Physiological Integration of Taste and Metabolism

You might also like

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

The n e w e ng l a n d j o u r na l of m e dic i n e

Review Article

Nutrition in Medicine
Dan L. Longo, M.D., Editor

Physiological Integration of Taste


and Metabolism
Josephine M. Egan, M.D.​​

“Y
ou don’t know what you’re missing till it’s gone” is a truism From the Diabetes Section, Laboratory of
that certainly applies to taste. It took a pandemic for taste to get attention. Clinical Investigation, National Institute
on Aging, National Institutes of Health,
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections Baltimore. Dr. Egan can be contacted at
can cause acute loss or distortions of taste as a result of infection within taste buds1 ­eganj@​­grc​.­nia​.­nih​.­gov or at the National
and, in some cases, can result in long-term taste dysfunction.2,3 Since chemosensory Institutes of Health, National Institute
on Aging, Biomedical Research Center,
disorders can substantially dampen a person’s enjoyment of life, this is an opportune 251 Bayview Blvd., Ste. 100, Baltimore,
time to appreciate recent advances in our understanding of taste. It is time to let go of MD 21224.
old ideas, such as the myth of the tongue taste map (which persists in the collective N Engl J Med 2024;390:1699-710.
consciousness despite decades of research debunking it) and the notion of taste as DOI: 10.1056/NEJMra2304578
limited to the mouth. Research reveals that downstream signaling of extraoral taste Copyright © 2024 Massachusetts Medical Society.

receptors regulates our physiological balance long after conscious gustation has faded.
In this review, I highlight two important areas of progress. First, I provide an up-
date on taste physiology and biochemistry, the receptors mediating prototypical taste
perception, and taste neurotransmission. Second, I examine taste in the context of
recent press and research reports on the health consequences of added sugars and
nonnutritive sweeteners. This area of research offers delectable “food for thought,”
highlighting the role of extraoral taste receptors and their putative involvement in
food intake, metabolism, and obesity.

Ta s te R ecep t or s a nd Ta s te T r a nsduc t ion Mech a nisms


Taste perception begins when nonvolatile chemicals in food, called tastants, are
placed in the mouth and activate taste receptor cells (TRCs), which recognize all
five primary taste sensory qualities: sweet, umami (savory), bitter, salty, and sour.
There is also evidence for the taste of “fat.”4 Taste is described in affective ways as
being pleasant (appetitive) or aversive (revulsive). TRCs are present in taste buds
buried in taste papillae, which are fleshy protuberances on the tongue (Fig. 1).
Once activated, TRCs send tastant information that the brain integrates with smell
and trigeminal-nerve transduction (registering texture, temperature, and pain) to
craft a complex perception of whatever we consume.
TRCs are morphologically classified into types I, II, and III, which can be subclas-
sified on the basis of differences in TRC function, molecular markers, or both (Fig. 1).
We also acknowledge a fourth cell, type IV, at the base and sides of taste buds.5 Al-
though there was once a belief that taste-sensing cells responsive to each of the basic
taste qualities were concentrated in separate areas, this is not accurate. TRCs sensitive
to each tastant are present in taste buds across the tongue, meaning that there
is no taste map representing regions of the tongue corresponding to specific tastes.5
In taste buds, glial-type type I TRCs support the structure of the buds. On the
plasma membranes of these cells, the enzyme NTPDase2 degrades intragemmal
ATP secreted by type II cells in response to tastants. Type II TRCs detect sweet,

n engl j med 390;18 nejm.org May 9, 2024 1699


The New England Journal of Medicine
Downloaded from nejm.org on June 17, 2024. For personal use only.
No other uses without permission. Copyright © 2024 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A
TASTE BUD
Circumvallate papillae (CVP)
Type I cell
Type II cell
Type III cell

Foliate papillae (FLP)

TONGUE

Fungiform papillae (FFP)

Intragemmal
nerve fiber
Type IV cell

B
TYPE II CELL TYPE I CELL TYPE II CELL TYPE III CELL
Sweet/umami/bitter Salt appetitive Sour

Adenosine OTOP1
ENaC
T1R or AMP H+
T2R Na+
ADP

ATP 5-HT
NTPDase2

Mitochondrion

Mitochondrion
ATP
ATP

CALHM1/3
CALHM1/3
P2X2/3 5-HTR
P2X2/3

Intragemmal nerve fiber

umami, and bitter tastants through subfamilies ing cascade follows a common downstream path-
of G protein–coupled receptors (GPCRs): the way, involving phospholipase Cβ2, inositol tri-
TAS1R (TAS1R1, TAS1R2, and TAS1R3) and phosphate (IP3) production, which causes Ca2+
TAS2R classes. These taste receptors initiate in- release from the endoplasmic reticulum, followed
tracellular signal transduction by stimulating the by Ca2+-dependent activation of transient receptor
heterotrimeric G protein α-gustducin. The result- potential melastatin 5 (TRPM5) channels and cul-

1700 n engl j med 390;18 nejm.org May 9, 2024

The New England Journal of Medicine


Downloaded from nejm.org on June 17, 2024. For personal use only.
No other uses without permission. Copyright © 2024 Massachusetts Medical Society. All rights reserved.
Physiological Integr ation of Taste and Metabolism

Figure 1 (facing page). Anatomy and Physiology of the Concentrations of NaCl between 30 and 150 mM
Tongue and Taste Buds. rely on the epithelial sodium channel (ENaC) in
There are three types of papillae (Panel A): fungiform a subset of TRCs for salty, appetitive transmis-
(FFP), foliate (FLP), and circumvallate (CVP). CVP, up sion. In mice, this channel is blocked by amiloride.
to 11 in number, are the largest and are easily visual- ENaCs allow Na+ influx, followed by cell depolar-
ized on the back of the tongue. FLP are parallel folds on
ization, action potential generation, and ATP re-
the sides of the tongue. FFP, several hundred in num-
ber and just visible to the naked eye, are scattered on lease through CALHM1/3 channels onto afferent
the anterior tongue, interspersed with a fourth papilla fibers.10 This process has not been confirmed in
type, filiform, that do not contain taste buds; they help humans, and humans actually seem appetitive-
cleanse the mouth, move food around, and serve mech- salty-taste insensitive to amiloride.11 A possible
anosensory functions by responding to touch, pain, and
explanation is that human ENaCs are structurally
temperature. Each taste bud has a pore surrounded
by a bushy network of villi of the taste receptor cells dissimilar or that appetitive salty taste in humans
(TRCs). Humans may have up to 4500 taste buds, but is mediated by a subpopulation of TRCs that is
the number varies greatly, and 50 to 60% of them are completely different from the subpopulation in
in CVP. Each taste bud, regardless of location, contains mice. Very high NaCl concentrations, which are
about 60 TRCs (morphologic types I, II, and III) and
aversive to both humans and mice, may activate
precursor cells (type IV). Types I, II, and III can be sub-
classified on the basis of differences in TRC function, responses in bitter type II TRCs, as well as type
molecular markers, or both (Panel B). Type I TRCs, which III cells12 and perhaps also free nerve endings of
wrap around the other TRCs, contain NTPDase2 on the trigeminal nerve. Bitter type II TRCs express
their plasma membrane, which degrades ATP released multiple TAS2Rs, and at least 25 TAS2R genes are
from type II TRCs. The G protein–coupled receptors,
known to be expressed in humans.13 These recep-
necessary for sweet taste (TAS1R3 and TAS1R2), umami
taste (TAS1R3 and TAS1R1), and bitter taste (TAS2Rs) tors are capable of discerning thousands of bit-
are present on type II TRCs. In response to their re- ter tastants, since some receptors are narrowly
spective tastants, they release ATP as their signaling tuned to one or a few tastants, whereas others are
molecule through CALHM1/3 channels onto purinergic broadly tuned to many bitter tastants.13
receptors on intragemmal nerve fibers throughout taste
Type III TRCs have dense synaptic vesicles
buds. Sodium ions, responsible for a pleasant salty
taste, enter through epithelial sodium channels (ENaCs) containing classic neurotransmitters such as
on TRCs, and also use ATP as their signaling molecule. 5-hydroxytryptamine (5-HT), which they release in
Type III TRCs, the only TRC type with classical neuronal response to acid stimulation. These are the only
synapses, contain a proton channel (OTOP1) that detects TRCs that form classical synapses with nearby
sour or acidic tastants leading to neurotransmitter re-
nerve fibers. They are also unique in containing the
lease, such as 5-hydroxytryptamine (5-HT).
proton channel OTOP1, which is necessary for
perceiving sour taste (acid sensing, such as citrus
minating in cell depolarization and ATP release fruits) and ammonium chloride (breakdown prod-
through specialized channels (CALHM1/3).6 ATP is ucts of amino acids and decaying meats).14 For most
a bona fide TRC neurotransmitter that activates species, sour taste is aversive, but humans enjoy
purinergic receptors on nerve fibers, which then sour taste triggered by acidic foods, up to a certain
transduce tastant information to the brain. concentration that seems to be individually deter-
The sweet and umami type II TRCs express mined, with higher concentrations becoming aver-
TAS1R3, a coreceptor that forms heterodimers sive, probably because they induce pain.15 Possible
with TAS1R2 and TAS1R1 to detect sweet and reasons for this affinity to sour taste are that acids
umami, respectively. In some situations, TAS1R3 may inhibit harmful microbial growth, signal the
itself is a low-affinity sweet receptor,7 and TAS1R3 presence of amino acids (which might also trigger
homodimers, especially in nontaste tissue, can umami perception), or indicate that a food may
function as cell-surface glucose sensors.8 Recently, have been fermented and may be psychoactive (e.g.,
researchers discovered that the Cl− ion, found in alcohol). Also, sour taste may have evolutionarily
table salt (sodium chloride [NaCl] concentration, guided humans toward sources of vitamin C.15
<30 mM), is also a ligand for TAS1R3 and evokes Type III TRCs also respond to carbon dioxide
appetitive preferences, adding to the notion that by means of the enzyme carbonic anhydrase 4,
TAS1R3 transmits pleasant tastes corresponding which integrates with other somatosensory inputs
to that which is necessary for life (i.e., energy that are necessary for the taste of carbonation.16
and salt).9 In addition, optogenetic research (i.e., research

n engl j med 390;18 nejm.org May 9, 2024 1701


The New England Journal of Medicine
Downloaded from nejm.org on June 17, 2024. For personal use only.
No other uses without permission. Copyright © 2024 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Sensory cortex
(postcentral gyrus)

Gustatory cortex

Ventral
posteromedial
nucleus

Principal nucleus

Trigeminal nerve
(CN V)
Trigeminal ganglion

Ophthalmic nerve
(CN V1)

Maxillary nerve
(CN V2)
Mandibular nerve
(CN V3) Nucleus of the
tractus solitarius
Geniculate ganglion

Petrosal ganglion
Chorda tympani
Nodose ganglion

Facial nerve
(CN VII)
Glossopharyngeal
nerve (CN IX)
Superior laryngeal
nerve (CN X)
Vagus nerve
(CN X)

in which light is used experimentally to influ- At the base and sides of taste buds are type IV
ence cell behavior) in mice suggests that these cells, which are precursors for the other three
cells are involved in recognizing the taste of TRC types. Although mature TRCs differentiate
water.17 Further research into type III tastant throughout life, mouse organoid culture studies
detection and subsequent signal integration in suggest that this process changes over time.19
the brain should help clarify how we distinguish Investigators are working to fully characterize
among water, sour, and ammonium stimuli. Fi- TRC precursors in humans, a project that is rele-
nally, a subset of type III TRCs in mice has been vant to an understanding of long-term taste loss
described as responsive to a wide range of taste and dysfunction as a result, for example, of SARS-
stimuli, such as sweet, bitter, and umami.18 CoV-2 infections.2,5 Despite interest in human taste

1702 n engl j med 390;18 nejm.org May 9, 2024

The New England Journal of Medicine


Downloaded from nejm.org on June 17, 2024. For personal use only.
No other uses without permission. Copyright © 2024 Massachusetts Medical Society. All rights reserved.
Physiological Integr ation of Taste and Metabolism

Figure 2 (facing page). Transmission of Taste Information.


found that sweet TRCs induce appetitive behav-
Fibers innervating the taste buds transmit signals to
iors, whereas bitter and sour TRCs induce aversive
the gustatory region of the nucleus of the tractus soli- behaviors.22 Since downstream signaling is the
tarius (NTS), then to the thalamus and the gustatory same for sweet, umami, and bitter, a labeled-line
cortex (pathway depicted in red). Each taste bud is in- type of transmission to the insula can be inferred,
nervated by primary intragemmal gustatory fibers, whereby dedicated nerve fibers and neurons de-
which extensively branch and connect with multiple
taste buds, interacting with many TRCs. This results in
code the three tastants, as well as NaCl. Proof
electrical activity that reflects the input from numer- of this comes from two-photon calcium imaging,
ous TRCs. Taste buds in the fungiform papillae are in- which has shown spatial segregation in the gus-
nervated by sensory neurons of the geniculate gangli- tatory cortex, with neurons that responded to
on, traveling through the chorda tympani branch of the each tastant.23
facial nerve (cranial nerve [CN] VII). Taste buds in the
posterior third of the tongue are innervated by sensory
Such strict specificity is hotly debated, how-
neurons of the petrosal ganglion, traveling through the ever, and other evidence favors combinatorial
lingual branch of the glossopharyngeal nerve (CN IX). coding, in which taste information is transmitted
Isolated taste buds in the palate are innervated by the through neuronal circuits that have patterns of
greater superficial petrosal branch of CN VII, whereas impulse firings. (For an in-depth discussion, see
those on the epiglottis and esophagus receive innerva-
tion from the superior laryngeal branch of the vagus
Roper.24) A parsimonious explanation is as fol-
nerve (CN X). Sensory fibers from CNs VII, IX, and X lows. At low concentrations of a single stimulus,
enter the medulla, synapsing on a slender column of a labeled line of taste transduction is operational
cells within the gustatory region situated in the rostral and is exquisitely elegant in separating appetitive
and lateral part of the NTS. From there, neurons proj- from aversive stimuli (life or death, to be or not
ect to the ventral posteromedial nucleus of the thala-
mus. Next, neurons project to the anterior insula and
be, to swallow or not to swallow — immediate
frontal operculum in the cerebral cortex, which facili- decision requirements). But complex food re-
tates the conscious perception and discrimination of quires a cross-fiber pattern with nerve fibers that
tastes. Taste sensations often include somatosensory are more broadly responsive and that perhaps
aspects like texture, temperature, and responses to respond maximally only to particular food con-
spicy and minty foods. This component is transmitted
by the branches of the trigeminal nerve (CN V), origi-
tents, allowing for comparison of activity across
nating in the trigeminal ganglion. From there, informa- the whole fiber repertoire so that the brain can
tion goes to the principal nucleus in the caudal pons, extract information related to total food quality.
which also projects to the ventral posteromedial nucleus Adding to the orchestration of taste percep-
of the thalamus, and finally to the somatosensory cortex tion are the hormones produced in TRCs that
in the brain’s parietal lobe (pathway depicted in blue).
modulate TRC signaling. Hormones produced by
enteroendocrine cells of the gut (cholecystokinin,
buds, studies in humans encounter methodologic glucagon-like peptide 1 [GLP-1], ghrelin, pep-
constraints. Although mouse studies primarily tide YY, and vasoactive intestinal peptide) and
use circumvallate papillae because they contain islets of Langerhans (glucagon and insulin), as
half the taste buds, most human data come from well as by some central nervous system neurons
fungiform papillae, since it is the only papilla (neuropeptide Y and vasoactive intestinal pep-
type proven to regrow and is easily biopsied in tide), are also synthesized in TRCs.25 More work
living subjects. There is large variation in the is needed to uncover all their functions in taste
density of fungiform papillae and the number of buds, but so far we know that receptors for some
taste buds between persons, and these decrease of those hormones, such as GLP-1, are present
as people age, a phenomenon reported in longitu- on the intragemmal nerve fibers in taste buds
dinal20 and cross-sectional21 studies. where GLP-1 receptor (GLP-1R) activation in mice
Since TRCs undergo rapid turnover, maintaining modulates sweet perception (Fig. 3).26
neurosensory integrity (i.e., sweet taste receptors We are also now aware of the presence of tastant
signaling to “sweet” neurons) is essential (Fig. 2). signal transduction machinery in nontaste tissue,
The labeled-line model of taste transmission con- including enteroendocrine cells (Fig. 4). Researchers
nects taste reception and signal integration in the have discovered diverse roles for extraoral taste re-
brain. Using chemogenetic manipulation in ro- ceptors, such as regulating male fertility27 and pro-
dents to activate intracellular signaling that is tecting tissue in the pulmonary vasculature.28 The
independent of cell surface receptors, researchers gut has emerged as a site for exploring the involve-

n engl j med 390;18 nejm.org May 9, 2024 1703


The New England Journal of Medicine
Downloaded from nejm.org on June 17, 2024. For personal use only.
No other uses without permission. Copyright © 2024 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

ment of taste receptors and their downstream sig- but our innate aversion to such tastants can be
naling pathways in appetite, nutrition, and disease. overcome by acquired preference and masking
with sweet tastants. Mutations in the TAS2R
genes and other genotypes that increase bitter
Rol e of Ta s te in Fo od In ta k e ,
Me ta bol ism, a nd Obe si t y taste thresholds are associated with increased
consumption of bitter beverages, such as alcohol
Obesity and obesity-related noncommunicable and coffee, a behavior that probably also reflects
diseases are at epidemic levels, with one projection a learned liking of their physiological effects.34
forecasting that by 2030, nearly 1 in 2 adults in the Since taste functions to guide what we should
United States will be obese.29 Although there are not ingest as much as what we should, altera-
many contributing factors,30 those relevant to this tions in taste perception can change our dietary
review concern the ways in which the contempo- patterns.35 Our diet also shapes our taste. West-
rary food environment, with its cornucopia of ap- ern diets rich in fat and carbohydrates change
petitive offerings, encourages overeating by stimu- the proteomic landscape of the tongue,36 and
lating our deeply ingrained reward systems. Taste, obese, diabetic mice and their offspring have an
by guiding us toward gastronomic delights (i.e., increased preference for sweet stimuli.37
tasty, energy-containing food) and away from Current observations suggest that obesity is re-
dangerous toxins, functions as an evolutionary lated to disruptions in the neural pathways that
gatekeeper for the substances that enter our body. encourage reward-related eating and suppress ho-
Sweet preference is innate, developed well before meostatic feedback that curbs hunger, although we
birth, and consuming sweet tastants triggers sat- have yet to fully elucidate the precise physiological
isfaction through central reward pathways.31 Stud- mechanisms.38 However, a direct connection be-
ies in humans have shown both immediate and tween obesity and taste perception in humans is
delayed dopamine signaling in response to palat- not proven. Overweight is associated with a pro-
able food, which suggests that reward pathways clivity for energy-dense, ultraprocessed foods39 and
respond to oral sensation and postingestive pro- for sweet tastants and fats. Researchers have pro-
cessing in the gut.32 There is even some evidence posed impaired lipid perception as a reason for
suggesting that sugar can be addictive in the lipid overconsumption in some obese people.40
same way that nicotine is.33 There is also evidence of different taste percep-
On the flip side of sweet and umami, bitter tion in obesity-prone populations,41 with reports
and sour tastes detect potentially toxic substances, that increased weight is associated with decreased

A B GLP-1 C Neuropeptide Y (NPY)

10 µm 50 µm

Figure 3. Immunohistochemical Features and Examples of Hormones in Human FFP.


Two typical-appearing taste buds are present in an FFP (Panel A, hematoxylin and eosin), and GLP-1 and neuropeptide Y (NPY) are pres-
ent in type II TRCs in taste buds (Panels B and C, respectively, immunofluorescence staining). In mice, GLP-1 maintains or enhances
sweet perception through GLP-1 receptors on the intragemmal nerve fibers, and NPY enhances sweet and bitter perception through Y1
receptors. In FFP from humans, there are approximately 60 TRCs in each bud, at least 50% of which are type II. However, as shown in
these examples, the size of the cells varies greatly between persons and even varies among individual taste buds.

1704 n engl j med 390;18 nejm.org May 9, 2024

The New England Journal of Medicine


Downloaded from nejm.org on June 17, 2024. For personal use only.
No other uses without permission. Copyright © 2024 Massachusetts Medical Society. All rights reserved.
Physiological Integr ation of Taste and Metabolism

papilla density42 and higher sweetness thresh- Me ta bol ic C onsequence s of


olds.43 Other studies have shown no association Nonnu t r i t i v e S w ee tener s
between perceived taste intensity and obesity.44
These variations in study findings are likely to As studies implicated added sugars in the rising
reflect, at least in part, differences in the meth- incidence of cardiovascular disease,54 diabetes,55
odologies and scales used to characterize taste and obesity,56 public health messaging began
intensity and perception, neurophysiological di- recommending restrictions on sugar intake,57,58
versity among study participants, and population- and food manufacturers sought sugar substitutes
level confounding factors. Studies examining (Table 1). Despite the subsequent proliferation of
molecular signaling and gene expression have the nonnutritive sweeteners,59 however, obesity and
potential to provide more clarity. A study in mice metabolic dysfunction continued their stubborn
suggested that obesity-related proinflammatory upward trajectory. There is ongoing discussion
cascades were responsible for diminished taste about whether nonnutritive sweeteners are them-
bud abundance and renewal in obese mice,45 and selves contributing to the rising obesity levels and
genomic work in humans has identified increased related coexisting conditions, but any link be-
inflammatory and decreased taste-associated gene tween these sweeteners and long-term morbidity
expression in obese persons.46 Many other factors lacks consensus. In 2023, the World Health Orga-
can also influence taste perception, including nization released new guidelines discouraging
age,47,48 medications, and disease.1,49 the use of nonnutritive sweeteners to lose weight
On a broader level, taste transduction machin- and reduce the risk of noncommunicable diseas-
ery affects health through its impact on the es, a recommendation based on a large system-
body’s hormone response to food. GLP-1, classi- atic review.60 In brief, the literature suggests that
cally characterized as an incretin hormone that there may be short-term weight loss associated
enhances glucose-mediated insulin secretion, is with the initiation of nonnutritive sweetener con-
secreted by enteroendocrine cells that are present sumption as a result of lower calorie intake, but
from the duodenum to the early colon. (The sec- more work is needed to understand the possible
ond known incretin is glucose-dependent insuli- consequences of long-term consumption.61 Some
notropic polypeptide [GIP].) Secretion of GLP-1 is studies have shown an elevated risk of major car-
regulated by TAS1Rs; sodium–glucose cotrans- diovascular events and disease,62 whereas others
porter 1 (SGLT1); fatty acids through GPCR40, have shown no such effect.63 Nonnutritive sweet-
GPCR119, and GPCR120; bile acid receptors; en- eners may dampen T-cell–mediated immune re-
dogenous cannabinoids; and microbial products. sponses,64 and their long-term use is implicated
GLP-1 inhibits gastric emptying, causing gastric in an increased incidence of type 2 diabetes.55
distention through ileal neurons that connect by Long-term, observational studies in humans sug-
means of celiac ganglia to gastric neurons and gest a link between nonnutritive sweeteners and
ultimately contribute to food rejection.50 In ad- obesity,65,66 and although such studies must often
dition, long-acting GLP-1R agonists affect re- contend with the possibility of reverse causality,
ward behavior and food desirability, possibly current observations highlight the need for a
decreasing food intake, at least in part by reduc- better understanding of possible mechanisms.
ing palatability.51 GLP-1Rs, as stated above, are One proposed mechanism for how consump-
present on intragemmal nerve fibers, where tion of nonnutritive sweeteners induces meta-
their activation in mice diminishes the percep- bolic dysfunction is an uncoupling of sweet taste
tion of sweetness.26 These findings may help from caloric value. The seminal studies on this
explain, in addition to gastric and CNS effects, mechanism showed that mice that had been
some other nonpancreatic actions of GLP-1R conditioned to associate sweetness with calories
agonists, three of which (liraglutide, semaglu- gained less weight and consumed less food than
tide, and tirzepatide) are approved by the Food mice that could not rely on sweet taste to predict
and Drug Administration for use in weight man- caloric value.66,67 Researchers proposed that these
agement. Liraglutide and semaglutide have been results showed how nonnutritive sweeteners alter
found to promote weight loss possibly by damp- Pavlovian associations between sweet taste and
ening a preference for appetitive tastes, such as calories, leading to disrupted metabolic regula-
sweet and umami.52,53 tion and changing eating behavior.67 The first

n engl j med 390;18 nejm.org May 9, 2024 1705


The New England Journal of Medicine
Downloaded from nejm.org on June 17, 2024. For personal use only.
No other uses without permission. Copyright © 2024 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

TAS1R (sweet/umami) TAS2R (bitter)

Brain Brain

Chemosensory cells Chemosensory cells

Nose, sinuses

Vagus nerve
Thyroid

Trachea

Adipose tissue

Bronchi

Heart Heart

Stomach Stomach

Pancreas

Bile ducts

Kidney Kidney

Small intestine Small intestine

Colon Colon

Bladder

Testis Testis

steps in proving the biologic plausibility of “un- Recent work has drawn a distinction between
coupling” would be to uncover the different path- two glucose-sensing pathways: the traditional TAS1R
ways for sensing energy content as compared with sweet tasting pathway and the pathway involving
taste qualities and then to determine whether SGLTs, which transport glucose but not nonnutri-
natural sugars and nonnutritive sweeteners have tive sweeteners.68 Studies exploring differential
differential effects on these pathways. brain responses in mice have identified distinct

1706 n engl j med 390;18 nejm.org May 9, 2024

The New England Journal of Medicine


Downloaded from nejm.org on June 17, 2024. For personal use only.
No other uses without permission. Copyright © 2024 Massachusetts Medical Society. All rights reserved.
Physiological Integr ation of Taste and Metabolism

Figure 4 (facing page). Extraoral Taste Receptors.


resonance imaging study showed that as compared
TAS1Rs are present not just on the tongue but through-
with glucose, sucralose initiated a weakened sati-
out the body in tissues such as the gut, brain, pancreas, ety response in the hypothalamus, as well as a
bladder, bone, adipose tissue, airway epithelium, skele- prolonged hedonic response in the ventral tegmen-
tal muscle, and testes. There are also extraoral TAS2Rs tal area, which plays a key role in the mesolimbic
in the larynx, gut, and brain and on immune cells, as pathway that drives reward-seeking behavior.72
well as throughout the respiratory and genitourinary
systems. TAS2Rs, along with TAS1R3 and α-gustducin,
Although these results support the need to draw
are also present in testes. Animals in which TAS2Rs are a distinction between the neural effects of sugars
used to avoid noxious foods also appear to have highly and the neural effects of nonnutritive sweeteners,
efficient spermatogenesis and, as a consequence, may most studies have not investigated the possibility of
produce more offspring. OTOP1, the proton channel in combined effects. Recent studies examining both
type III TRCs, was first identified in the vestibular sys-
tem, where it is necessary for the formation of calcium
gastrointestinal and neurologic responses to sweet
carbonate–based otoconia and otoliths. It is also ex- taste complicate a strict uncoupling model. One
pressed in brown adipose cells. elegant human study showed that pairing sucralose
with carbohydrate intake impaired the central re-
ward response to sugar and induced insulin insen-
Table 1. Common Sugars, Sweeteners, and Amino Acids sitivity after only 2 weeks.73 These effects were not
and Proteins Known to Taste Sweet to Humans.
observed in the groups consuming carbohydrates
Sugars or sucralose independently, and there was no
Glucose change in perceptual sensitivity to taste. These re-
Fructose sults contradict the theory that nonnutritive sweet-
Maltose eners lead to metabolic dysfunction because of the
Sucrose
Galactose lack of calories or disturbance of the conditioned
Nonnutritive and low-calorie sweeteners
oral sweet sensation response. The authors instead
theorize that the combination of carbohydrate and
Aspartame
Acesulfame potassium (Ace-K) sucralose overactivated glucose transport through
Sucralose binding of both carbohydrate-derived glucose and
Neotame sucralose to gut sweet taste receptors. The results
Advantame
Saccharin of this study do not contradict the findings in
Cyclamate the uncoupling experiments in animals, although
Alitame they elucidate a different mechanism.
Certain steviol glycosides
Mogrosides: extracts from Siraitia grosvenorii (monk fruit, More studies of this kind offer a promising
also known as Swingle fruit or luo han guo) path for understanding the effects of nonnutritive
Sweet-tasting amino acids and proteins sweeteners on human physiology. However, many
D-phenylalanine studies use more than one nonnutritive sweetener,
D-serine and it can be hard to compare results across dif-
D-tryptophan ferent sweeteners because of variations in such
Monellin
Brazzein factors as sweetener intensity and binding affini-
Thaumatin ties and mechanisms. These problems, combined
with the inherent difficulties in performing long-
term human studies, make blanket conclusions
neural pathways for sweet sensing as compared and establishment of causation difficult. Further
with energy sensing69 and have shown how pro- work that compares habitual users of nonnutritive
longed activation of reward pathways in the sweeteners with nonhabitual users and investi-
brain leads to compulsive sucrose consumption.70 gates the effects of nonnutritive sweeteners paired
Even in flies, a high-fat, high-sugar diet impairs with caloric sources will help clarify the neuro-
central processing of sweet taste, weakening satia- physiological effects of sweeteners in the context
tion and encouraging overeating.71 Neuroimaging of typical food consumption patterns. These in-
is helping to corroborate these findings in hu- vestigations will not only strengthen causal and
mans, and indeed some studies suggest that hu- mechanistic links but will also supply crucial evi-
mans process sugar differently from nonnutritive dence to better inform dietary recommendations
sweeteners. For example, one functional magnetic and introduce new therapeutic paths.

n engl j med 390;18 nejm.org May 9, 2024 1707


The New England Journal of Medicine
Downloaded from nejm.org on June 17, 2024. For personal use only.
No other uses without permission. Copyright © 2024 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Ta s te , the Vagus, a nd the sweeteners not only in food and drink but also in
En teroend o cr ine S ys tem products such as toothpaste).
Advances in optogenetic work are enhancing
Despite the competing mechanistic theories, the our understanding of how integrated gut–brain
evidence thus far supports the notion that nonnu- signaling of taste sensation helps drive behavior
tritive sweeteners and natural sugars elicit distinct and metabolic outcomes. A spate of work suggests
homeostatic and hedonic responses in the body. that long-term energy homeostasis requires signal
Therefore, it is more accurate to call nonnutritive integration from nutrient-sensing receptors in the
sweeteners simulacra of sugars, not sugar substi- gut.81-83 In blinded testing, humans and mice prefer
tutes. TRC machinery in extraoral tissues, espe- nutritive sugars over noncaloric sweeteners, and
cially the gut, and physiological mechanisms of when scientists genetically manipulate mice to be
tastant binding have been “sweet” avenues for unable to taste sweetness in their taste buds, the
exploring the unique effects of sugars as com- mice still develop a preference for sugar over non-
pared with nonnutritive sweeteners. The latter nutritive sweeteners. This preference is modulated
have been linked to gut epithelial-cell death and by a population of neurons that are activated by the
increased gut-wall permeability74 and to altera- gut–brain axis to respond to sugar but not to non-
tions in the composition of the gastrointestinal nutritive sweeteners.81 Some enteroendocrine cells
microbiota (affecting pathways such as those in- have long, basal extensions, called neuropods,
volved in purine metabolism, glycolysis, and fatty which synapse with vagal afferent fibers to directly
acid synthesis),75 which have potential down- convey sugar-sensing information to the brain
stream consequences for hormone secretion, within milliseconds.81,82 Vagal afferent signals enter
metabolic homeostasis, and obesity. More directly, the medulla, and from there the information travels
rodent studies suggest that activation of the in a network of many overlapping circuits that are
sweet taste receptors in the gut by sugars and involved in eating signaling in the hypothalamus.
nonnutritive sweeteners is linked to hormonal This homeostatic effect leads to changes in con-
up-regulation and increased SGLT1 expression,76 sumption by altering how the brain learns the
glucose transporter 2 (GLUT2) induction,77 and value of sugar and controls behavior through
glucose absorption. There may be long-term con- dopaminergic reward circuits in the basal ganglia.
sequences, as suggested by a recent study in mice Sugars and nonnutritive sweeteners stimulate
that showed a dose–response effect of increased different neurotransmitter responses from cells
glucose absorption in the gut after long-term ex- containing cholecystokinin and cells containing
posure to sucralose.78 However, these effects and GLP-1. Nonnutritive sweeteners stimulate puri-
the role of sweet taste receptors are debated,79 as nergic neurotransmission only through TAS1Rs
is the existence of such a mechanism in humans, (exactly as they stimulate neurotransmission in
with decades of work providing evidence both for TRCs in taste buds), whereas glucose, which also
and against an effect of nonnutritive sweeteners activates TAS1Rs, has an additional pathway,
on incretin release.80 dependent on downstream signaling from SGLT1,
More large-scale clinical studies are necessary that stimulates glutamatergic neurotransmission
to start resolving these seeming inconsistencies, from the neuropods.83 The preference for nutritive
which reflect differences in study design and sugars over nonnutritive sweeteners is dependent
study populations. Current research is beginning on the glutamatergic signaling pathway, probably
to address these challenges, which will require because it stimulates vagal afferents that function
grappling with metabolic diversity among various as appetitive or reward neurons.84 Moreover, the
population groups (e.g., persons without obesity role of enteroendocrine cells in food preference
vs. persons with obesity and persons who habitu- conditioning and gastrointestinal control of re-
ally use nonnutritive sweeteners vs. persons who ward circuits is, in part, how the gut itself influ-
do not), the many different types and dose-depen- ences food intake.
dent effects of nonnutritive sweeteners, and the
effect of such sweeteners under typical consump- C onclusions
tion patterns (with the recognition, for example,
that nonnutritive sweeteners are rarely consumed The physiology of taste provides insight into our
in isolation and that humans are exposed to these relationship with food and our metabolic well-

1708 n engl j med 390;18 nejm.org May 9, 2024

The New England Journal of Medicine


Downloaded from nejm.org on June 17, 2024. For personal use only.
No other uses without permission. Copyright © 2024 Massachusetts Medical Society. All rights reserved.
Physiological Integr ation of Taste and Metabolism

being. Research has long dispelled the taste map Disclosure forms provided by the author are available with the
full text of this article at NEJM.org.
myth and is now venturing into new territory,
providing a complex understanding of how tas- I thank all the staff of the Diabetes Section, Laboratory of Clini-
cal Investigation, National Institute on Aging, for their input; Dr.
tants activate hedonic and homeostatic pathways Qin Yao for the original images of human fungiform papillae; Tom
and a recognition of the gut’s involvement in food Wynn and Lauren Brick of the visual media core of the Intramural
intake. This knowledge may, in turn, inform up- Research Program, National Institute on Aging, and Dr. Caio Ma-
zucanti, also of the Intramural Research Program, National Insti-
dates to dietary guidelines and clinical practice tute on Aging for initial drafts of the figures; and Adeline Choo
guidelines for what constitutes an ideal diet. for assistance in preparing an earlier version of the manuscript.

References
1. Doyle ME, Appleton A, Liu Q-R, Yao Q, The molecular receptive ranges of human blockade of testes-expressed taste genes
Mazucanti CH, Egan JM. Human type II TAS2R bitter taste receptors. Chem Senses causes male sterility. Proc Natl Acad Sci
taste cells express angiotensin-converting 2010;​35:​157-70. U S A 2013;​110:​12319-24.
enzyme 2 and are infected by severe acute 14. Liang Z, Wilson CE, Teng B, Kinnamon 28. Harrington EO, Vang A, Braza J, Shil
respiratory syndrome coronavirus 2 (SARS- SC, Liman ER. The proton channel OTOP1 A, Chichger H. Activation of the sweet
CoV-2). Am J Pathol 2021;​191:​1511-9. is a sensor for the taste of ammonium taste receptor, T1R3, by the artificial
2. Yao Q, Doyle ME, Liu QR, et al. Long- chloride. Nat Commun 2023;​14:​6194. sweetener sucralose regulates the pulmo-
term dysfunction of taste papillae in 15. Frank HER, Amato K, Trautwein M, et nary endothelium. Am J Physiol Lung Cell
SARS-CoV-2. NEJM Evid 2023;​2(9). al. The evolution of sour taste. Proc Biol Mol Physiol 2018;​314:​L165-L176.
3. Behnood S, Newlands F, O’Mahoney Sci 2022;​289:​20211918. 29. Ward ZJ, Bleich SN, Cradock AL, et al.
L, et al. Persistent symptoms are associ- 16. Chandrashekar J, Yarmolinsky D, von Projected U.S. state-level prevalence of
ated with long term effects of COVID-19 Buchholtz L, et al. The taste of carbon- adult obesity and severe obesity. N Engl
among children and young people: results ation. Science 2009;​326:​443-5. J Med 2019;​381:​2440-50.
from a systematic review and meta-analysis 17. Zocchi D, Wennemuth G, Oka Y. The 30. Heymsfield SB, Wadden TA. Mecha-
of controlled studies. PLoS One 2023;​ cellular mechanism for water detection in nisms, pathophysiology, and management
18(12):​e0293600. the mammalian taste system. Nat Neurosci of obesity. N Engl J Med 2017;​376:​254-66.
4. Liu D, Archer N, Duesing K, Hannan 2017;​20:​927-33. 31. Mennella JA, Bobowski NK. The
G, Keast R. Mechanism of fat taste per- 18. Dutta Banik D, Benfey ED, Martin LE, sweetness and bitterness of childhood: in-
ception: association with diet and obesity. et al. A subset of broadly responsive type sights from basic research on taste prefer-
Prog Lipid Res 2016;​63:​41-9. III taste cells contribute to the detection ences. Physiol Behav 2015;​152:​502-7.
5. Doyle ME, Premathilake HU, Yao Q, of bitter, sweet and umami stimuli. PLoS 32. Thanarajah SE, Backes H, DiFelicean-
Mazucanti CH, Egan JM. Physiology of Genet 2020;​16(8):​e1008925. tonio AG, et al. Food intake recruits oro-
the tongue with emphasis on taste trans- 19. Ren W, Liu Q, Zhang X, Yu Y. Age- sensory and post-ingestive dopaminergic
duction. Physiol Rev 2023;​103:​1193-246. related taste cell generation in circumval- circuits to affect eating desire in humans.
6. Ahmad R, Dalziel JEG. G protein-coupled late papillae organoids via regulation of Cell Metab 2019;​29(3):​695-706.e4.
receptors in taste physiology and pharma- multiple signaling pathways. Exp Cell Res 33. Wiss DA, Avena N, Rada P. Sugar ad-
cology. Front Pharmacol 2020;​11:​587664. 2020;​394:​112150. diction: from evolution to revolution.
7. Zhao GQ, Zhang Y, Hoon MA, et al. 20. Karikkineth AC, Tang EY, Kuo P-L, Front Psychiatry 2018;​9:​545.
The receptors for mammalian sweet and Ferrucci L, Egan JM, Chia CW. Longitudi- 34. Cornelis MC, van Dam RM. Genetic
umami taste. Cell 2003;​115:​255-66. nal trajectories and determinants of hu- determinants of liking and intake of cof-
8. Nakagawa Y, Ohtsu Y, Nagasawa M, man fungiform papillae density. Aging fee and other bitter foods and beverages.
Shibata H, Kojima I. Glucose promotes its (Albany NY) 2021;​13:​24989-5003. Sci Rep 2021;​11:​23845.
own metabolism by acting on the cell- 21. Fischer ME, Cruickshanks KJ, Schubert 35. Gervis JE, Fernández-Carrión R, Chui
surface glucose-sensing receptor T1R3. CR, et al. Factors related to fungiform pa- KKH, et al. Associations between taste per-
Endocr J 2014;​61:​119-31. pillae density: the Beaver Dam Offspring ception profiles and empirically derived
9. Atsumi N, Yasumatsu K, Takashina Y, Study. Chem Senses 2013;​38:​669-77. dietary patterns: an exploratory analysis
et al. Chloride ions evoke taste sensations 22. Peng Y, Gillis-Smith S, Jin H, Trän- among older adults with metabolic syn-
by binding to the extracellular ligand- kner D, Ryba NJP, Zuker CS. Sweet and drome. Nutrients 2021;​14:​142.
binding domain of sweet/umami taste bitter taste in the brain of awake behaving 36. Dutt M, Ng Y-K, Molendijk J, et al.
receptors. Elife 2023;​12:​e84291. animals. Nature 2015;​527:​512-5. Western diet induced remodelling of the
10. Nomura K, Nakanishi M, Ishidate F, 23. Chen X, Gabitto M, Peng Y, Ryba NJP, tongue proteome. Proteomes 2021;​9:​22.
Iwata K, Taruno A. All-electrical Ca2+- Zuker CS. A gustotopic map of taste qual- 37. Choo E, Wong L, Chau P, Bushnell J,
independent signal transduction mediates ities in the mammalian brain. Science Dando R. Offspring of obese mice display
attractive sodium taste in taste buds. Neu- 2011;​333:​1262-6. enhanced intake and sensitivity for palat-
ron 2020;​106(5):​816-829.e6. 24. Roper SD. Encoding taste: from recep- able stimuli, with altered expression of taste
11. Desor JA, Finn J. Effects of amiloride tors to perception. Handb Exp Pharmacol signaling elements. Sci Rep 2020;​10:​12776.
on salt taste in humans. Chem Senses 2022;​275:​53-90. 38. de Araujo IE, Simon SA. The gustatory
1989;​14:​793-803 (https://academic​.­oup​ 25. Calvo SS-C, Egan JM. The endocrinol- cortex and multisensory integration. Int J
.­com/​­chemse/​­article​-­abstract/​­14/​­6/​­793/​ ogy of taste receptors. Nat Rev Endocrinol Obes (Lond) 2009;​33:​Suppl 2:​S34-S43.
­551510). 2015;​11:​213-27. 39. Bartoshuk LM, Duffy VB, Hayes JE,
12. Chandrashekar J, Kuhn C, Oka Y, et 26. Shin Y-K, Martin B, Golden E, et al. Moskowitz HR, Snyder DJ. Psychophysics
al. The cells and peripheral representa- Modulation of taste sensitivity by GLP-1 of sweet and fat perception in obesity:
tion of sodium taste in mice. Nature 2010;​ signaling. J Neurochem 2008;​106:​455-63. problems, solutions and new perspectives.
464:​297-301. 27. Mosinger B, Redding KM, Parker MR, Philos Trans R Soc Lond B Biol Sci 2006;​
13. Meyerhof W, Batram C, Kuhn C, et al. et al. Genetic loss or pharmacological 361:​1137-48.

n engl j med 390;18 nejm.org May 9, 2024 1709


The New England Journal of Medicine
Downloaded from nejm.org on June 17, 2024. For personal use only.
No other uses without permission. Copyright © 2024 Massachusetts Medical Society. All rights reserved.
Physiological Integr ation of Taste and Metabolism

40. Besnard P, Christensen JE, Brignot H, et ages, artificially sweetened beverages, and 69. Tellez LA, Han W, Zhang X, et al.
al. Obese subjects with specific gustatory fruit juice and incidence of type 2 diabetes: Separate circuitries encode the hedonic
papillae microbiota and salivary cues dis- systematic review, meta-analysis, and esti- and nutritional values of sugar. Nat Neu-
play an impairment to sense lipids. Sci Rep mation of population attributable fraction. rosci 2016;​19:​465-70.
2018;​8:​6742. BMJ 2015;​351:​h3576. 70. Nieh EH, Matthews GA, Allsop SA, et
41. Salbe AD, DelParigi A, Pratley RE, 56. Qi Q, Chu AY, Kang JH, et al. Sugar- al. Decoding neural circuits that control
Drewnowski A, Tataranni PA. Taste pref- sweetened beverages and genetic risk of compulsive sucrose seeking. Cell 2015;​160:​
erences and body weight changes in an obesity. N Engl J Med 2012;​367:​1387-96. 528-41.
obesity-prone population. Am J Clin Nutr 57. DiFrancesco L, Fulgoni VL III, Gaine 71. May CE, Rosander J, Gottfried J, Den-
2004;​79:​372-8. PC, Scott MO, Ricciuto L. Trends in added nis E, Dus M. Dietary sugar inhibits satia-
42. Kaufman A, Kim J, Noel C, Dando R. sugars intake and sources among U.S. tion by decreasing the central processing
Taste loss with obesity in mice and men. adults using the National Health and Nu- of sweet taste. Elife 2020;​9:​e54530.
Int J Obes (Lond) 2020;​44:​739-43. trition Examination Survey (NHANES) 72. van Opstal AM, Kaal I, van den Berg-
43. Park DC, Yeo JH, Ryu IY, Kim SH, 2001-2018. Front Nutr 2022;​9:​897952. Huysmans AA, et al. Dietary sugars and
Jung J, Yeo SG. Differences in taste detec- 58. Department of Agriculture, Depart- non-caloric sweeteners elicit different ho-
tion thresholds between normal-weight ment of Health and Human Services. Di- meostatic and hedonic responses in the
and obese young adults. Acta Otolaryngol etary guidelines for Americans, 2020- brain. Nutrition 2019;​60:​80-6.
2015;​135:​478-83. 2025. 9th ed. December 2020 (https:// 73. Dalenberg JR, Patel BP, Denis R, et al.
44. Fischer ME, Cruickshanks KJ, Schubert www​.­dietaryguidelines​.­gov/​­sites/​­default/​ Short-term consumption of sucralose
CR, et al. Taste intensity in the Beaver Dam ­f iles/​­2020​-­12/​­Dietary_Guidelines_for_ with, but not without, carbohydrate im-
Offspring Study. Laryngoscope 2013;​123:​ Americans_2020​-­2025​.­pdf). pairs neural and metabolic sensitivity to
1399-404. 59. Sylvetsky AC, Jin Y, Clark EJ, Welsh JA, sugar in humans. Cell Metab 2020;​31(3):​
45. Kaufman A, Choo E, Koh A, Dando R. Rother KI, Talegawkar SA. Consumption 493-502.e7.
Inflammation arising from obesity re- of low-calorie sweeteners among children 74. Shil A, Olusanya O, Ghufoor Z, For-
duces taste bud abundance and inhibits and adults in the United States. J Acad son B, Marks J, Chichger H. Artificial
renewal. PLoS Biol 2018;​16(3):​e2001959. Nutr Diet 2017;​117(3):​441-448.e2. sweeteners disrupt tight junctions and
46. Archer N, Shaw J, Cochet-Broch M, et 60. Rios-Leyvraz M, Montez J. Health ef- barrier function in the intestinal epithe-
al. Obesity is associated with altered gene fects of the use of non-sugar sweeteners: lium through activation of the sweet taste
expression in human tastebuds. Int J Obes a systematic review and meta-analysis. receptor, T1R3. Nutrients 2020;​12:​1862.
(Lond) 2019;​43:​1475-84. Geneva:​World Health Organization, 2022 75. Suez J, Cohen Y, Valdés-Mas R, et al.
47. Barragán R, Coltell O, Portolés O, et al. (https://iris​.­who​.­int/​­handle/​­10665/​­353064). Personalized microbiome-driven effects of
Bitter, sweet, salty, sour and umami taste 61. Wilk K, Korytek W, Pelczyńska M, non-nutritive sweeteners on human glucose
perception decreases with age: sex-specific Moszak M, Bogdański P. The effect of ar- tolerance. Cell 2022;​185(18):​3307-3328.e19.
analysis, modulation by genetic variants and tificial sweeteners use on sweet taste per- 76. Margolskee RF, Dyer J, Kokrashvili Z,
taste-preference associations in 18 to 80 ception and weight loss efficacy: a review. et al. T1R3 and gustducin in gut sense sug-
year-old subjects. Nutrients 2018;​10:​1539. Nutrients 2022;​14:​1261. ars to regulate expression of Na+-glucose
48. Chia CW, Yeager SM, Egan JM. Endo- 62. Witkowski M, Nemet I, Alamri H, et al. cotransporter 1. Proc Natl Acad Sci U S A
crinology of taste with aging. Endocrinol The artificial sweetener erythritol and cardio- 2007;​104:​15075-80.
Metab Clin North Am 2023;​52:​295-315. vascular event risk. Nat Med 2023;​29:​710-8. 77. Mace OJ, Affleck J, Patel N, Kellett GL.
49. Cattaneo C, Mameli C, D’Auria E, 63. Ebbeling CB, Feldman HA, Steltz SK, Sweet taste receptors in rat small intestine
Zuccotti G, Pagliarini E. The influence of Quinn NL, Robinson LM, Ludwig DS. Ef- stimulate glucose absorption through
common noncommunicable diseases on fects of sugar-sweetened, artificially sweet- apical GLUT2. J Physiol 2007;​582:​379-92.
chemosensory perception and clinical im- ened, and unsweetened beverages on cardio- 78. Shi Q, Zhu X, Deng S. Sweet taste re-
plications in children and adolescents. metabolic risk factors, body composition, ceptor expression and its activation by su-
Adv Nutr 2022;​13:​234-47. and sweet taste preference: a randomized cralose to regulate glucose absorption in
50. Zhang T, Perkins MH, Chang H, Han controlled trial. J Am Heart Assoc 2020;​ mouse duodenum. J Food Sci 2021;​86:​540-5.
W, de Araujo IE. An inter-organ neural 9(15):​e015668. 79. Saltiel MY, Kuhre RE, Christiansen
circuit for appetite suppression. Cell 64. Zani F, Blagih J, Gruber T, et al. The CB, et al. Sweet taste receptor activation
2022;​185(14):​2478-2494.e28. dietary sweetener sucralose is a negative in the gut is of limited importance for
51. Müller TD, Finan B, Bloom SR, et al. modulator of T cell-mediated responses. glucose-stimulated GLP-1 and GIP secre-
Glucagon-like peptide 1 (GLP-1). Mol Metab Nature 2023;​615:​705-11. tion. Nutrients 2017;​9:​418.
2019;​30:​72-130. 65. Chia CW, Shardell M, Tanaka T, et al. 80. Ahmad SY, Friel JK, Mackay DS. Effect
52. Kadouh H, Chedid V, Halawi H, et al. Chronic low-calorie sweetener use and risk of of sucralose and aspartame on glucose
GLP-1 analog modulates appetite, taste abdominal obesity among older adults: a co- metabolism and gut hormones. Nutr Rev
preference, gut hormones, and regional hort study. PLoS One 2016;​11(11):​e0167241. 2020;​78:​725-46.
body fat stores in adults with obesity. 66. Fowler SPG. Low-calorie sweetener 81. Tan H-E, Sisti AC, Jin H, et al. The
J Clin Endocrinol Metab 2020;​105:​1552-63. use and energy balance: Results from ex- gut-brain axis mediates sugar preference.
53. Friedrichsen M, Breitschaft A, Taday- perimental studies in animals, and large- Nature 2020;​580:​511-6.
on S, Wizert A, Skovgaard D. The effect of scale prospective studies in humans. 82. Han W, Tellez LA, Perkins MH, et al.
semaglutide 2.4 mg once weekly on ener- Physiol Behav 2016;​164:​517-23. A neural circuit for gut-induced reward.
gy intake, appetite, control of eating, and 67. Davidson TL, Sample CH, Swithers SE. Cell 2018;​175(3):​665-678.e23.
gastric emptying in adults with obesity. An application of Pavlovian principles to the 83. Buchanan KL, Rupprecht LE, Kael-
Diabetes Obes Metab 2021;​23:​754-62. problems of obesity and cognitive decline. berer MM, et al. The preference for sugar
54. Yang Q, Zhang Z, Gregg EW, Flanders Neurobiol Learn Mem 2014;​108:​172-84. over sweetener depends on a gut sensor
WD, Merritt R, Hu FB. Added sugar intake 68. Breslin PAS, Izumi A, Tharp A, et al. cell. Nat Neurosci 2022;​25:​191-200.
and cardiovascular diseases mortality among Evidence that human oral glucose detec- 84. Liu WW, Bohórquez DV. The neural ba-
US adults. JAMA Intern Med 2014;​174:​516-24. tion involves a sweet taste pathway and a sis of sugar preference. Nat Rev Neurosci
55. Imamura F, O’Connor L, Ye Z, et al. glucose transporter pathway. PLoS One 2022;​23:​584-95.
Consumption of sugar sweetened bever- 2021;​16(10):​e0256989. Copyright © 2024 Massachusetts Medical Society.

1710 n engl j med 390;18 nejm.org May 9, 2024

The New England Journal of Medicine


Downloaded from nejm.org on June 17, 2024. For personal use only.
No other uses without permission. Copyright © 2024 Massachusetts Medical Society. All rights reserved.

You might also like