perception, it is notable that olfactory dysfunc- AgRP neuron activity (Fig. 4) (88). Elegant ma- sensory vagal afferents, but not chemosensory
tion represents a strong predictor of 5-year nipulation studies uncovered that vagal afferents subtypes, has been shown to potently inhibit
mortality (85). that innervate the stomach and duodenum food intake in mice (98, 102).
mediate the satiating action of CCK and nu- The molecular mechanisms by which vagal
Neuronal basis of gut-brain communication trient sensing (96). Notably, activation of this afferents detect mechanical stimuli in GI tract
The rapid activity changes in AgRP and POMC vagal gut-brain pathway was found to be organs remain incompletely understood. How-
neurons that are triggered by sensory food highly rewarding as assessed by using self- ever, many of the receptors and ion channels
perception remain at a relatively stable level stimulation and place preference assays, and that are expressed by somatosensory neurons
if mice subsequently obtain and consume food to recruit substantia nigra dopaminergic re- that detect mechanical stimuli in other periph-
ad libitum (Fig. 4) (68–70, 86). If food is, how- ward neurons (96). Thus, vagal afferents con- eral organs (104, 105), such as the skin, are also

Downloaded from https://www.science.org at Universidade Estadual de Campinas on October 16, 2023


ever, not accessible, or provides no or only few vey gut-derived signals that decrease the found in vagal afferents (97, 98). Thus, these
calories, the acute activity changes are readily activity of aversive AgRP neurons and that molecular sensors likely account, at least in
reversed (68–70, 86). Thus, after transient, enhance reward-related signaling through part, for the stretch- and distension-induced
sensory-driven signals, postingestive signals the dopaminergic pathway. activation of vagal afferents whose periph-
eventually reach the ARC and sustain neuro- Vagal afferents are functionally and anato- eral terminals innervate the muscular layers
nal activity at a new level. Recent studies have mically heterogeneous, and their peripheral of the stomach and intestines (99). In addi-
demonstrated that food-derived signals that axons also innervate other internal organs in tion, activation of EECs may contribute to gut-
arise in the gut play an important role in addition to the stomach and the intestine (90). brain communication by mechanosensory
mediating these responses (87–89). Specifically, RNA-sequencing of NG, including single-cell vagal afferents to inhibit food intake. EECs
calcium recordings from AgRP neurons showed RNA-sequencing of retrogradely labeled organ- can convert mechanical stimuli into the pro-
that their inhibition after food consumption specific innervating neurons, recently char- duction of gut hormones, which in turn acti-
is proportional to the caloric content of the acterized the population(s) that transmit vate vagal afferents (106–108). This could
food (87, 89). This correlative inhibition is re- gut-derived information to the brain in mice include CCK release and the activation of
capitulated when nutrients are directly delivered (97–101). These studies revealed that vagal stomach-innervating, stretch-responsive Glp-1r
into the stomach or small intestine, bypassing afferents express various genetic markers, in- subtypes. Indeed, selective stimulation of CCK-
any sensory and oral cues (Fig. 4) (87, 89). Thus, cluding calcitonin gene-related peptide 1 (Calca), expressing EECs suppressed feeding in mice
AgRP neurons integrate online information Glp-1r, the proton-sensing G protein–coupled (109, 110). This was abolished by removal of
from the gut that precedes assimilation of in- receptor Gpr65, vasoactive intestinal peptide vagus nerve innervation of the GI tract or
gested calories and eventually results in changes (Vip), and Oxtr. The subtypes marked by blocking CCK receptors, which are highly ex-
of circulating energy state–communicating these five genes are particularly noteworthy pressed in Glp-1r vagal afferents (98, 99, 102).
hormones. Molecularly distinct vagal sensory because as demonstrated through selective It is likewise unclear what central pathways
neurons, which bridge the gut and the brain, histological assessment, they show distinct downstream of vagal afferents relay gut-
are a major afferent pathway of this commu- innervation patterns and morphologies in derived information to the ARC and how this
nication and mediate various physiological re- GI tract organs (98, 99, 101, 102). Calca and route actually contributes to the regulation of
sponses to maintain systemic metabolism. Glp-1r define subtypes that primarily innervate feeding behavior as well as other aspects of
Sensory neurons of the vagus nerve, com- the stomach (98, 99, 102, 103), where Calca metabolism. Systemic glucose regulation is
monly called vagal afferents, are pseudouni- terminals form putative chemosensory muco- of particular interest in this regard because
polar neurons, whose cell bodies are located sal endings (98, 103). Glp-1r terminals are neural gut-brain communication and ARC-
in two nodose ganglia (NG) at the base of the enriched in gastric muscular layers and form based circuits both have long been recognized
skull in proximity to the carotid arteries (90, 91). intraganglionic laminar endings (IGLE) as essential for glycemic control, especially in
Their central axons project to the dorsal (98, 99, 102), which are thought to be me- the postprandial state (13, 111). Vagal afferent
hindbrain, where they synaptically activate chanosensors. By contrast, Gpr65, Vip, and signaling onto distinct neurons in the hind-
neurons in the nucleus of the solitary tract Oxtr subtypes primarily innervate the small brain NTS/AP is believed to be an important
and in the area postrema (NTS/AP) (Fig. 4) intestine, where Gpr65 and Vip mucosal control point for the integration and routing
(90, 91). Their peripheral axons target the organs endings are enriched in intestinal villi, and of gut-derived information (Fig. 4). Molecu-
of the gastrointestinal (GI) tract, where they Oxtr form IGLE, with the highest density larly defined NTS/AP neurons are activated
sense mechanical signals, such as stretch or in the muscular layers of the proximal small upon selective opto- or chemogenetic stimu-
distension (92). In addition, they sense chem- intestine (98, 99, 102). Notably, as revealed by lation of gut-innervating vagal afferents in
ical signals, primarily through hormones simultaneously monitoring AgRP neuron mice (96, 98, 102) and by natural stimuli that
secreted by enteroendocrine cells (EECs), activity while chemogenetically manipulating excite them, such as organ stretch, CCK ad-
including cholecystokinin (CCK), GLP-1, pep- distinct vagal afferents, stimulation of mecha- ministration, meal ingestion, or gastric delivery
tide YY (PYY), and serotonin [5-hydroxy- nosensory (Oxtr and Glp-1r) but not chemo- of nutrients (112, 113). Further, manipulating
tryptamine (5-HT)] (91, 93, 94). Vagal afferent sensory (Gpr65 and Vip) subtypes were found distinct NTS/AP neurons produces effects on
activation by these gut hormones is likely to to selectively inhibit AgRP neuron activity food consumption (114–121), appetitive and
dominate the sustained inhibition of AgRP (Fig. 4) (98). These observations suggest that aversive behavior (115, 116), and systemic
neurons after food consumption. Infusion AgRP neuron–based hunger circuits integrate glucose homeostasis (120, 122). From the NTS/
of nutrients directly into the small intestine online mechanical information from the gut. AP, gut-derived signals could reach the ARC
(87–89, 95), which causes the natural release This pathway has likely far-reaching func- through direct projections or relay through

Brüning and Fenselau, Science 381, eabl7398 (2023) 29 September 2023 6 of 10


RES EARCH | R E V I E W

other nodes. The lateral parabrachial nucleus creased hypothalamic inflammation, in part ingly, hopes were high for leptin treatment of
(LPBN) is a strong candidate node for relaying from enhanced astrocyte and microglia acti- obesity. However, it was soon realized that
gut mechano- and chemosensory signals be- vation, has been shown to cause neuronal although leptin provided a cure for humans
cause it is heavily innervated by the NTS/AP leptin and insulin resistance, even preceding with rare leptin gene mutations, the majority
(123). Moreover, recent calcium imaging stud- the manifestation of systemic metaflamma- of obese humans presented with elevated leptin
ies have established that anorexigenic neurons tion in obesity (132). The complexity of the par- levels, which is indicative of leptin resistance.
in the LPBN are activated by meal ingestion and ticipating CNS cell types in these processes— As such, treating obese humans with leptin
selective stimulation of the stomach (124, 125). microglia, circulating macrophages, and only yielded disappointing outcomes (140).
However, neurons in the NTS/AP, and also in astrocytes—have been reviewed elsewhere (133). Nevertheless, recent studies have identified
the LPBN, are remarkably heterogeneous, and Exaggerated endoplasmic reticulum (ER) subtypes of obese humans who exhibit rela-
further research is needed to fully map their stress contributes to altered hormone respon- tively low circulating leptin levels in proportion
afferent and efferent connections (114, 118, 126). siveness in critical metabolism regulatory neuro- to their body-mass index, which is indicative
Another major obstacle in deciphering the circuits such as POMC and AgRP neurons (134). of relative leptin deficiency, and these individ-
functionally relevant circuits between vagal In addition, ectopic lipid deposition and asso- uals exhibit the most profound weight reduc-
afferents and the ARC has been the technical ciated lipotoxicity has also been linked to leptin tion upon administering recombinant leptin.
difficulties associated with targeting vagal and insulin resistance in the ARC, and identify- Thus, identifying the proper patient class could

Downloaded from https://www.science.org at Universidade Estadual de Campinas on October 16, 2023


afferents in NGs, which are extremely small and ing the responsible lipid species accumulating make leptin a therapeutic option for a substan-
located close to delicate structures. Recently, in the ARC to induce hormone resistance is still tial number of obese patients. Given that leptin
the metabolic functions of specific vagal affer- subject to intense research (135). The dynamic resistance may occur in the majority of patients,
ents have been investigated by using an inter- regulation of mitochondrial function and dyna- strategies to resensitize leptin action in obesity
sectional approach to facilitate genetic entry mics in metabolism regulatory neurons has may provide a promising avenue for new ther-
into molecularly defined subtypes (102). This been identified as a key determinant of ARC apeutics. Targeting pathways such as exagger-
revealed that stomach-innervating mechano- neuronal populations, and deregulated balance ated ER stress and lipotoxicity in hypothalamic
sensory Glp-1r vagal afferents relay anorexi- of mitochondrial fission and fusion in POMC neurons may thus offer new strategies. Appli-
genic signals (99, 102) and play a crucial role and AgRP neurons has been linked to altered cation of chemical chaperones or improving
in controlling glycemia during feeding (102). fuel metabolism of these cells and their fine- leptin action with celastrol, a chemical compound
By contrast, selective manipulations of Gpr65 tuned, energy state–dependent regulation (136). isolated from the root extracts of Tripterygium
vagal afferents, which are activated by intes- To add more complexity, these mechanisms wilfordii, have been shown to enhance leptin
tinal nutrients (99), revealed that their activa- are closely interconnected because ER stress sensitivity and reduce food intake and body
tion increases hepatic glucose production (102), and mitochondrial stress responses are inti- weight in HFD-fed obese mice, which currently
but that they are dispensable for food-intake mately linked, and lipotoxic species accumu- awaits further clinical development (141).
regulation (98, 102). Future work is required lating in obesity have been found to disrupt Alternative pathways targeted to improve
to map the functional neurocircuits between both mitochondrial dynamics and function weight loss include treatment with long-acting
these and other gut-innervating vagal afferents, as well as to cause ER stress, also in the CNS. GLP-1 analogs. These drugs represent one of
NTS/AP neurons, and ARC neurons and to Although most of the present studies have the few effective approaches currently in cli-
detail the connections between gut mechano- identified cell-autonomous neuronal changes nical practice. Liraglutide and more recently
sensation and chemosensation and their im- in obesity in mice, the effect of HFD-feeding semaglutide are approved to treat obesity in
pact on systemic metabolic homeostasis. and obesity on neuronal wiring and commu- some countries; semaglutide treatment is
nication remains ill defined. Studies on the capable of reducing body weight by up to 20%
Disease-associated circuit alterations effects of altered maternal metabolism during (142). Although the targets of GLP-1 to reduce
Soon after the discovery of leptin, it was rec- pregnancy on the long-term predisposition of food intake have been extensively studied over
ognized that obese humans and obese mouse the offspring to the development of obesity recent years—and clearly include POMC neu-
models exhibit elevated leptin levels and dis- and metabolic disorders have revealed that rons in the ARC, as well as through an only
play an attenuated response to exogenously deregulated insulin signaling and ER stress recently identified Lepr/Glp1r–expressing neu-
applied leptin (127, 128). This led to the foun- activation inhibit projection formation of ron population in the dorsal medial hypo-
dation for a concept of leptin resistance in POMC neurons in offspring during critical thalamus (DMH) (143)—GLP-1R is expressed
obesity, particularly in the ARC (129). Similarly, developmental periods (137, 138). This effect in multiple cell types in the central and periph-
mouse obesity models exhibit resistance to the then causes altered POMC neuron communi- eral nervous system, and the weight-reducing
regulatory function of the hormones insulin cation to downstream effector neurons in the effect of GLP-1 analogs has been shown to
and ghrelin in the brain (130). These findings offspring, leading to increased propensity of depend on both systems (121). The above-
have nurtured research into the molecular obesity and type 2 diabetes mellitus develop- mentioned NTS/AP region of the hindbrain,
mechanisms of central hormone resistance in ment. Interestingly, mutations in axon guidance which relays gut-derived information, likely
obesity in analogy to the mechanisms under- molecules, which are critical for POMC neuron plays an important role in mediating the
lying insulin resistance in peripheral tissues of projection development in mice, have also been weight loss–promoting effects of GLP-1 ana-
humans and mouse models of insulin resist- identified in humans with monogenic obesity logs. Studies in mice, rats, and nonhuman pri-
ance and type 2 diabetes mellitus. Numerous (139). Thus, the field of metabolic perinatal mates have revealed that GLP-1–responsive
mechanisms leading to obesity associated leptin programming of critical feeding circuits clearly cells are distributed throughout the NTS/AP
and insulin resistance mainly in the ARC, deserves further study. (118, 144–146) and that the activity of these
particularly in POMC, and AgRP neurons have cells contributes to the observed feeding sup-
been identified. Such mechanisms include Therapeutic strategies to control metabolism pression and body weight loss upon adminis-
increased expression of the suppressor of cyto- The discovery of leptin has provided a paradigm- tration of GLP-1 analogs (118). Although
kine signaling 3 (SOCS-3), which is a well- shifting molecular correlate that has paved further investigations are needed to fully define
characterized inhibitor of LEPR and insulin the way for the functional interrogation of the functional cellular and circuit mechanisms,
receptor (INSR) signaling (131). Moreover, in- metabolism regulatory neurocircuits. Accord- these data provide support that this hindbrain

Brüning and Fenselau, Science 381, eabl7398 (2023) 29 September 2023 7 of 10


RES EARCH | R E V I E W

region is a promising target for the metabolic more innovative therapeutic approaches. Use signaling. Front. Neurosci. 13, 240 (2019). doi: 10.3389/
benefits of peripherally administered com- of optogenetics in humans is entering clinical fnins.2019.00240; pmid: 30941008
19. C. García-Cáceres et al., Astrocytic insulin signaling
pounds that enable the treatment of obesity. trials for vision restoration (152). Therefore, couples brain glucose uptake with nutrient availability. Cell
Another very promising approach for weight development of improved opto- or chemogenet- 166, 867–880 (2016). doi: 10.1016/j.cell.2016.07.028;
reduction and improvement of metabolism ic tools alongside the more defined identifi- pmid: 27518562
20. J. G. Kim et al., Leptin signaling in astrocytes regulates
in obesity builds on the recent development of cation of targetable critical neuronal nodes in
hypothalamic neuronal circuits and feeding. Nat. Neurosci. 17,
polypeptide agonists. This new class of thera- metabolic control may lead to the develop- 908–910 (2014). doi: 10.1038/nn.3725; pmid: 24880214
peutics unifies receptor specificity of two or ment of gene therapeutic approaches for the 21. L. Varela et al., Hunger-promoting AgRP neurons trigger an
more neuropeptides in a single molecule and remote control of feeding behavior and sys- astrocyte-mediated feed-forward autoactivation loop in mice.
J. Clin. Invest. 131, e144239 (2021). doi: 10.1172/JCI144239;
among others includes GLP-1-glucagon, GLP- temic metabolism (153). Nonetheless, all of pmid: 33848272
1–gastric inhibitory polypeptide (GIP), or other these developments to treat or even prevent 22. S. Kohnke et al., Nutritional regulation of oligodendrocyte
polyagonists (147). These substances exhibit obesity will critically depend on furthering differentiation regulates perineuronal net remodeling in the
median eminence. Cell Rep. 36, 109362 (2021). doi: 10.1016/
tremendous efficacy in preclinical models, and our understanding of the detailed regulatory j.celrep.2021.109362; pmid: 34260928
recently, the first GLP-1–GIP agonist provided principles of neurocircuits in metabolism. 23. M. Valdearcos et al., Microglial inflammatory signaling
superior efficiency as compared with the weight- orchestrates the hypothalamic immune response to dietary
RE FERENCES AND NOTES excess and mediates obesity susceptibility. Cell Metab. 26,
reducing effect of semaglutide and has obtained 185–197.e3 (2017). doi: 10.1016/j.cmet.2017.05.015;

Downloaded from https://www.science.org at Universidade Estadual de Campinas on October 16, 2023


1. W. B. Cannon, [Homeostasis adaptations]. An. Med. Ateneo
approval for treatment of type 2 diabetes (148). Ramon Cajal Mex. 3, 1–9 (1945) [Homeostasis adaptations]. pmid: 28683286
Multiple other combined modalities of neuro- pmid: 20982179 24. C. García-Cáceres et al., Role of astrocytes, microglia, and
2. P. Sterling, Allostasis: A model of predictive regulation. tanycytes in brain control of systemic metabolism. Nat.
peptide combinations are currently undergoing Neurosci. 22, 7–14 (2019). doi: 10.1038/s41593-018-0286-y;
Physiol. Behav. 106, 5–15 (2012). doi: 10.1016/
advanced clinical testing (149). However, the j.physbeh.2011.06.004; pmid: 21684297 pmid: 30531847
molecular targets of these complex drugs still 3. G. C. Kennedy, The role of depot fat in the hypothalamic 25. J. N. Campbell et al., A molecular census of arcuate
control of food intake in the rat. Proc. R. Soc. Lond. Ser. B hypothalamus and median eminence cell types.
await detailed elucidation, and approaches such
140, 578–596 (1953). doi: 10.1098/rspb.1953.0009; Nat. Neurosci. 20, 484–496 (2017). doi: 10.1038/nn.4495;
as single-cell sequencing and cell type–specific pmid: 13027283 pmid: 28166221
modulation of neuronal activity will aid in the 4. Y. Zhang et al., Positional cloning of the mouse obese gene 26. R. A. Romanov et al., Molecular interrogation of hypothalamic
elucidation of their exact therapeutic mecha- and its human homologue. Nature 372, 425–432 (1994). organization reveals distinct dopamine neuronal subtypes.
doi: 10.1038/372425a0; pmid: 7984236 Nat. Neurosci. 20, 176–188 (2017). doi: 10.1038/nn.4462;
nism(s) (Box 1 and Fig. 2). 5. M. Tschöp, D. L. Smiley, M. L. Heiman, Ghrelin induces pmid: 27991900
Another important therapeutic challenge is adiposity in rodents. Nature 407, 908–913 (2000). 27. D. Atasoy, J. N. Betley, H. H. Su, S. M. Sternson,
posed by weight regain after successful weight doi: 10.1038/35038090; pmid: 11057670 Deconstruction of a neural circuit for hunger. Nature 488,
6. J. C. Brüning et al., Role of brain insulin receptor in control of 172–177 (2012). doi: 10.1038/nature11270; pmid: 22801496
reduction, which also holds true after dis- 28. Q. Tong, C. P. Ye, J. E. Jones, J. K. Elmquist, B. B. Lowell,
body weight and reproduction. Science 289, 2122–2125
continuation of long-acting GLP-1 analogs (150). (2000). doi: 10.1126/science.289.5487.2122; pmid: 11000114 Synaptic release of GABA by AgRP neurons is required for
Further defining the underlying mechanisms, 7. M. Blüher, Obesity: Global epidemiology and pathogenesis. normal regulation of energy balance. Nat. Neurosci. 11,
Nat. Rev. Endocrinol. 15, 288–298 (2019). doi: 10.1038/ 998–1000 (2008). doi: 10.1038/nn.2167; pmid: 19160495
which include processes of long-term adapta- 29. M. J. Krashes, B. P. Shah, S. Koda, B. B. Lowell, Rapid versus
s41574-019-0176-8; pmid: 30814686
tions in the neural pathways that coordinate 8. E. K. Speliotes et al, Association analyses of 249,796 delayed stimulation of feeding by the endogenously released
the activity of ARC-based feeding circuits, may individuals reveal 18 new loci associated with body mass AgRP neuron mediators GABA, NPY, and AgRP. Cell Metab.
index. Nat. Genet. 42, 937–948 (2010). doi: 10.1038/ng.686; 18, 588–595 (2013). doi: 10.1016/j.cmet.2013.09.009;
lead to the development of molecules that pmid: 24093681
pmid: 20935630
counteract this well-described yo-yo effect. 9. R. J. F. Loos, G. S. H. Yeo, The genetics of obesity: From 30. X. Zhang, A. N. van den Pol, Hypothalamic arcuate nucleus
Weight loss upon caloric deprivation in mice discovery to biology. Nat. Rev. Genet. 23, 120–133 (2022). tyrosine hydroxylase neurons play orexigenic role in
leads to a remarkably long-lasting enhance- doi: 10.1038/s41576-021-00414-z; pmid: 34556834 energy homeostasis. Nat. Neurosci. 19, 1341–1347 (2016).
10. J. M. Friedman, A war on obesity, not the obese. Science doi: 10.1038/nn.4372; pmid: 27548245
ment of the excitatory synaptic input to AgRP 299, 856–858 (2003). doi: 10.1126/science.1079856; 31. S. Spencer et al., Distribution of catecholamine-containing
neurons of the ARC, causing their elevated pmid: 12574619 neurons in the normal human hypothalamus. Brain Res. 328,
neural activity and concomitant increase in 11. J. T. Clark, P. S. Kalra, S. P. Kalra, Neuropeptide Y stimulates 73–80 (1985). doi: 10.1016/0006-8993(85)91324-1;
feeding but inhibits sexual behavior in rats. Endocrinology pmid: 2857591
hunger drive until lost weight is regained (151). 117, 2435–2442 (1985). doi: 10.1210/endo-117-6-2435; 32. B. J. Everitt, T. Hökfelt, J. Y. Wu, M. Goldstein, Coexistence of
Identifying the molecular mechanisms respon- pmid: 3840737 tyrosine hydroxylase-like and gamma-aminobutyric acid-like
sible for the observed long-term effects will be a 12. T. L. Horvath, F. Naftolin, S. P. Kalra, C. Leranth, immunoreactivities in neurons of the arcuate nucleus.
Neuropeptide-Y innervation of beta-endorphin-containing Neuroendocrinology 39, 189–191 (1984). doi: 10.1159/
major task for future investigations to advance
cells in the rat mediobasal hypothalamus: A light and 000123977; pmid: 6147773
targeting strategies that enable inhibiting the electron microscopic double immunostaining analysis. 33. P. J. Keller, W. Lichtensteiger, Stimulation of tubero-infundibular
induction, or reversing the maintenance, of this Endocrinology 131, 2461–2467 (1992). doi: 10.1210/ dopamine neurones and gonadotrophin secretion. J. Physiol.
neuronally induced synaptic plasticity mecha- endo.131.5.1425443; pmid: 1425443 219, 385–401 (1971). doi: 10.1113/jphysiol.1971.sp009668;
13. A. Jais, J. C. Brüning, Arcuate nucleus-dependent regulation pmid: 4945749
nism. Because reorganization of the synaptic of metabolism-pathways to obesity and diabetes mellitus. 34. Z. A. Knight et al., Molecular profiling of activated neurons by
inputs to AgRP and POMC neurons are ob- Endocr. Rev. 43, 314–328 (2022). doi: 10.1210/endrev/ phosphorylated ribosome capture. Cell 151, 1126–1137
served in mouse models of obesity (36), tar- bnab025; pmid: 34490882 (2012). doi: 10.1016/j.cell.2012.10.039; pmid: 23178128
14. R. Dali, J. Estrada-Meza, F. Langlet, Tanycyte, the neuron 35. A. Jais et al., PNOCARC neurons promote hyperphagia and
geting the underlying signaling pathways may whisperer. Physiol. Behav. 263, 114108 (2023). doi: 10.1016/ obesity upon high-fat-diet feeding. Neuron 106, 1009–1025.
also offer new ways to offset obesity-induced j.physbeh.2023.114108; pmid: 36740135 e10 (2020). doi: 10.1016/j.neuron.2020.03.022;
alterations in their energy state–dependent 15. M. Porniece Kumar et al., Insulin signalling in tanycytes gates pmid: 32302532
hypothalamic insulin uptake and regulation of AgRP neuron 36. S. Pinto et al., Rapid rewiring of arcuate nucleus feeding
activity regulation as well as how these neu-
activity. Nat. Metab. 3, 1662–1679 (2021). doi: 10.1038/ circuits by leptin. Science 304, 110–115 (2004). doi: 10.1126/
ronal populations integrate gut-derived and s42255-021-00499-0; pmid: 34931084 science.1089459; pmid: 15064421
anticipatory signals when highly palatable 16. W. Bakker et al., Acute changes in systemic glycemia gate 37. M. A. Cowley et al., Leptin activates anorexigenic POMC
energy-dense food is available in obese indi- access and action of GLP-1R agonist on brain structures neurons through a neural network in the arcuate nucleus.
controlling energy homeostasis. Cell Rep. 41, 111698 (2022). Nature 411, 480–484 (2001). doi: 10.1038/35078085;
viduals (95). doi: 10.1016/j.celrep.2022.111698; pmid: 36417883 pmid: 11373681
17. M. Duquenne et al., Leptin brain entry via a tanycytic LepR- 38. A. R. Rau, S. T. Hentges, GABAergic inputs to POMC neurons
Conclusions EGFR shuttle controls lipid metabolism and pancreas originating from the dorsomedial hypothalamus are regulated
function. Nat. Metab. 3, 1071–1090 (2021). doi: 10.1038/ by energy state. J. Neurosci. 39, 6449–6459 (2019).
Further refinement of the molecular-systems s42255-021-00432-5; pmid: 34341568 doi: 10.1523/JNEUROSCI.3193-18.2019; pmid: 31235650
neuroscience approaches described for pre- 18. S. Yoo, D. Cha, D. W. Kim, T. V. Hoang, S. Blackshaw, 39. L. Vong et al., Leptin action on GABAergic neurons prevents
clinical research may pave the way for even Tanycyte-independent control of hypothalamic leptin obesity and reduces inhibitory tone to POMC neurons.

Brüning and Fenselau, Science 381, eabl7398 (2023) 29 September 2023 8 of 10


RES EARCH | R E V I E W

Neuron 71, 142–154 (2011). doi: 10.1016/j.neuron.2011.05.028; 61. M. L. Power, J. Schulkin, Anticipatory physiological regulation 84. W. Chen et al., Nutrient-sensing AgRP neurons relay control
pmid: 21745644 in feeding biology: Cephalic phase responses. Appetite 50, of liver autophagy during energy deprivation. Cell Metab. 35,
40. Y. Qi et al., Agrp-negative arcuate NPY neurons drive feeding 194–206 (2008). doi: 10.1016/j.appet.2007.10.006; 786–806.e13 (2023). doi: 10.1016/j.cmet.2023.03.019;
under positive energy balance via altering leptin pmid: 18045735 pmid: 37075752
responsiveness in POMC neurons. Cell Metab. 35, 979–995. 62. B. K. Anand, R. V. Pillai, Activity of single neurones in the 85. J. M. Pinto, K. E. Wroblewski, D. W. Kern, L. P. Schumm,
e7 (2023). doi: 10.1016/j.cmet.2023.04.020; pmid: 37201523 hypothalamic feeding centres: Effect of gastric distension. M. K. McClintock, Olfactory dysfunction predicts 5-year
41. C. Zhan et al., Acute and long-term suppression of feeding J. Physiol. 192, 63–77 (1967). doi: 10.1113/jphysiol.1967. mortality in older adults. PLOS ONE 9, e107541 (2014).
behavior by POMC neurons in the brainstem and sp008288; pmid: 4860993 doi: 10.1371/journal.pone.0107541; pmid: 25271633
hypothalamus, respectively. J. Neurosci. 33, 3624–3632 63. E. Arnauld, J. du Pont, Vasopressin release and firing of 86. Y. Livneh et al., Homeostatic circuits selectively gate food cue
(2013). doi: 10.1523/JNEUROSCI.2742-12.2013; supraoptic neurosecretory neurones during drinking in the responses in insular cortex. Nature 546, 611–616 (2017).
pmid: 23426689 dehydrated monkey. Pflugers Arch. 394, 195–201 (1982). doi: 10.1038/nature22375; pmid: 28614299
42. L. Yaswen, N. Diehl, M. B. Brennan, U. Hochgeschwender, doi: 10.1007/BF00589091; pmid: 7145599 87. L. R. Beutler et al., Dynamics of gut-brain communication
Obesity in the mouse model of pro-opiomelanocortin 64. S. Maddison, R. I. Horrell, Hypothalamic unit responses to underlying hunger. Neuron 96, 461–475.e5 (2017).
deficiency responds to peripheral melanocortin. Nat. Med. 5, alimentary perfusions in the anesthetised rat. Brain Res. Bull. doi: 10.1016/j.neuron.2017.09.043; pmid: 29024666
1066–1070 (1999). doi: 10.1038/12506; pmid: 10470087 4, 259–266 (1979). doi: 10.1016/0361-9230(79)90290-9; 88. N. Goldstein et al., Hypothalamic detection of macronutrients
43. A. W. Xu et al., Effects of hypothalamic neurodegeneration on pmid: 466512 via multiple gut-brain pathways. Cell Metab. 33, 676–687.e5
energy balance. PLOS Biol. 3, e415 (2005). doi: 10.1371/ 65. L. A. Gunaydin et al., Natural neural projection dynamics (2021). doi: 10.1016/j.cmet.2020.12.018; pmid: 33450178
journal.pbio.0030415; pmid: 16296893 underlying social behavior. Cell 157, 1535–1551 (2014). 89. Z. Su, A. L. Alhadeff, J. N. Betley, Nutritive, Post-ingestive
44. H. Krude et al., Severe early-onset obesity, adrenal doi: 10.1016/j.cell.2014.05.017; pmid: 24949967 signals are the primary regulators of AgRP neuron
insufficiency and red hair pigmentation caused by POMC 66. G. Cui et al., Concurrent activation of striatal direct and activity. Cell Rep. 21, 2724–2736 (2017). doi: 10.1016/
mutations in humans. Nat. Genet. 19, 155–157 (1998). indirect pathways during action initiation. Nature 494, j.celrep.2017.11.036; pmid: 29212021

Downloaded from https://www.science.org at Universidade Estadual de Campinas on October 16, 2023


doi: 10.1038/509; pmid: 9620771 238–242 (2013). doi: 10.1038/nature11846; pmid: 23354054 90. H. R. Berthoud, W. L. Neuhuber, Functional and chemical
45. Y. Aponte, D. Atasoy, S. M. Sternson, AGRP neurons are 67. J. Y. Cohen, S. Haesler, L. Vong, B. B. Lowell, N. Uchida, anatomy of the afferent vagal system. Auton. Neurosci. 85,
sufficient to orchestrate feeding behavior rapidly and without Neuron-type-specific signals for reward and punishment in 1–17 (2000). doi: 10.1016/S1566-0702(00)00215-0;
training. Nat. Neurosci. 14, 351–355 (2011). doi: 10.1038/ the ventral tegmental area. Nature 482, 85–88 (2012). pmid: 11189015
nn.2739; pmid: 21209617 doi: 10.1038/nature10754; pmid: 22258508 91. S. J. Brookes, N. J. Spencer, M. Costa, V. P. Zagorodnyuk,
46. M. Koch et al., Hypothalamic POMC neurons promote 68. J. N. Betley et al., Neurons for hunger and thirst transmit a Extrinsic primary afferent signalling in the gut. Nat. Rev.
cannabinoid-induced feeding. Nature 519, 45–50 (2015). negative-valence teaching signal. Nature 521, 180–185 Gastroenterol. Hepatol. 10, 286–296 (2013). doi: 10.1038/
doi: 10.1038/nature14260; pmid: 25707796 (2015). doi: 10.1038/nature14416; pmid: 25915020 nrgastro.2013.29; pmid: 23438947
47. N. Biglari et al., Functionally distinct POMC-expressing 69. Y. Chen, Y. C. Lin, T. W. Kuo, Z. A. Knight, Sensory 92. H. R. Berthoud, L. A. Blackshaw, S. J. Brookes, D. Grundy,
neuron subpopulations in hypothalamus revealed by detection of food rapidly modulates arcuate feeding circuits. Neuroanatomy of extrinsic afferents supplying the
intersectional targeting. Nat. Neurosci. 24, 913–929 (2021). Cell 160, 829–841 (2015). doi: 10.1016/j.cell.2015.01.033; gastrointestinal tract. Neurogastroenterol. Motil. 16 (Suppl 1),
doi: 10.1038/s41593-021-00854-0; pmid: 34002087 pmid: 25703096 28–33 (2004). doi: 10.1111/j.1743-3150.2004.00471.x;
48. H. Fenselau et al., A rapidly acting glutamatergic ARC→PVH 70. Y. Mandelblat-Cerf et al., Arcuate hypothalamic AgRP and pmid: 15066001
satiety circuit postsynaptically regulated by a-MSH. putative POMC neurons show opposite changes in spiking 93. G. J. Dockray, Enteroendocrine cell signalling via the vagus
Nat. Neurosci. 20, 42–51 (2017). doi: 10.1038/nn.4442; across multiple timescales. eLife 4, e07122 (2015). nerve. Curr. Opin. Pharmacol. 13, 954–958 (2013).
pmid: 27869800 doi: 10.7554/eLife.07122; pmid: 26159614 doi: 10.1016/j.coph.2013.09.007; pmid: 24064396
49. L. Engström Ruud, M. M. A. Pereira, A. J. de Solis, 71. Y. Chen, Z. A. Knight, Making sense of the sensory 94. F. M. Gribble, F. Reimann, Enteroendocrine Cells: Chemosensors
H. Fenselau, J. C. Brüning, NPY mediates the rapid feeding regulation of hunger neurons. BioEssays 38, 316–324 (2016). in the Intestinal Epithelium. Annu. Rev. Physiol. 78, 277–299
and glucose metabolism regulatory functions of AgRP doi: 10.1002/bies.201500167; pmid: 26898524 (2016). doi: 10.1146/annurev-physiol-021115-105439;
neurons. Nat. Commun. 11, 442 (2020). doi: 10.1038/ 72. M. L. Andermann, B. B. Lowell, Toward a wiring diagram pmid: 26442437
s41467-020-14291-3; pmid: 31974377 understanding of appetite control. Neuron 95, 757–778 95. C. M. Mazzone et al., High-fat food biases hypothalamic
50. Y. Chen et al., Sustained NPY signaling enables AgRP (2017). doi: 10.1016/j.neuron.2017.06.014; pmid: 28817798 and mesolimbic expression of consummatory drives.
neurons to drive feeding. eLife 8, e46348 (2019). 73. R. J. Seeley, K. C. Berridge, The hunger games. Cell 160, Nat. Neurosci. 23, 1253–1266 (2020). doi: 10.1038/
doi: 10.7554/eLife.46348; pmid: 31033437 805–806 (2015). doi: 10.1016/j.cell.2015.02.028; s41593-020-0684-9; pmid: 32747789
51. N. Balthasar et al., Divergence of melanocortin pathways in pmid: 25723156 96. W. Han et al., A neural circuit for gut-induced reward. Cell
the control of food intake and energy expenditure. Cell 123, 74. S. M. Sternson, A. K. Eiselt, Three pillars for the neural 175, 665–678.e23 (2018). doi: 10.1016/j.cell.2018.08.049;
493–505 (2005). doi: 10.1016/j.cell.2005.08.035; control of appetite. Annu. Rev. Physiol. 79, 401–423 pmid: 30245012
pmid: 16269339 (2017). doi: 10.1146/annurev-physiol-021115-104948; 97. J. Kupari, M. Häring, E. Agirre, G. Castelo-Branco, P. Ernfors,
52. A. S. Garfield et al., A neural basis for melanocortin-4 pmid: 27912679 An atlas of vagal sensory neurons and their molecular
receptor-regulated appetite. Nat. Neurosci. 18, 863–871 75. Y. Chen, Y. C. Lin, C. A. Zimmerman, R. A. Essner, specialization. Cell Rep. 27, 2508–2523.e4 (2019).
(2015). doi: 10.1038/nn.4011; pmid: 25915476 Z. A. Knight, Hunger neurons drive feeding through a doi: 10.1016/j.celrep.2019.04.096; pmid: 31116992
53. T. Branco et al., Near-perfect synaptic integration by nav1.7 sustained, positive reinforcement signal. eLife 5, e18640 98. L. Bai et al., Genetic identification of vagal sensory neurons
in hypothalamic neurons regulates body weight. Cell 165, (2016). doi: 10.7554/eLife.18640; pmid: 27554486 that control feeding. Cell 179, 1129–1143.e23 (2019).
1749–1761 (2016). doi: 10.1016/j.cell.2016.05.019; 76. A. S. Garfield et al., Dynamic GABAergic afferent modulation doi: 10.1016/j.cell.2019.10.031; pmid: 31730854
pmid: 27315482 of AgRP neurons. Nat. Neurosci. 19, 1628–1635 (2016). 99. E. K. Williams et al., Sensory neurons that detect stretch and
54. K. W. Williams et al., Segregation of acute leptin and insulin doi: 10.1038/nn.4392; pmid: 27643429 nutrients in the digestive system. Cell 166, 209–221 (2016).
effects in distinct populations of arcuate proopiomelanocortin 77. J. Berrios et al., Food cue regulation of AGRP hunger neurons doi: 10.1016/j.cell.2016.05.011; pmid: 27238020
neurons. J. Neurosci. 30, 2472–2479 (2010). doi: 10.1523/ guides learning. Nature 595, 695–700 (2021). doi: 10.1038/ 100. R. B. Chang, D. E. Strochlic, E. K. Williams, B. D. Umans,
JNEUROSCI.3118-09.2010; pmid: 20164331 s41586-021-03729-3; pmid: 34262177 S. D. Liberles, Vagal sensory neuron subtypes that
55. B. Y. H. Lam et al., Heterogeneity of hypothalamic pro- 78. I. E. de Araujo, T. Lin, M. G. Veldhuizen, D. M. Small, differentially control breathing. Cell 161, 622–633 (2015).
opiomelanocortin-expressing neurons revealed by single-cell Metabolic regulation of brain response to food cues. Curr. doi: 10.1016/j.cell.2015.03.022; pmid: 25892222
RNA sequencing. Mol. Metab. 6, 383–392 (2017). Biol. 23, 878–883 (2013). doi: 10.1016/j.cub.2013.04.001; 101. Q. Zhao et al., A multidimensional coding architecture of the
doi: 10.1016/j.molmet.2017.02.007; pmid: 28462073 pmid: 23643837 vagal interoceptive system. Nature 603, 878–884 (2022).
56. L. Steuernagel et al., HypoMap-a unified single-cell gene 79. G. T. Dodd et al., Insulin signaling in AgRP neurons regulates doi: 10.1038/s41586-022-04515-5; pmid: 35296859
expression atlas of the murine hypothalamus. Nat. Metab. 4, meal size to limit glucose excursions and insulin resistance. 102. D. Borgmann et al., Gut-brain communication by distinct
1402–1419 (2022). doi: 10.1038/s42255-022-00657-y; Sci. Adv. 7, eabf4100 (2021). doi: 10.1126/sciadv.abf4100; sensory neurons differently controls feeding and glucose
pmid: 36266547 pmid: 33637536 metabolism. Cell Metab. 33, 1466–1482.e7 (2021).
57. A. Secher et al., The arcuate nucleus mediates GLP- 80. S. M. Steculorum et al., AgRP neurons control systemic doi: 10.1016/j.cmet.2021.05.002; pmid: 34043943
1 receptor agonist liraglutide-dependent weight loss. J. Clin. insulin sensitivity via myostatin expression in brown adipose 103. W. S. Kim et al., Organ-specific, multimodal, wireless
Invest. 124, 4473–4488 (2014). doi: 10.1172/JCI75276; tissue. Cell 165, 125–138 (2016). doi: 10.1016/ optoelectronics for high-throughput phenotyping of
pmid: 25202980 j.cell.2016.02.044; pmid: 27015310 peripheral neural pathways. Nat. Commun. 12, 157 (2021).
58. C. Quarta et al., POMC neuronal heterogeneity in energy 81. C. Brandt et al., Food perception primes hepatic ER doi: 10.1038/s41467-020-20421-8; pmid: 33420038
balance and beyond: An integrated view. Nat. Metab. 3, homeostasis via melanocortin-dependent control of mTOR 104. J. M. Kefauver, A. B. Ward, A. Patapoutian, Discoveries in
299–308 (2021). doi: 10.1038/s42255-021-00345-3; activation. Cell 175, 1321–1335.e20 (2018). doi: 10.1016/ structure and physiology of mechanically activated ion
pmid: 33633406 j.cell.2018.10.015; pmid: 30445039 channels. Nature 587, 567–576 (2020). doi: 10.1038/
59. M. Feldman, C. T. Richardson, Role of thought, sight, smell, 82. H. Tsuneki et al., Food odor perception promotes systemic s41586-020-2933-1; pmid: 33239794
and taste of food in the cephalic phase of gastric acid lipid utilization. Nat. Metab. 4, 1514–1531 (2022). 105. D. Usoskin et al., Unbiased classification of sensory neuron
secretion in humans. Gastroenterology 90, 428–433 (1986). doi: 10.1038/s42255-022-00673-y; pmid: 36376564 types by large-scale single-cell RNA sequencing. Nat.
doi: 10.1016/0016-5085(86)90943-1; pmid: 3940915 83. C. López-Otín, M. A. Blasco, L. Partridge, M. Serrano, Neurosci. 18, 145–153 (2015). doi: 10.1038/nn.3881;
60. J. LeBlanc, M. Cabanac, Cephalic postprandial thermogenesis G. Kroemer, Hallmarks of aging: An expanding universe. Cell pmid: 25420068
in human subjects. Physiol. Behav. 46, 479–482 (1989). 186, 243–278 (2023). doi: 10.1016/j.cell.2022.11.001; 106. A. Mercado-Perez, A. Beyder, Gut feelings: Mechanosensing
doi: 10.1016/0031-9384(89)90024-3; pmid: 2623073 pmid: 36599349 in the gastrointestinal tract. Nat. Rev. Gastroenterol. Hepatol.

Brüning and Fenselau, Science 381, eabl7398 (2023) 29 September 2023 9 of 10


RES EARCH | R E V I E W

19, 283–296 (2022). doi: 10.1038/s41575-021-00561-y; 376–380 (2020). doi: 10.1038/s41586-020-2167-2; 145. K. M. Heppner et al., Expression and distribution of glucagon-
pmid: 35022607 pmid: 32296182 like peptide-1 receptor mRNA, protein and binding in the
107. C. Alcaino et al., A population of gut epithelial 126. J. L. Pauli et al., Molecular and anatomical characterization of male nonhuman primate (Macaca mulatta) brain.
enterochromaffin cells is mechanosensitive and requires parabrachial neurons and their axonal projections. eLife 11, Endocrinology 156, 255–267 (2015). doi: 10.1210/
Piezo2 to convert force into serotonin release. Proc. Natl. e81868 (2022). doi: 10.7554/eLife.81868; pmid: 36317965 en.2014-1675; pmid: 25380238
Acad. Sci. U.S.A. 115, E7632–E7641 (2018). doi: 10.1073/ 127. M. W. Schwartz, E. Peskind, M. Raskind, E. J. Boyko, 146. C. B. Jensen et al., Characterization of the glucagonlike
pnas.1804938115; pmid: 30037999 D. Porte Jr., Cerebrospinal fluid leptin levels: Relationship to peptide-1 receptor in male mouse brain using a novel
108. A. J. Treichel et al., Specialized mechanosensory epithelial plasma levels and to adiposity in humans. Nat. Med. 2, antibody and in situ hybridization. Endocrinology 159,
cells in mouse gut intrinsic tactile sensitivity. 589–593 (1996). doi: 10.1038/nm0596-589; pmid: 8616722 665–675 (2018). doi: 10.1210/en.2017-00812;
Gastroenterology 162, 535–547.e13 (2022). doi: 10.1053/ 128. R. C. Frederich et al., Leptin levels reflect body lipid content pmid: 29095968
j.gastro.2021.10.026; pmid: 34688712 in mice: Evidence for diet-induced resistance to leptin action. 147. M. H. Tschöp et al., Unimolecular polypharmacy for
109. L. Bai et al., Enteroendocrine cell types that drive food Nat. Med. 1, 1311–1314 (1995). doi: 10.1038/nm1295-1311; treatment of diabetes and obesity. Cell Metab. 24, 51–62
reward and aversion. eLife 11, e74964 (2022). doi: 10.7554/ pmid: 7489415 (2016). doi: 10.1016/j.cmet.2016.06.021; pmid: 27411008
eLife.74964; pmid: 35913117 129. J. Friedman, The long road to leptin. J. Clin. Invest. 126, 148. J. P. Frías et al., tirzepatide versus semaglutide once weekly
110. M. Hayashi et al., Enteroendocrine cell lineages that 4727–4734 (2016). doi: 10.1172/JCI91578; pmid: 27906690 in patients with type 2 diabetes. N. Engl. J. Med. 385,
differentially control feeding and gut motility. eLife 12, e78512 130. H. Cui, M. López, K. Rahmouni, The cellular and molecular 503–515 (2021). doi: 10.1056/NEJMoa2107519;
(2023). doi: 10.7554/eLife.78512; pmid: 36810133 bases of leptin and ghrelin resistance in obesity. Nat. Rev. pmid: 34170647
111. F. A. Duca, T. M. Z. Waise, W. T. Peppler, T. K. T. Lam, Endocrinol. 13, 338–351 (2017). doi: 10.1038/ 149. T. D. Müller, M. Blüher, M. H. Tschöp, R. D. DiMarchi, Anti-
The metabolic impact of small intestinal nutrient sensing. nrendo.2016.222; pmid: 28232667 obesity drug discovery: Advances and challenges. Nat. Rev.
Nat. Commun. 12, 903 (2021). doi: 10.1038/s41467-021- 131. C. Bjørbaek, J. K. Elmquist, J. D. Frantz, S. E. Shoelson, Drug Discov. 21, 201–223 (2022). doi: 10.1038/
21235-y; pmid: 33568676 J. S. Flier, Identification of SOCS-3 as a potential mediator of s41573-021-00337-8; pmid: 34815532

Downloaded from https://www.science.org at Universidade Estadual de Campinas on October 16, 2023


112. C. Ran, J. C. Boettcher, J. A. Kaye, C. E. Gallori, S. D. Liberles, central leptin resistance. Mol. Cell 1, 619–625 (1998). 150. J. P. H. Wilding et al., Weight regain and cardiometabolic
A brainstem map for visceral sensations. Nature 609, doi: 10.1016/S1097-2765(00)80062-3; pmid: 9660946 effects after withdrawal of semaglutide: The STEP 1 trial
320–326 (2022). doi: 10.1038/s41586-022-05139-5; 132. J. P. Thaler et al., Obesity is associated with hypothalamic extension. Diabetes Obes. Metab. 24, 1553–1564 (2022).
pmid: 36045291 injury in rodents and humans. J. Clin. Invest. 122, 153–162 doi: 10.1111/dom.14725; pmid: 35441470
113. H. J. Grill, M. R. Hayes, Hindbrain neurons as an essential hub (2012). doi: 10.1172/JCI59660; pmid: 22201683 151. K. Grzelka et al., A synaptic amplifier of hunger for
in the neuroanatomically distributed control of energy 133. M. D. Dorfman, J. P. Thaler, Hypothalamic inflammation and regaining body weight in the hypothalamus. Cell Metab. 35,
balance. Cell Metab. 16, 296–309 (2012). doi: 10.1016/ gliosis in obesity. Curr. Opin. Endocrinol. Diabetes Obes. 22, 770–785.e5 (2023). doi: 10.1016/j.cmet.2023.03.002;
j.cmet.2012.06.015; pmid: 22902836 325–330 (2015). doi: 10.1097/MED.0000000000000182; pmid: 36965483
114. C. Zhang et al., Area postrema cell types that pmid: 26192704 152. J. A. Sahel et al., Partial recovery of visual function in
mediate nausea-associated behaviors. Neuron 109, 134. L. Ozcan et al., Endoplasmic reticulum stress plays a central a blind patient after optogenetic therapy. Nat. Med. 27,
461–472.e5 (2021). doi: 10.1016/j.neuron.2020.11.010; role in development of leptin resistance. Cell Metab. 9, 35–51 1223–1229 (2021). doi: 10.1038/s41591-021-01351-4;
pmid: 33278342 (2009). doi: 10.1016/j.cmet.2008.12.004; pmid: 19117545 pmid: 34031601
115. C. W. Roman, V. A. Derkach, R. D. Palmiter, Genetically and 135. A. Jais, J. C. Brüning, Hypothalamic inflammation in obesity 153. C. J. Magnus et al., Ultrapotent chemogenetics for research
functionally defined NTS to PBN brain circuits mediating and metabolic disease. J. Clin. Invest. 127, 24–32 (2017). and potential clinical applications. Science 364, eaav5282
anorexia. Nat. Commun. 7, 11905 (2016). doi: 10.1038/ doi: 10.1172/JCI88878; pmid: 28045396 (2019). doi: 10.1126/science.aav5282; pmid: 30872534
ncomms11905; pmid: 27301688 136. C. M. Nasrallah, T. L. Horvath, Mitochondrial dynamics in the 154. L. E. Mickelsen et al., Single-cell transcriptomic analysis
116. C. W. Roman, S. R. Sloat, R. D. Palmiter, A tale of two circuits: central regulation of metabolism. Nat. Rev. Endocrinol. 10, of the lateral hypothalamic area reveals molecularly
CCKNTS neuron stimulation controls appetite and induces 650–658 (2014). doi: 10.1038/nrendo.2014.160; distinct populations of inhibitory and excitatory neurons.
opposing motivational states by projections to distinct brain pmid: 25200564 Nat. Neurosci. 22, 642–656 (2019). doi: 10.1038/
regions. Neuroscience 358, 316–324 (2017). doi: 10.1016/ 137. M. C. Vogt et al., Neonatal insulin action impairs hypothalamic s41593-019-0349-8; pmid: 30858605
j.neuroscience.2017.06.049; pmid: 28684275 neurocircuit formation in response to maternal high-fat 155. L. Madisen et al., Transgenic mice for intersectional targeting
117. I. Aklan et al., NTS catecholamine neurons mediate feeding. Cell 156, 495–509 (2014). doi: 10.1016/ of neural sensors and effectors with high specificity and
hypoglycemic hunger via medial hypothalamic feeding j.cell.2014.01.008; pmid: 24462248 performance. Neuron 85, 942–958 (2015). doi: 10.1016/
pathways. Cell Metab. 31, 313–326.e5 (2020). doi: 10.1016/ 138. S. Park, A. Jang, S. G. Bouret, Maternal obesity-induced j.neuron.2015.02.022; pmid: 25741722
j.cmet.2019.11.016; pmid: 31839488 endoplasmic reticulum stress causes metabolic alterations 156. L. E. Fenno et al., Comprehensive dual- and triple-feature
118. M. Q. Ludwig et al., A genetic map of the mouse dorsal vagal and abnormal hypothalamic development in the intersectional single-vector delivery of diverse functional
complex and its role in obesity. Nat. Metab. 3, 530–545 offspring. PLOS Biol. 18, e3000296 (2020). doi: 10.1371/ payloads to cells of behaving mammals. Neuron 107,
(2021). doi: 10.1038/s42255-021-00363-1; pmid: 33767443 journal.pbio.3000296; pmid: 32163401 836–853.e11 (2020). doi: 10.1016/j.neuron.2020.06.003;
119. G. D’Agostino et al., Appetite controlled by a cholecystokinin 139. A. A. van der Klaauw et al., Human semaphorin 3 variants link pmid: 32574559
nucleus of the solitary tract to hypothalamus neurocircuit. melanocortin circuit development and energy balance. Cell
eLife 5, e12225 (2016). doi: 10.7554/eLife.12225; 176, 729–742.e18 (2019). doi: 10.1016/j.cell.2018.12.009; AC KNOWLED GME NTS
pmid: 26974347 pmid: 30661757 We apologize to all colleagues whose contributions to this broad
120. R. P. Gaykema et al., Activation of murine pre-proglucagon- 140. S. B. Heymsfield et al., Recombinant leptin for weight loss in field of active research could not be covered because of the focus
producing neurons reduces food intake and body weight. obese and lean adults: A randomized, controlled, dose- and space limitations of the present Review. This work has
J. Clin. Invest. 127, 1031–1045 (2017). doi: 10.1172/JCI81335; escalation trial. JAMA 282, 1568–1575 (1999). doi: 10.1001/ received funding from the European Research Council (ERC) under
pmid: 28218622 jama.282.16.1568; pmid: 10546697 the European Union’s Horizon 2020 research and innovation
121. D. I. Brierley et al., Central and peripheral GLP-1 systems 141. J. Liu, J. Lee, M. A. Salazar Hernandez, R. Mazitschek, program (grant agreement ID 742106 to J.C.B. and ID 851778 to
independently suppress eating. Nat. Metab. 3, 258–273 U. Ozcan, Treatment of obesity with celastrol. Cell 161, H.F.) and the BMBF through the German Center for Diabetes
(2021). doi: 10.1038/s42255-021-00344-4; pmid: 33589843 999–1011 (2015). doi: 10.1016/j.cell.2015.05.011; Research (DZD) to J.C.B.; J.C.B. has received research funds
122. C. R. Boychuk et al., A hindbrain inhibitory microcircuit pmid: 26000480 through collaboration agreements with Novo Nordisk, Denmark, and
mediates vagally-coordinated glucose regulation. Sci. Rep. 9, 142. W. Ghusn et al., Weight loss outcomes associated with Sanofi Aventis, Germany, and is founder of Cerapeutix. H.F. has
2722 (2019). doi: 10.1038/s41598-019-39490-x; semaglutide treatment for patients with overweight or received research funds through collaboration agreements with
pmid: 30804396 obesity. JAMA Netw. Open 5, e2231982 (2022). doi: 10.1001/ Novo Nordisk, Denmark. We thank members of the Brüning and
123. I. E. de Araujo, Gustatory and homeostatic functions of the jamanetworkopen.2022.31982; pmid: 36121652 Fenselau labs for critical input and discussions. J.C.B. has received
rodent parabrachial nucleus. Ann. N. Y. Acad. Sci. 1170, 143. A. C. Rupp et al., Suppression of food intake by Glp1r/Lepr- consultancy fees from Eli Lilly and Company and Novo Nordisk.
383–391 (2009). doi: 10.1111/j.1749-6632.2009.03923.x; coexpressing neurons prevents obesity in mouse models. License information: Copyright © 2023 the authors, some rights
pmid: 19686163 J. Clin. Invest.e157515 (2023). doi: 10.1172/JCI157515; reserved; exclusive licensee American Association for the
124. C. A. Campos, A. J. Bowen, C. W. Roman, R. D. Palmiter, pmid: 37581939 Advancement of Science. No claim to original US government
Encoding of danger by parabrachial CGRP neurons. 144. I. Merchenthaler, M. Lane, P. Shughrue, Distribution of pre- works. https://www.science.org/about/science-licenses-journal-
Nature 555, 617–622 (2018). doi: 10.1038/nature25511; pro-glucagon and glucagon-like peptide-1 receptor messenger article-reuse
pmid: 29562230 RNAs in the rat central nervous system. J. Comp. Neurol. 403,
125. D. Y. Kim et al., A neural circuit mechanism for 261–280 (1999). doi: 10.1002/(SICI)1096-9861(19990111)403: Submitted 3 April 2023; accepted 31 August 2023
mechanosensory feedback control of ingestion. Nature 580, 2<261::AID-CNE8>3.0.CO;2-5; pmid: 9886047 10.1126/science.abl7398

Brüning and Fenselau, Science 381, eabl7398 (2023) 29 September 2023 10 of 10


Integrative neurocircuits that control metabolism and food intake
Jens C. Brüning and Henning Fenselau

Science 381 (6665), eabl7398. DOI: 10.1126/science.abl7398

Downloaded from https://www.science.org at Universidade Estadual de Campinas on October 16, 2023


Editor’s summary
The maintenance of body weight is regulated through a complex network of neuronal signaling, hormones, and gut-
brain interactions that allow adaptation to food intake and energy expenditure. Defining the nature of the underlying
neurocircuitry and how feedback regulation is achieved are important in understanding the development of obesity
and its attendant pathologies, such as type 2 diabetes mellitus and cardiovascular diseases. In a Review, Brüning and
Fenselau discuss our developing understanding of the neural pathways that underpin food intake, energy expenditure,
and systemic metabolism and discuss how this knowledge provides new therapeutic targets to treat obesity. —Gemma
Alderton

View the article online


https://www.science.org/doi/10.1126/science.abl7398
Permissions
https://www.science.org/help/reprints-and-permissions

Use of this article is subject to the Terms of service

Science (ISSN 1095-9203) is published by the American Association for the Advancement of Science. 1200 New York Avenue NW,
Washington, DC 20005. The title Science is a registered trademark of AAAS.

Copyright © 2023 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim
to original U.S. Government Works

You might also like