ArticlePDF AvailableLiterature Review

Effects of Non-nutritive Sweeteners on Sweet Taste Processing and Neuroendocrine Regulation of Eating Behavior

Authors:
  • Harvard T.H. Chan School of Public Health

Abstract and Figures

Purpose of review: Non-nutritive sweeteners (NNS) are increasingly used as a replacement for nutritive sugars as means to quench the desire for "sweets" while contributing few or no dietary calories. However, there is concern that NNS may uncouple the evolved relationship between sweet taste and post-ingestive neuroendocrine signaling. In this review, we examine the effects of NNS exposure on neural and peripheral systems in humans. Recent findings: NNS exposure during early development may influence sweet taste preferences, and NNS consumption might increase motivation for sweet foods. Neuroimaging studies provide evidence that NNS elicit differential neuronal responsivity in areas related to reward and satiation, compared with caloric sweeteners. Findings are heterogenous regarding whether NNS affect physiological responses. Additional studies are warranted regarding the consequences of NNS on metabolic outcomes and neuroendocrine pathways. Given the widespread popularity of NNS, future studies are essential to establish their role in long-term health.
Content may be subject to copyright.
NUTRITION AND THE BRAIN (J NASSER, SECTION EDITOR)
Effects of Non-nutritive Sweeteners on Sweet Taste Processing
and Neuroendocrine Regulation of Eating Behavior
Alexandra G. Yunker
1,2
&Reshma Patel
1,2
&Kathleen A. Page
1,2
#Springer Science+Business Media, LLC, part of Springer Nature 2020
Abstract
Purpose of Review Non-nutritive sweeteners (NNS) are increasingly used as a replacement for nutritive sugars as means to
quench the desire for sweetswhile contributing few or no dietary calories. However, there is concern that NNS may uncouple
the evolved relationship between sweet taste and post-ingestive neuroendocrine signaling. In this review, we examine the effects
of NNS exposure on neural and peripheral systems in humans.
Recent Findings NNS exposure during early development may influence sweet taste preferences, and NNS consumption might
increase motivation for sweet foods. Neuroimaging studies provide evidence that NNS elicit differential neuronal responsivity in
areas related to reward and satiation, compared with caloric sweeteners. Findings are heterogenous regarding whether NNS affect
physiological responses.
Summary Additional studies are warranted regarding the consequences of NNS on metabolic outcomes and neuroendocrine
pathways. Given the widespread popularity of NNS, future studies are essential to establish their role in long-term health.
Keywords Non-nutritive sweeteners .Low-calorie sweeteners .Sweeteners .Artificial sweeteners .Sweet taste .Hormones .
Obesity .Insulin .Incretins .GLP-1 .Brain .fMRI .Neuroimaging .Obesity .Reward .Satiety .Food intake .Feeding
behavior .Appetite .Hypothalamus .Striatum .Insula .Amygdala
Introduction
A growing body of evidence has linked increased caloric sug-
ar consumption with obesity risk [13]. Accordingly, non-
nutritive sweeteners (NNS) have become a popular alternative
for added sugar intake, satisfying the craving for sweets
while providing few or no calories. NNS use is rapidly in-
creasing; currently, over 40% of US adults and 25% of ado-
lescents and children are habitual NNS consumers [4].
Notably, among American children and adolescents, NNS in-
take has increased by 200% since 1999 [4]. Furthermore,
given the widespread distribution of NNS in drinks and foods,
consumers are often unaware that they are even ingesting
NNS [5,6]. Despite the increasing usage of NNS, and NNS
being largely marketed as strategic tools for weight manage-
ment, the prevalence of obesity and associated metabolic dis-
orders has not decreased; rather, rates of the co-epidemics of
obesity and type 2 diabetes (T2DM) have continued to rise
over the past several decades [7,8]. Notably, a recent advisory
from the American Heart Association recommended against
prolonged consumption of NNS beverages by children, while
also concluding that NNS beverages could potentially be a
useful replacement strategy for adult chronic high consumers
of sugar-sweetened beverages [9]. In addition, while epidemi-
ological evidence suggests that NNS exposure throughout the
lifespan (and as early as in utero) can contribute to risk for
weight gain [1013] and risk for metabolic derangements,
including type 2 diabetes [14], other studies using experimen-
tal designs have reported that NNS have neutral or beneficial
effects regarding body weight [1518] and glucose metabo-
lism [19]. Given the equivocal evidence regarding the efficacy
of NNS, and the paradoxical increase in prevalence of both
NNS use and metabolic disorders, it is imperative that the
This article is part of the Topical Collection on Nutrition and the Brain
*Kathleen A. Page
kpage@usc.edu
1
Division of Endocrinology, Department of Medicine, Keck School of
Medicine, Diabetes and Obesity Research Institute, University of
Southern California, 2250 Alcazar Street; CSC 209, Los
Angeles, CA 90089, USA
2
Diabetes and Obesity Research Institute, Keck School of Medicine,
University of Southern California, Los Angeles, CA 90089, USA
Current Nutrition Reports
https://doi.org/10.1007/s13668-020-00323-3
potential neural and peripheral implications of NNS consump-
tion throughout the lifespan are understood (Fig. 1). The pur-
pose of this article is to review and summarize the current
literature, and to address the gaps in knowledge regarding
the effects of both acute and chronic NNS exposure across
the lifespan on glucose metabolism, sweet taste perception
and preference, and neural systems involved in appetite and
reward, with an emphasis on findings from human studies.
NNS and Sweet Taste
While caloric sugars and NNS have varying chemical struc-
tures, they all interact with the heterodimeric sweet taste re-
ceptor complex, T1R2/T1R3 [20]. Notably, sweet taste per-
ception plays a role in carbohydrate metabolism and reward
[21], and sweet taste preference has been linked to the likeli-
hood of children becoming overweight or obese [22].
Additionally, sweet-liking is predictive of weight gain over
time in some adult populations [23,24]. Given the potential
metabolic consequences of heightened hedonic liking for
sweet, it is important that the possible influences of NNS on
taste perception and preference are well understood.
A growing body ofevidence in rodents,and a limited num-
ber of studies in humans, suggests that NNS exposure or con-
sumption during early development may influence sweet taste
processing. The formation of taste preferences, including pref-
erence for sweet, begins before birth; both rodent and human
studies havedemonstrated that maternal diets during pregnan-
cy and lactation, in addition to the offspringsdietduringthe
first months of life, influence flavor learning, conditioning,
and acceptance [2527]. The effects of in utero and early-
life exposure to NNS on offspring sweet taste learning, per-
ception, and preference have in large part been elucidated by
rodent studies. Rosales-Gomez et al. recently reported that
young mice who habitually consumed oral sucralose directly
after weaning had heightened preference for sweetened water
and increased weight gain at approximately 15 weeks of life
[28]. Other rodent studies have addressed how in utero and
lactational NNS exposure influences offspring taste prefer-
ence. Mouse pups exposed to acesulfame potassium (aceK)
prenatally and during lactation via maternal diet exhibit great-
er sweet taste preference in adulthood compared with control
Fig. 1 Downstream potential
effects of NNS intake on
neuroendocrine systems involved
in appetite regulation and eating
behavior. The taste of a sweet
substance activates sweet taste
receptors, including in the oral
cavity and brain. Exposure to
NNS, which uncouple sweet taste
and energy content, may
dysregulate sweet taste signaling
pathways and affect brain and
metabolic processes across the
lifespan, which, in turn, may
contribute to altered eating
behavior, T2DM, and chronic
disease. Figure created with
BioRender
Curr Nutr Rep
mice [29,30], and furthermore, infusion of aceK during the
early postnatal stage promoted unfavorable gustatory system
changes in young mice [31].
Together, these findings in animal models have particular
clinical implications for pediatric populations. NNS are fre-
quently ingested by nursing infants; it has been shown that
saccharin, sucralose, and aceK were present in 65% of breast
milk samples from twenty lactating mothers, independent of
the mothersNNS dietary intake [32]. However, the specific
magnitude and prevalence of fetal NNS exposure in humans
remains unknown [33]. Studies in children examining the
effects of habitual NNS consumption on dietary preference
for sweet foods are limited to cross-sectional analyses. A
study among UK children and adolescents (ages 418) found
that boys who reported any dietary consumption of artificially
sweetened beverage(s) (ASBs) had higher dietary intake of
sugar from solid foods when compared with boys who report-
ed consuming only sugar-sweetened beverages (SSBs) or
those who were non-consumers of either SSBs or ASBs
[34]. However, the majority of this cohort consumed both
SSBs and ASBs (43%), while a subset of 18% consumed only
ASBs [34]. More recently, Sylvetsky et al. reported the first
results from a US study that used 20112016 National Health
and Nutrition Examination Survey (NHANES) data to exam-
ine associations between NNS beverage consumption and di-
etary intake in children and adolescents. They found that con-
sumers of low-calorie sweetened beverages, whether con-
sumed alone or in conjunction with SSBs, displayed higher
energy, carbohydrate, total sugar, and added sugar intake
compared with children and adolescents who were classified
as only water consumers [35••]. In a separate study that used
NHANES data, Sylvetsky and colleagues also demonstrated
positive associations between dietary NNS consumption and
obesity in adolescents [36]. It should be noted that the
NHANES data were limited to self-reported dietary intake,
based on only a single or two-day recall, and that given the
cross-sectional nature of the studies, confounding by reverse
causation is possible. Nevertheless, the findings from
Sylvetsky et al. are consistent with one of the proposed mech-
anisms by which early-life NNS exposure may impact future
body composition via dysregulation of the developmental pro-
gramming of taste preferences. Chronic NNS consumption
may uncouple the functionality of sweet taste to signal the
post-ingestive caloric consequences of eating sweet foods, in
turn, enhancing sugar intake [3741]. Taken together, further
studies examining the effects of early-life NNS exposure on
taste preference, dietary intake, and body weight regulation
are warranted. Future areas of study include replication of
findings in animal models and studies that include more rig-
orous experimental methods in humans.
Few human studies have addressed how NNS consumption
influences sweet taste preferences in adulthood. A study by
Casperson etal. aimed to determine the effects of consuming a
SSB or NNS beverage on the reinforcing value of sweet foods.
Young adults ingested either acute oral sucralose (Splenda®)
or sucrose, matched for sweetness and pleasantness, with a
standardized meal. Consumption of sucralose, but not sucrose,
heightened the motivation to gain access to sweet foods post
meal [42]. Sucralose increased the relative reinforcing values
of sweet snacks, compared with salty or savory snacks, sug-
gesting that acute NNS consumption might alter desire for
sweet foods and eating behavior [42]. In contrast, a recent
industry-funded study reported that among French adults, wa-
ter and low-calorie sweetened (LCS) beverage ingestion did
not have differential effects on the selection of, or motivation-
al ratings towards, sweet foods [43]. Furthermore, appetite for
sweet foods was neither affected by acute nor longer-term
exposure to the LCS beverage [43]. It is important to note that
the respective experimental studies by Casperson et al. and
Fantino et al. utilized different NNS methodologies, the latter
employing a LCS lemonade that included several NNS (aceK,
aspartame, and sucralose) in combination with other com-
pounds. Hill and colleagues examined the acute effects of
consuming a SSB (Sprite®), a NNS beverage (Sprite
Zero®), or an unsweetened beverage (carbonated water), in
combination with a standardized meal, on subsequent product
choice and subjective responses to a sugar-sweetened food
among young adults. They found that participants who con-
sumed the NNS drink, relativeto those who had consumed the
SSB or water, were more likely to choose a high calorie food
item (specifically, candy) during a food product choice task,
compared to other food options [44]. In addition, participants
who consumed the NNS beverage felt less satisfied after eat-
ing a sugar-sweetened snack (cookies), compared with sub-
jects who consumed the SSB or water [44]. Taken together,
these studies provide equivocal evidence for the impact of
NNS consumption on adult sweet taste preference and eating
behavior, and future experimental studies are warranted.
NNS and Metabolic Hormones
There has been much debate regarding whether NNS have
effects on metabolic hormones. In vitro studies showed that
NNS bind with high affinity to the T1R3 subunit of the sweet
taste receptor complex, which is expressed on the tongue and
throughoutthe digestive tract [4547], and that NNS stimulate
incretin and insulin release in both the enteroendocrine cells in
the gut as well as beta cells in the pancreas [20,4852]. While
evidence from in vitro studies has been compelling, in vivo
studies testing the effects of NNS on metabolic hormone se-
cretion have produced mixed results. Studies in rodent models
have been reviewed elsewhere [53], and in this review, we
highlight human studies that have examined the effects of
NNS on metabolic hormones (Table 1).
Curr Nutr Rep
Table 1 Summary of findings from human studies examining NNS effect on hormone levels, specifically insulin, glucagon like peptide 1 (GLP-1), peptide YY (PYY), gastric inhibitory polypeptide
(GIP), leptin, ghrelin, and glucagon
Author Year Age Group/
Size
Participant
Characteristics
NNS Used Dosage and Method
of Delivery
Insulin C-
peptide
GLP-1 PYY GIP Leptin Ghrelin Glucagon
Abdallah et al. 1997 12 Male
Adults
Lean Aspartame 18mg aspartame,
tasted
Decreased n/a n/a n/a n/a n/a n/a No effect
Ahmad et al. 2019 17 Adults
(10F)
Lean Aspartame,
Sucralose
425mg aspartame,
136mg sucralose,
ingested for 2 weeks
No effect n/a No effect n/a n/a No effect n/a n/a
Anton et al. 2010 31 Adults 19 Lean, 12 Obese Stevia,
Asparta-
me,
Sucrose
400g preload meal
with stevia,
aspartame or
sucrose
Lower with stevia
and aspartame
than sucrose
n/a n/a n/a n/a n/a n/a n/a
Argyri et al. 2013 70 Adults
(28F)
T2DM Diabetic
dessert
Not specified, ingested No effect No effect n/a n/a n/a n/a n/a n/a
Bonnet et al. 2018 50 Adults
(28F)
28 Lean, 22
Overweight
Aspartame+
aceK
129mg aspartame +
13mg aceK for 12
weeks, 4 week
washout, then 258
mg aspartame +
26mg aceK for 12
weeks
No effect n/a n/a n/a n/a n/a n/a n/a
Brown, R et al. 2009 22
Adolesce-
nts and
Young
Adults
(12F)
Overweight Sucralose+
aceK
68mg sucralose +
41mg aceK (Diet
Rite Cola), then
OGTT
No effect n/a Increased n/a n/a n/a n/a n/a
Brown, R et al. 2012 44
Adolesce-
nts and
Young
Adults
(27F)
25 Overweight
(Nondiabetic),
9 T1DM lean,
10 T2DM
obese
Sucralose+
aceK
68mg sucralose +
41mg aceK (Diet
Rite Cola), then
OGTT
n/a No effect Increased in
T1DM and
nondiabetic
overweight
groups, not
T2DM group
No effect n/a n/a n/a n/a
Brown, A et al. 2011 8 Female
Adults
Lean Sucralose 42mg sucralose drink,
42mg sucralose +
50g sucrose drink,
then standard meal
No effect n/a n/a n/a n/a n/a No effect No effect
Dhillon et al. 2017 64 Adults
(41F)
Obese Sucralose Not specified, in solid
and liquid form;
taste vs. ingestion
Early rise higher
with taste of
solid
n/a n/a n/a n/a n/a n/a n/a
Ford et al. 2011 8 Adults
(7F)
Lean Sucralose 2mmol/L in water, or
2mmol/L in water
with maltodextrin,
tasted and ingested
No effect n/a No effect No effect n/a n/a n/a n/a
Grotz et al. 2003 128 Adults
(42F)
Obese Sucralose 667mg sucralose, via
ingested capsule, 13
week exposure
n/a No effect n/a n/a n/a n/a n/a n/a
Grotz et al. 2017 Lean Sucralose No effect No effect n/a n/a n/a n/a n/a n/a
Curr Nutr Rep
Table 1 (continued)
Author Year Age Group/
Size
Participant
Characteristics
NNS Used Dosage and Method
of Delivery
Insulin C-
peptide
GLP-1 PYY GIP Leptin Ghrelin Glucagon
47 Male
Adults
667mg sucralose, via
ingested capsule, 12
week exposure
Higgins and
Mattes
2019 154 Adults
(87F)
85 Overweight, 69
Obese
Saccharin,
Asparta-
me,
Stevia,
Sucralose
73mg saccharin, 58mg
aspartame, 66mg
stevia, 16mg
sucralose, via drink,
12 week exposure
No effect n/a n/a n/a n/a n/a n/a n/a
Higgins et al. 2018 100 Adults
(50F)
Lean Aspartame 0, 350, 1050mg
aspartame, via
drink, 12 week
exposure
No effect n/a No effect n/a No effect No effect n/a n/a
Lertrit et al. 2018 15 Adults
(11F)
8 Lean, 1
Overweight, 6
Obese
Sucralose 200mg sucralose, via
capsule, 4 week
exposure
Decreased n/a Increased n/a n/a n/a n/a n/a
Ma et al. 2009 7 Adults Lean Sucralose 80mg sucralose,
800mg sucralose,
via NG tube
No effect n/a No effect n/a No effect n/a n/a n/a
Ma et al. 2010 10 Adults
(2F)
Lean Sucralose 960mg sucralose, via
NG tube
n/a n/a No effect n/a n/a n/a n/a n/a
Nichol et al. 2019 21 Adults
(17F)
10 Lean, 11 Obese Sucralose 48mg sucralose, t aste
vs. ingestion then
OGTT
Increased in taste
vs. ingestion
for both weight
groups, late
increase in
obese group
during OGTT
No effect n/a n/a No effect n/a n/a n/a
Overduin et al. 2016 20 Adults
(10F)
10 Lean, 10 Obese Erythritol+
Sucralose
8g erythritol, 4mg
sucralose in drink
and meal, compared
to sucrose control
No effect n/a Increased
compared to
sucrose
Increased
compared to
sucrose
n/an/an/an/a
Pepino et al. 2013 17 Adults
(15F)
Obese Sucralose 48mg sucralose,
ingested, followed
by OGTT
Increased Increased No effect n/a No effect n/a n/a No effect
Romo-Romo et al. 2018 66 Adults
(49F)
Lean Sucralose 12mg sucralose
consumed 3x/day,
via drink, 14 day
exposure
Decreased insulin
sensitivity,
increased acute
insulin
response
n/a n/a n/a n/a n/a n/a n/a
Sakurai et al. 2012 21 Male
Adults
Lean Sucralose,
aceK,
Asparta-
me,
Erythritol
Not specified, mixed
with 5mg sucrose
via drink,
co-ingested with a
meal
n/a n/a No effect n/a n/a n/a n/a n/a
Curr Nutr Rep
Table 1 (continued)
Author Year Age Group/
Size
Participant
Characteristics
NNS Used Dosage and Method
of Delivery
Insulin C-
peptide
GLP-1 PYY GIP Leptin Ghrelin Glucagon
Steinert et al. 2011 12 Adults
(6F)
Lean Sucralose,
Asparta-
me, aceK
169mg aspartame,
220mg aceK, 62mg
sucralose, via NG
tube
No effect n/a No effect No effect n/a n/a No effect n/a
Sylvetsky et al. 2016 61 Adults
(34F)
Overweight Sucralose,
Sucralos-
e+aceK
Experiment 1: 68mg,
170mg and 250mg
sucralose;
Experiment 2: Diet
Rite Cola: 68mg
sucralose + 41mg
aceK; Diet
Mountain Dew:
18mg sucralose,
18mg aceK, 57mg
aspartame; 68mg
sucralose + 41mg
aceK dissolved in
seltzer water, all
preloads to OGTT
No effect No effect Increased after
sucralose +
aceK, Diet Rite
Cola, and Diet
Mountain Dew;
no effect of
sucralose alone
n/a No effect n/a n/a n/a
Temizkhan et al. 2015 16 Adults
(8F)
8Obese,8Obese
T2DM
Aspartame,
Sucralose
72mg aspartame,
24mg sucralose,
ingested, then
OGTT
No effect No effect Increased after
sucralose
n/a n/a n/a n/a n/a
Tey et al. 2017 30 Male
Adults
Lean Aspartame,
Monk
fruit,
Stevia
440mg aspartame,
630mgmonkfruit,
330mg stevia, via
drink, prior to test
meal, sucrose as a
positive control
Increased acutely
after test meal,
no difference in
AUC
n/a n/a n/a n/a n/a n/a n/a
Wu et al. 2012 10 Adults
(3F)
Overweight Sucralose 60mg sucralose drink,
followed by test
meal
No effect n/a No effect n/a No effect n/a n/a n/a
Wu et al. 2013 10 Male
Adults
Overweight Sucralose,
aceK,
Sucralos-
e+aceK
52mg sucralose,
200mg aceK, 46mg
sucralose + 26mg
aceK, then OGTT
No effect n/a No effect n/a n/a n/a n/a n/a
Curr Nutr Rep
The effects of acute ingestion of NNS on incretin and in-
sulin responses have been studied using a variety of delivery
methods as well as different dosages and types of NNS
[5466]. Interestingly, Diet Rite Cola®, which contains sucra-
lose and aceK, among other colorants and preservatives, was
found to increase GLP-1 secretion when compared with a
water or a seltzer control when consumed prior to an oral
glucose tolerance test (OGTT) in overweight and obese indi-
viduals [5456]. However, studies examining the effects of
NNS dissolved solely in water have been mixed, with the
majority showing no effect of acute NNS consumption on
hormone secretion [56,57,59,64]. While Temizkhan et al.
observed an increase in GLP-1 levels when sucralose vs. wa-
ter was consumed prior to glucose ingestion, these effects
were not observed with an aspartame preload [57].
Sylvetsky and colleagues found no significant difference be-
tween varying concentrations of sucralose dissolved in water
compared with water alone when consumed prior to an oral
glucose load on peripheral insulin, glucose, C-peptide, or
GLP-1 levels in overweight individuals [56]. Likewise,
Ford and colleagues found no difference between sucralose
compared with water preloads on insulin or GLP-1 responses
to oral glucose [64]. Additionally, Wu and colleagues found
no effect of sucralose or aceK, when consumed alone or in
combination, on peripheral insulin or GLP-1 concentrations
before or during an OGTT [59]. Future work is needed to
determine if some of the observed NNS effects on peripheral
GLP-1 are attributable to the colorants or preservative present
in diet soda given that the majority of studies that found an
effect on GLP-1 secretion utilized Diet Rite Cola®.
The studies mentioned above utilized acute glucose inges-
tion as a caloric load, whereas other studies used a standard-
ized meal to examine the effects of NNS consumption on
hormone responses in a more real-lifescenario. These stud-
ies have largely found no effects of acute NNS ingestion on
hormone responses to standardized meals [60,65,67,68]. Wu
and colleagues examined the effects of sucralose on the hor-
monal response to a mashed potato meal in overweight indi-
viduals and found no significant effect of a sucralose preload
on insulin, GLP-1, or GIP [60]. Likewise, NNS co-ingested
with a meal of chicken soup and biscuits had no effect on
GLP-1 levels in lean males [67]. Similarly, Brown and col-
leagues found no effect of a sucralose preload on insulin,
ghrelin, and glucagon levels in response to a standardized
breakfast in lean females [68]. More recently, Tey and col-
leagues showed that the consumption of drinks containing the
NNS, aspartame, stevia, or monk fruit extract when compared
with drinks containing sucrose resulted in higher insulin levels
at 120 min after lunch, but reported no difference between the
four drinks on insulin or glucose area under the curve (AUC)
over the 3-h period after lunch [65].
The longer-term effects of NNS consumption on metabolic
regulation in humans have also been examined with no clear
consensus on their physiological effects. Two recent studies
suggested that sucralose ingestion may negatively affect insu-
lin sensitivity [69,70]. Lertrit and colleagues showed that 4-
week consumption of sucralose (in capsule form) vs. an empty
capsule increased peripheral GLP-1 levels and decreased in-
sulin sensitivity in lean, overweight, and obese adults [69].
This finding was replicated by another study showing that a
14-day ingestion of sucralose in beverage form led to de-
creased insulin sensitivity in lean adults [70]. In contrast, other
studies have shown no effect of longer-term NNS consump-
tion on metabolic hormones [18,7173]. Ahmad and col-
leagues found no change in insulin, GLP-1, or leptin levels
and no change in insulin sensitivity in lean adults exposed for
12 weeks to aspartame or sucralose mixed in water [71].
Higgins and Mattes tested 12-week exposure to aspartame,
sucralose, saccharin, and stevia in overweight and obese
adults and showed no change in insulin levels with any of
the NNS examined [18]. Similarly, another study showed
no effects of aspartame and aceK administered in differing
dosages over 12 weeks on insulin levelsin response to glucose
ingestion [72]. Furthermore, Grotz et al. found no difference
in glucose, C-peptide, or hemoglobin A1c after ingestion of
sucralose in a capsule vs. cellulose placebo over 1213 weeks
in obese and lean adults [74,75].
Collectively, the current evidence provides equivocal evi-
dence on the effects of NNS consumption on hormones in-
volved in appetite regulation and glucose homeostasis. More
work is necessary to determine the specific concentrations and
types of NNS that may elicit hormone secretion, whether ef-
fects of NNS are dependent on delivery method, and whether
consumption of NNS in isolation or in the presence of carbo-
hydrate produces different effects. Future studies should con-
sider how individual characteristics, including habitual NNS
consumption, age, sex, adiposity, and insulin resistance, affect
metabolic hormone responses to NNS consumption.
Of note, while the majority of work has been done in
adults, recent evidence suggests that NNS may affect fetal
development and potential programming later in life.
Exposure to NNS in utero and during early life was associated
with risk of metabolic syndrome later in life in mice [76,77].
A longitudinal study in children demonstrated that mothers
with gestational diabetes who consumed daily NNS compared
with NNS non-consumers had children who were larger for
gestational age at birth as well as a higher BMI z-score and
increased risk of obesity at 7 years [13]. These results were
corroborated by a longitudinal cohort study showing that daily
consumption of NNS was linked with a 0.2 unit increase in
infant BMI z-score as well as a greater risk for being over-
weight at 1 year of age [11], suggesting that maternal pro-
gramming with NNS exposure may affect a childsmetabolic
development. These studies further underscore the need for
studies on the effects of NNS in early development and
childhood.
Curr Nutr Rep
NNS and Neural Systems Involved in Appetite
and Reward
There has been increasing interest towards elucidating the
effects of NNS on brain regulation of appetite and reward. A
growing body of evidence reported via fMRI studies suggests
that NNS can provoke differential brain responses in humans,
compared with nutritive sweeteners. Frank et al. reported that
taste pathways in the brain can distinguish nutritive versus
non-nutritive sweet taste; sucrose, relative to sucralose, elicit-
ed stronger blood oxygen leveldependent (BOLD) brain re-
sponse activation of regions involved in reward processing,
such as the main gustatory complex (frontal operculum/
anterior insula) and the contralateral insula and midbrain, in-
cluding the ventral tegmental area (VTA),and substantia nigra
[78]. In accordance with these findings, Smeets et al. demon-
strated that small tastes of sucrose provoked increased BOLD
activation in the striatum, while in contrast, small tastes of a
mix of several NNS (aspartame, aceK, sodium cyclamate, and
sodium saccharin) led to heightened activation in the amyg-
dala, among lean adult males [79]. Taken together, these find-
ings support that either small or large tastes of NNS, when
compared with caloric sugars, can evoke differential re-
sponses within neural areas involved in processing of reward
and primary regions of taste activation.
Notably, several fMRI studies indicate that NNS may have
dampened hypothalamic satiety signaling effects, compared
with nutritive sugars. The hypothalamus is a brain region that
regulates appetite and energy homeostasis. Prior fMRI studies
have consistently shown a reduction in hypothalamic activa-
tion following the ingestion of glucose, which is interpreted as
a biomarker of satiety [8082], and obesity is associated with
an altered glucose-linked hypothalamic response [83,84]; fur-
thermore, alterations in glucose-linked hypothalamic activa-
tion predicted longitudinal weight gain in children [85].
Smeets and colleagues first established that decreases in hy-
pothalamic activation in response to sweetened beverages
might be dependent on both sweet taste and energy content.
They found that among young adults, glucose ingestion pro-
voked a signal reduction in the hypothalamus, while water,
maltodextrin, and aspartame had no effect on hypothalamic
activation [86]. Most recently, Van Opstal et al. investigated
the hypothalamic response to acute ingestion of sucralose,
relative to nutritive sugar; sucralose led to the smallest de-
crease in BOLD activity in the hypothalamus, similar to water,
when compared with glucose, fructose, and sucrose ingestion
[87]. Another recent fMRI study also demonstrated that con-
sumption of a fat/protein milkshake sweetened with glucose
resulted in a widespread effect on the brain: decreased BOLD
signal in the posterior cingulate cortex, brainstem, VTA, and
insula and also decreased voxel based connectivity in the hy-
pothalamus and VTA [88••]. In contrast, shakes containing
allulose and sucralose showed no effect on BOLD signaling
within any of the regions of interest indicating that the NNS
had no immediate effect on the activation of brain areas related
to eating behavior [88••]. This finding further supports that
sweet taste, in the absence of nutritive carbohydrates, may
not lead to hypothalamic connectivity changes that are typi-
cally linked to satiation. It is important to note that there is an
abundant expression of sweet taste receptors within the hypo-
thalamus; RNA expression levels of the sweet taste receptor
complex (T1R2/T1R3) in the hypothalamus are significantly
higher than those in other brain regions implicated in eating
behavior, such as the cortex or hippocampus [89]. Given that
NNS interact with the sweet taste receptor complex, future
areas of investigation could consider how sweet taste prefer-
ence impacts neural satiety signaling in response to sugars and
NNS.
Other recent findings from Creze and colleagues utilize
electroencephalographic (EEG) methods to assess whether in-
gestion of sucrose and NNS drinks would elicit different neu-
ral responses to food cues and subsequent food intake at an ad
libitum buffet. The acute ingestion of a NNS beverage (con-
taining a mix of cyclamate, aceK, and aspartame) produced
differential neural activity in response to food cues, compared
with sucrose and water ingestion. Sucrose or water, but not
NNS, led to increased insula activation, whereas NNS con-
sumption increased neural activity in ventrolateral prefrontal
regions associated with inhibition of reward, consistent with
prior findings in humans [79,90]. The investigators concluded
that their findings showing differential brain responses to
acute NNS consumption may be indicative of early-stage ad-
aptation to taste-calorie uncoupling [90••]. However, there
was no difference in food intake during the buffet between
the water and NNS conditions, which the investigators pro-
posed could be due to limitations in experimental design and
that the design may not have been sensitive enough to capture
all secondary outcome differences between the NNS and wa-
ter groups [90••]. Another recent EEG study by Creze et al.
featured an interventional design; daily consumers of SSB
were asked to undergo a 3-month replacement period with
NNS beverage equivalents, which contained a varying mix
of NNS, such as aspartame, cyclamate, aceK, and sucralose.
Participants neither experienced weight loss over the replace-
ment period nor changes in food liking towards visual cues;
however, neural activity in response to high-fat, sweet food
cues was decreased in prefrontal regions linked to impulse
control after the intervention period [91]. Interestingly, the
post-intervention neural modulations in prefrontal areas were
predictive of weight loss failure, implying individual dimin-
ished ability over food intake control [91].
Additional studies in humans utilizing fMRI methods sug-
gest that frequent dietary consumption of NNS may condition
altered neural processing of sweet taste. Rudenga and Small
showed a negative association between self-reported chronic
NNS use and amygdala response, with a similar trend in the
Curr Nutr Rep
insula, to acute in-scanner tastes of varying concentrations of
sucrose among lean and overweight adults [92]. Given that the
amygdala and insula are key regions involved in integrating
flavor nutrient signals, these findings are consistent with those
of rodent literature suggesting that chronic NNS use may un-
couple the association between sweet taste and post-ingestive
consequences of predicted calories. In addition, Green and
colleagues showed that among individuals who were non-
habitual diet soda drinkers, patterns of activation in the
orbitofrontal cortex (OFC), a region implicated in processing
of reward, differed in response to acute tastes of saccharin
compared with sucrose; in contrast, among the habitual con-
sumers of diet sodas, neural activation patterns did not differ
between either the sucrose or saccharin condition [93].
Furthermore, habitual diet soda drinkers exhibited greater ac-
tivation in the OFC, lentiform nucleus, dopaminergic mid-
brain, and right amygdala in response to both sucrose and
saccharin, compared with non-diet soda drinkers [93].
Together, these findings support that chronic NNS consump-
tion may compromise the efficacy of brain regions related to
appetite and reward to process sweet taste.
Neuroimaging studies that have assessed brain responses to
NNS have largely been focused on lean and healthy cohorts
[78,79,86,87,9094]. Studies that examine potential obesity
related differences in neural responses to NNS are warranted.
In addition, many of the neuroimaging studies that examine
brain responses to NNS have been limited to same-sex cohorts
[78,79,86,87,90,91,94]. Given that sex differences regard-
ing sweet taste perception have been previously reported in
rodents [95], future investigators should include both males
and females. Finally, to the best of our knowledge, there have
been no studies to date that examine the effects of NNS on
brain regulation of appetite and reward in children.
Conclusion
While NNS seem to elicit differential brain responses in ap-
petite and reward regions, compared with caloric sweeteners,
findings are equivocal as to whether these divergent brain
responses are predictive of subsequent metabolic conse-
quences. Gaps in the knowledge include how NNS affect both
glucose metabolism and the neural regulation of eating behav-
ior in particularly vulnerable populations such as pregnant and
lactating women, children, obese individuals, and persons
with metabolic disease. A key goal of future research should
investigate how both chronic and acute intake of NNS influ-
ence the neural and peripheral responses of these populations.
In addition, exposure to NNS during development and
throughout the lifespan may also influence sweet taste prefer-
ences; given that there is an abundance of sweet taste receptors
in the brain, it would be of interest to examine how individual
variation in sweet taste preference affects the neural
processing of NNS. Considering both the increasing preva-
lence of dietary NNS intake and the rising rates of obesity
and chronic disease, additional studies in humans are critical
to determine how NNS consumption impacts neuroendocrine
systems across the lifespan.
Funding Information This work was supported by the National Institutes
of Health (NIH) National Institute of Diabetes and Digestive and Kidney
Diseases R01DK102794 (PI: K.A.P).
Compliance with Ethical Standards
Conflict of Interest The authors have nothing to disclose.
Human and Animal Rights and Informed Consent All cited studies by
the authors were approved by the institutional review boards of their
respective institutions.
References
Papers of particular interest, published recently, have been
highlighted as:
Of importance
•• Of major importance
1. Hu FB, Malik VS. Sugar-sweetened beverages and risk of obesity
and type 2 diabetes: epidemiologic evidence. Physiol Behav.
2010;100:4754.
2. Malik VS, Willett WC, Hu FB. Global obesity: trends, risk factors
and policy implications. Nat Rev Endocrinol. 2013;9:1327.
3. Bray GA, Popkin BM. Dietary sugar and body weight: have we
reached a crisis in the epidemic of obesity and diabetes?: health be
damned! Pour on the sugar. Diabetes Care. 2014;37:9506.
4. Sylvetsky AC, Jin Y, Clark EJ, Welsh JA, Rother KI, Talegawkar
SA. Consumption of low-calorie sweeteners among children and
adults in the United States. J Acad Nutr Diet. 2017;117:441
448.e2.
5. Sylvetsky AC, Greenberg M, Zhao X, Rother KI. What parents
think about giving nonnutritive sweeteners to their children: a pilot
study. Int J Pediatr. 2014;2014:15. https://doi.org/10.1155/2014/
819872.
6. Sylvetsky AC, Walter PJ, Garraffo HM, Robien K, Rother KI.
Widespread sucralose exposure in a randomized clinical trial in
healthy young adults. Am J Clin Nutr. 2017;105:8203.
7. Hales CM, Carroll MD, Fryar CD, Ogden CL. Prevalence of obe-
sity among adults and youth : United States, 20152016. 2017.
8. Xu G, Liu B, Sun Y, Du Y, Snetselaar LG, Hu FB, et al. Prevalence
of diagnosed type 1 and type 2 diabetes among US adults in 2016
and 2017: population based study. BMJ. 2018;362:k1497.
9. Johnson RK, Lichtenstein AH, Anderson CAM, Carson JA,
Després J-P, Hu FB, et al. Low-calorie sweetened beverages and
cardiometabolic health: a science advisory from the American
Heart Association. Circulation. 2018;138:e12640. https://doi.org/
10.1161/CIR.0000000000000569.
10. Pepino MY. Metabolic effects of non-nutritive sweeteners. Physiol
Behav. 2015;152:4505.
11.Azad MB, Sharma AK, de Souza RJ, et al. Association between
artificially sweetened beverage consumption during pregnancy and
infant body mass index. JAMA Pediatr. 2016;170:66270. This
Curr Nutr Rep
epidemiological study showed that daily consumption of NNS
was associated with a 0.2 unit increase in infant BMI z-score as
well as a greater risk for being overweight at 1 year of age.
12. Murray S, Tulloch A, Criscitelli K, Avena NM. Recent studies of
the effects of sugars on brain systems involved in energy balance
and reward: relevance to low calorie sweeteners. Physiol Behav.
2016;164:5048.
13. Zhu Y, Olsen SF, Mendola P, Halldorsson TI, Rawal S, Hinkle SN,
et al. Maternal consumption of artificially sweetened beverages
during pregnancy, and offspring growth through 7 years of age: a
prospective cohort study. Int J Epidemiol. 2017;46:1499508.
14. The InterAct consortium. Consumption of sweet beverages and
type 2 diabetes incidence in European adults: results from EPIC-
InterAct. Diabetologia. 2013;56:152030.
15. Maersk M, Belza A, Holst JJ, Fenger-Grøn M, Pedersen SB, Astrup
A, et al. Satiety scores and satiety hormone response after sucrose-
sweetened soft drink compared with isocaloric semi-skimmed milk
and with non-caloric soft drink: a controlled trial. Eur J Clin Nutr.
2012;66:5239.
16. Peters JC, Wyatt HR,Foster GD, Pan Z, Wojtanowski AC, Vander
Veur SS, et al. The effects of water and non-nutritive sweetened
beverages on weight loss during a 12-week weight loss treatment
program. Obesity (Silver Spring). 2014;22:141521.
17. Peters JC, Beck J. Low calorie sweetener (LCS) use and energy
balance. Physiol Behav. 2016;164:5248.
18.Higgins KA, Mattes RD. A randomized controlled trial contrasting
the effects of 4 low-calorie sweeteners and sucrose on body weight
in adults with overweight or obesity. Am J Clin Nutr. 2019;109:
1288301. This study compared the effects of 4 different NNS
directly, and the authors found that a 12-week exposure to
aspartame, sucralose, saccharin, or stevia had no effect on pe-
ripheral insulin. Interestingly, while saccharin consumption in-
creased body weight, participants who consumed sucralose ex-
perienced a (non-significant) directionally negative change in
weight.
19. Tey SL, Salleh NB, Henry CJ, Forde CG. Effects of non-nutritive
(artificial vs natural) sweeteners on 24-h glucose profiles. Eur J Clin
Nutr. 2017;71:112932.
20. Margolskee RF, Dyer J, Kokrashvili Z, Salmon KSH, Ilegems E,
Daly K, et al. T1R3 and gustducin in gut sense sugars to regulate
expression of Na+glucose cotransporter 1. Proc Natl Acad Sci U S
A. 2007;104:1507580.
21. Veldhuizen MG, Babbs RK, Patel B, Fobbs W, Kroemer NB,
Garcia E, et al. Integration of sweet taste and metabolism deter-
mines carbohydrate reward. Curr Biol. 2017;27:24762485.e6.
22. Lanfer A, Knof K, Barba G, Veidebaum T, Papoutsou S, de
Henauw S, et al. Taste preferences in association with dietary habits
and weight status in European children: results from the IDEFICS
study. Int J Obes. 2012;36:2734.
23. Salbe AD, DelParigiA, Pratley RE, Drewnowski A, Tataranni PA.
Taste preferences and body weight changes in an obesity-prone
population. Am J Clin Nutr. 2004;79:3728.
24. Matsushita Y, Mizoue T, Takahashi Y, Isogawa A, Kato M, Inoue
M, et al. Taste preferences and body weight change in Japanese
adults: the JPHC study. Int J Obes. 2009;33:11917.
25. Mennella JA, Jagnow CP, Beauchamp GK. Prenatal and postnatal
flavor learning by human infants. Pediatrics. 2001;107:E88.
26. Mennella JA, Castor SM. Sensitive period in flavor learning: effects
of duration of exposure to formula flavors on food likes during
infancy. Clin Nutr. 2012;31:10225.
27. Frihauf JB, Fekete ÉM,Nagy TR, Levin BE, Zorrilla EP. Maternal
Western diet increases adiposity even in male offspring of obesity-
resistant rat dams: early endocrine risk markers. Am J Phys Regul
Integr Comp Phys. 2016;311:R104559.
28. Rosales-Gómez CA, Martínez-Carrillo BE, Reséndiz-Albor AA,
Ramírez-Durán N, Valdés-Ramos R, Mondragón-Velásquez T,
et al. Chronic consumption of sweeteners and its effect on
glycaemia, cytokines, hormones, and lymphocytes of GALT in
CD1 mice. Biomed Res Int. 2018;2018:115.
29. Zhang G-H, Chen M-L, Liu S-S, Zhan Y-H, Quan Y, Qin Y-M,
et al. Effects of mothers dietary exposure to acesulfame-K in preg-
nancy or lactation on the adult offsprings sweet preference. Chem
Senses. 2011;36:76370.
30. Chen M-L, Liu S-S, Zhang G-H, Quan Y, Zhan Y-H, Gu T-Y, et al.
Effects of early intraoral acesulfame-K stimulation to mice on the
adults sweet preference and the expression of α-gustducin in fun-
giform papilla. Chem Senses. 2013;38:44755.
31. Zhang G-H, Chen M-L, Liu S-S, Zhan Y-H, Quan Y, Qin Y-M,
et al. Facilitation of the development of fungiform taste buds by
early intraoral acesulfame-K stimulation to mice. J Neural Transm.
2010;117:12614.
32. Sylvetsky AC, Gardner AL, Bauman V, Blau JE, Garraffo HM,
Walter PJ, et al. Nonnutritive sweeteners in breast Milk. J Toxicol
Environ Health Part A. 2015;78:102932.
33.Sylvetsky AC, Conway EM, Malhotra S, Rother KI. Development
of sweet taste perception: implications for artificial sweetener use.
Endocr Dev. 2017;32:8799. An informative recent review re-
garding the potential effects of NNS exposure on taste percep-
tion and preferences throughout fetal development, childhood,
and adolescence.
34. Seferidi P, Millett C, Laverty AA. Sweetened beverage intake in
association to energy and sugar consumption and cardiometabolic
markers in children. Pediatr Obes. 2018;13:195203.
35.•• Sylvetsky AC, Figueroa J, Zimmerman T, Swithers SE, Welsh JA.
Consumption of low-calorie sweetened beverages is associated
with higher total energy and sugar intake among children,
NHANES 20112016. Pediatr Obes. 2019;14:e12535. This recent
large observational study using NHANES data found associa-
tions in children and adolescents between habitual NNS con-
sumption and higher dietary energy, carbohydrate, total sugar,
and added sugar intake.
36. Sylvetsky AC, Jin Y, Mathieu K, DiPietro L, Rother KI,
Talegawkar SA. Low-calorie sweeteners: disturbing the energy
balance equation in adolescents? Obesity (Silver Spring).
2017;25:204954.
37. Swithers SE. Artificial sweeteners produce the counterintuitive ef-
fect of inducing metabolic derangements. Trends Endocrinol
Metab. 2013;24:43141.
38. Swithers SE. Artificial sweeteners are not the answer to childhood
obesity. Appetite. 2015;93:8590.
39. Wang Q-P, Lin YQ, Zhang L, Wilson YA, Oyston LJ, Cotterell J,
et al. Sucralose promotes food intake through NPY and a neuronal
fasting response. Cell Metab. 2016;24:7590.
40. Musso P-Y, Lampin-Saint-Amaux A, Tchenio P, Preat T. Ingestion
of artificial sweeteners leads to caloric frustration memory in dro-
sophila. Nat Commun. 2017;8:1803.
41. Davidson TL, Martin AA, Clark K, Swithers SE. Intake of high-
intensity sweeteners alters the ability of sweet taste to signal caloric
consequences: implications for the learned control of energy and
body weight regulation. Q J Exp Psychol. 2011;64:143041.
42.Casperson SL, Johnson L, Roemmich JN. The relative reinforcing
value of sweet versus savory snack foods after consumption of
sugar- or non-nutritive sweetened beverages. Appetite. 2017;112:
1439. This study utilized experimental design to determine
how the acute ingestion of a NNS beverage, relative to a
sugar-sweetened drink, influenced eating behavior after a stan-
dardized meal, among healthy adults. The authors found that
the NNS drink, but not the sugar-sweetened beverage, in-
creased the reinforcing value of sweet snack foods.
43. Fantino M, Fantino A, Matray M, Mistretta F. Beverages contain-
ing low energy sweeteners do not differ from water in their effects
Curr Nutr Rep
on appetite, energy intake and food choices in healthy, non-obese
French adults. Appetite. 2018;125:55765.
44. Hill SE, Prokosch ML, Morin A, Rodeheffer CD. The effect of non-
caloric sweeteners on cognition, choice, and post-consumption sat-
isfaction. Appetite. 2014;83:828.
45. Farkas A, Híd J. The black agonist-receptor model of high potency
sweeteners, and its implication to sweetness taste and sweetener
design. J Food Sci. 2011;76:S4658.
46. Xu H, Staszewski L, Tang H, Adler E, Zoller M, Li X. Different
functional roles of T1R subunits in the heteromeric taste receptors.
Proc Natl Acad Sci U S A. 2004;101:1425863.
47. Nie Y, Vigues S, Hobbs JR, Conn GL, Munger SD. Distinct con-
tributions of T1R2 and T1R3 taste receptor subunits to the detection
of sweet stimuli. Curr Biol. 2005;15:194852.
48. Jang H-J, Kokrashvili Z, Theodorakis MJ, Carlson OD, Kim BJ,
Zhou J, et al. Gut-expressed gustducin and taste receptors regulate
secretion of glucagon-like peptide-1. Proc Natl Acad Sci U S A.
2007;104:1506974.
49. Nakagawa Y, Nagasawa M, Yamada S, Hara A, Mogami H,
Nikolaev VO, et al. Sweet taste receptor expressed in pancreatic
beta-cells activates the calcium and cyclic AMP signaling systems
and stimulates insulin secretion. PLoS One. 2009;4:e5106.
50. Ohtsu Y, Nakagawa Y, Nagasawa M, Takeda S, Arakawa H,
Kojima I. Diverse signaling systems activated by the sweet taste
receptor in human GLP-1-secreting cells. Mol Cell Endocrinol.
2014;394:709.
51. Kojima I, Nakagawa Y, Hamano K, Medina J, Li L, Nagasawa M.
Glucose-sensing receptor T1R3: a new signaling receptor activated
by glucose in pancreatic β-cells. Biol Pharm Bull. 2015;38:6749.
52. Li L, Ohtsu Y, Nakagawa Y, Masuda K, Kojima I. Sucralose, an
activator of the glucose-sensing receptor, increases ATP by
calcium-dependent and -independent mechanisms. Endocr J.
2016;63:71525.
53. Fowler SPG. Low-calorie sweetener use and energy balance: results
from experimental studies in animals, and large-scale prospective
studies in humans. Physiol Behav. 2016;164:51723.
54. Brown RJ, Walter M, Rother KI. Ingestion of diet soda before a
glucose load augments glucagon-like peptide-1 secretion. Diabetes
Care. 2009;32:21846.
55. Brown RJ, Walter M, Rother KI. Effects of diet soda on gut hor-
mones in youths with diabetes. Diabetes Care. 2012;35:95964.
56.Sylvetsky AC, Brown RJ, Blau JE, Walter M, Rother KI. Hormonal
responses to non-nutritive sweeteners in water and diet soda. Nutr
Metab (Lond). 2016;13:71. This study showed that diet soda,
relative to a NNS dissolved in water, increased GLP-1 release.
57. Temizkan S, Deyneli O, Yasar M, Arpa M, Gunes M, Yazici D,
et al. Sucralose enhances GLP-1 release and lowers blood glucose
in the presence of carbohydrate in healthy subjects but not in pa-
tients with type 2 diabetes. Eur J Clin Nutr. 2015;69:1626.
58. Pepino MY, Tiemann CD, Patterson BW, Wice BM, Klein S.
Sucralose affects glycemic and hormonal responses to an oral glu-
cose load. Diabetes Care 2013; DC_122221.
59. Wu T, Bound MJ, Standfield SD, Bellon M, Young RL, Jones KL,
et al. Artificial sweeteners have no effect on gastric emptying,
glucagon-like peptide-1, or glycemia after oral glucose in healthy
humans. Diabetes Care. 2013;36:e2023.
60. Wu T, Zhao BR, Bound MJ, Checklin HL, Bellon M, Little TJ,
et al. Effects of different sweet preloads on incretin hormone secre-
tion, gastric emptying, and postprandial glycemia in healthy
humans. Am J Clin Nutr. 2012;95:7883.
61. Ma J, Bellon M, Wishart JM, Young R, Blackshaw LA, Jones KL,
et al. Effect of the artificial sweetener, sucralose, on gastric empty-
ing and incretin hormone release in healthy subjects. Am J Physiol
Gastrointest Liver Physiol. 2009;296:G7359.
62. Ma J, Chang J, Checklin HL, Young RL, Jones KL, Horowitz M,
et al. Effect of the artificial sweetener, sucralose, on small intestinal
glucose absorption in healthy human subjects. Br J Nutr. 2010;104:
8036.
63. Steinert RE, Frey F, Toepfer A, Drewe J, Beglinger C. Effects of
carbohydrate sugars and artificial sweeteners on appetite and the
secretion of gastrointestinal satiety peptides. Br J Nutr. 2011;24:
19.
64. Ford HE, Peters V, Martin NM, Sleeth ML, Ghatei MA, Frost GS,
et al. Effectsof oral ingestion of sucralose on gut hormone response
and appetite in healthy normal-weight subjects. Eur J Clin Nutr.
2011;65:50813.
65.Tey SL, Salleh NB, Henry J, Forde CG. Effects of aspartame-,
monk fruit-, stevia- and sucrose-sweetened beverages on postpran-
dial glucose, insulin and energy intake. Int J Obes. 2017;41:4507.
This study showed that two different types of NNS (stevia and
monk fruit) both have an effect on post-prandial insulin release
in combination with a meal.
66.•• Nichol AD, Salame C, Rother KI, Pepino MY. Effects of sucralose
ingestion versus sucralose taste on metabolic responses to an oral
glucose tolerance test in participants with normal weight and obe-
sity: a randomized crossover trial. Nutrients. 2019. https://doi.org/
10.3390/nu12010029.This study demonstrated that sucralose
decreased plasma insulin concentrations in lean participants,
but increased insulin concentrations among obese
participants, when tasted or ingested. These findings suggest
that sucralose may have differential peripheral effects
depending on body weight and metabolic health.
67. Sakurai K, Lee EY, Morita A, Kimura S, Kawamura H, Kasamatsu
A, et al. Glucagon-like peptide-1 secretion by direct stimulation of
L cells with luminal sugar vs non-nutritive sweetener. J Diabetes
Invest. 2012;3:15663.
68. Brown AW, Bohan Brown MM, Onken KL, Beitz DC. Short-term
consumption of sucralose, a nonnutritive sweetener, is similar to
water with regard to select markers of hunger signaling and short-
term glucose homeostasis in women. Nutr Res. 2011;31:8828.
69. Lertrit A, Srimachai S, Saetung S, Chanprasertyothin S, Chailurkit
L, Areevut C, et al. Effects of sucralose on insulin and glucagon-
like peptide-1 secretion in healthy subjects: a randomized, double-
blind, placebo-controlled trial. Nutrition. 2018;5556:12530.
70. Romo-Romo A, Aguilar-Salinas CA, Brito-Córdova GX, Gómez-
Díaz RA, Almeda-Valdes P. Sucralose decreases insulin sensitivity
in healthy subjects: a randomized controlled trial. Am J Clin Nutr.
2018;108:48591.
71.Ahmad SY, Friel JK, MacKay DS. The effect of the artificial sweet-
eners on glucose metabolism in healthy adults: a randomized
double-blinded crossover clinical trial. Appl Physiol Nutr Metab.
2019. https://doi.org/10.1139/apnm-2019-0359.This study found
no change in peripheral insulin, GLP-1, or leptin levels or
insulin sensitivity in healthy, lean participants exposed for 12
weeks to aspartame or sucralose mixed in water.
72. Bonnet F, Tavenard A, Esvan M, Laviolle B, Viltard M, Lepicard
EM, et al. Consumption of a carbonated beverage with high-
intensity sweeteners has no effect on insulin sensitivity and secre-
tion in nondiabetic adults. J Nutr. 2018;148:12939.
73. Higgins KA, Considine RV, Mattes RD. Aspartame consumption
for 12 weeks does not affect glycemia, appetite, or body weight of
healthy, lean adults in a randomized controlled trial. J Nutr.
2018;148:6507.
74. Grotz VL, Henry RR, McGill JB, Prince MJ,Shamoon H, Trout JR,
et al. Lack of effect of sucralose on glucose homeostasis in subjects
with type 2 diabetes. J Am Diet Assoc. 2003;103:160712.
75. Grotz VL, Pi-Sunyer X, Porte D, Roberts A, Richard Trout J. A 12-
week randomized clinical trial investigating the potential for sucra-
lose to affect glucose homeostasis. Regul Toxicol Pharmacol.
2017;88:2233.
76. Collison KS, Makhoul NJ, Zaidi MZ, Al-Rabiah R, Inglis A,
Andres BL, et al. Interactive effects of neonatal exposure to
Curr Nutr Rep
monosodium glutamate and aspartame on glucose homeostasis.
Nutr Metab (Lond). 2012;9:58.
77. Collison KS, Inglis A, Shibin S, Andres B, Ubungen R, Thiam J,
et al. Differential effects of early-life NMDA receptor antagonism
on aspartame-impaired insulin tolerance and behavior. Physiol
Behav. 2016;167:20921.
78. Frank GKW, Oberndorfer TA, Simmons AN, Paulus MP, Fudge
JL, Yang TT, et al. Sucrose activates human taste pathways differ-
ently from artificial sweetener. NeuroImage. 2008;39:155969.
79. Smeets PAM, Weijzen P, de Graaf C, Viergever MA. Consumption
of caloric and non-caloric versions of a soft drink differentially
affects brain activation during tasting. NeuroImage. 2011;54:
136774.
80. Smeets PAM, de Graaf C, Stafleu A, van Osch MJP, van der Grond
J. Functional MRI of human hypothalamic responses following
glucose ingestion. NeuroImage. 2005;24:3638.
81. Page KA, Chan O, Arora J, Belfort-DeAguiar R, Dzuira J,
Roehmholdt B, et al. Effects of fructose vs glucose on regional
cerebral blood flow in brain regions involved with appetite and
reward pathways. JAMA. 2013;309:6370.
82. Luo S, Melrose AJ, Dorton H, Alves J, Monterosso JR, Page KA.
Resting state hypothalamic response to glucose predicts glucose-
induced attenuation in the ventral striatal response to food cues.
Appetite. 2017;116:46470.
83. Matsuda M, Liu Y, Mahankali S, Pu Y,MahankaliA, Wang J, et al.
Altered hypothalamic function in response to glucose ingestion in
obese humans. Diabetes. 1999;48:18016.
84. Jastreboff AM, Sinha R, Arora J, Giannini C, Kubat J, Malik S,
et al. Altered brain response to drinking glucose and fructose in
obese adolescents. Diabetes. 2016;65:192939.
85. Page KA, Luo S, Wang X, Chow T, Alves J, Buchanan TA, et al.
Children exposed to maternal obesity or gestational diabetes during
early fetal development have hypothalamic alterations that predict
future weight gain. Diabetes Care. 2019;42:147380. https://doi.
org/10.2337/dc18-2581.
86. Smeets PAM, de Graaf C, Stafleu A, van Osch MJP, van der Grond
J. Functional magnetic resonance imaging of human hypothalamic
responses to sweet taste and calories. Am J Clin Nutr. 2005;82:
10116.
87. van Opstal AM, Kaal I, van den Berg-Huysmans AA, Hoeksma M,
Blonk C, Pijl H, et al. Dietary sugars and non-caloric sweeteners
elicit different homeostatic and hedonic responses in the brain.
Nutrition. 2019;60:806.
88.•• van Opstal AM, Hafkemeijer A, van den Berg-Huysmans AA,
HoeksmaM,MulderTPJ,PijlH,etal.Brainactivityand
connectivity changes in response to nutritive natural sugars, non-
nutritive natural sugar replacements and artificial sweeteners. Nutr
Neurosci. 2019; This recent fMRI study expands upon prior
work demonstrating that NNS may not have a similar satiating
effect on the brain as nutritive sweeteners. In this study, healthy
adults are given fat/protein milkshakes sweetened with either
glucose, fructose, allulose, or sucralose. Unlike glucose, sucra-
lose had no effect on BOLD signaling, which supports the hy-
pothesis that sweet tasteabsent of nutritive carbohydrates, even
in the presence of fat/protein, may not lead to hypothalamic
connectivity changes generally associated with fullness.
89. Kohno D. Sweet taste receptor in the hypothalamus: a potential new
player in glucose sensing in the hypothalamus. J Physiol Sci.
2017;67:45965.
90.•• Crézé C, Candal L, Cros J, Knebel J-F, Seyssel K, Stefanoni N,
et al. The impact of caloric and non-caloric sweeteners on food
intake and brain responses to food: a randomized crossover con-
trolled trial in healthy humans. Nutrients. 2018;10:615. Findings
showed that the acute consumption of a NNS drink prompted
differential neural activation towards palatable food cues, com-
pared with a sucrose drink or a water control, among healthy
adults.
91. Crézé C, Notter-Bielser M-L, Knebel J-F, Campos V, Tappy L,
Murray M, et al. The impact of replacing sugar- by artificially-
sweetened beverages on brain and behavioral responses to food
viewing - an exploratory study. Appetite. 2018;123:1608.
92. Rudenga KJ, Small DM. Amygdala response to sucrose consump-
tion is inversely related to artificial sweetener use. Appetite.
2012;58:5047.
93. Green E, Murphy C. Altered processing of sweet taste in the brain
of diet soda drinkers. Physiol Behav. 2012;107:5607.
94. Opstal AMV, Hafkemeijer A, Berg-Huysmans AA van den,
Hoeksma M, Mulder TPJ, Pijl H, Rombouts SARB, Grond J van
der (2019) Brain activity and connectivity changes in response to
nutritive natural sugars, non-nutritive natural sugar replacements
and artificial sweeteners. Nutr Neurosci 0:111.
95. Curtis KS, Davis LM, Johnson AL, Therrien KL, Contreras RJ. Sex
differences in behavioral taste responses to and ingestion of sucrose
and NaCl solutions by rats. Physiol Behav. 2004;80:65764.
PublishersNoteSpringer Nature remains neutral with regard to jurisdic-
tional claims in published maps and institutional affiliations.
Curr Nutr Rep
... However, only a small percentage of participants (17.51%) reported frequently consuming non-nutritive sweeteners to replace sucrose in their diet, highlighting a discrepancy between preferences and actual behavior regarding sweetener consumption. Factors like taste preferences, cultural influences, availability of alternatives, and personal beliefs about the risks or benefits of non-nutritive sweeteners may shape participants' dietary choices (26). The considerable attention to the potential risks associated with non-nutritive sweeteners reflected a demand for clear and transparent information on health-related concerns. ...
Article
Full-text available
Introduction This study aimed to explore the knowledge, attitude, and practice (KAP) toward non-nutritive sweeteners among a population with reduced sugar intake requirements. Methods This cross-sectional study used self-developed questionnaires to collect demographic characteristics and KAP towards non-nutritive sweeteners among respondents with reduced sugar intake requirements, i.e., overweight or obese individuals and patients with pre-diabetes or diabetes. Results A total of 639 valid questionnaires were collected, and 51.64% of participants were male. The KAP scores were 7.63 ± 3.58 (range: 0–11), 34.28 ± 7.47 (range: 12–60), and 15.48 ± 3.97 (range: 7–35), respectively. Pearson’s correlation analysis showed that knowledge score was positively correlated with attitude (r = 0.229, p < 0.001) and practice score (r = 0.467, p < 0.001), while attitude was positively correlated with practice (r = 0.312, p < 0.001). The structural equation model showed that knowledge was directly and positively associated with attitude (path coefficient = 0.48, p < 0.001) and practice (path coefficient = 0.46, p < 0.001). In addition, the attitude was directly and positively associated with practice (path coefficient = 0.12, p < 0.001). Besides, diabetes was associated with lower knowledge (path coefficient = −0.81, p = 0.038) and practice (path coefficient = −0.42, p < 0.041). Discussion Population with the reduced sugar intake requirement showed poor knowledge, negative attitudes, and suboptimal practices toward non-nutritive sweeteners. To optimize the utilization of non-nutritive sweeteners in accordance with medical prescriptions, especially for individuals with diabetes, tailored educational interventions may be designed for participants with lower KAP.
... Regarding the effects of specific components and features of UPF, the content of low-/no-calorie sweeteners (LNCSs) [174][175][176], their organoleptic properties (i.e., taste, texture) [177], and design (e.g., ready to consume) [153] have been associated with reduced satiety and overeating [51]. Regarding LNCSs, neuroimaging studies have provided evidence that the sweet taste in the absence of nutritive carbohydrates may not lead to changes in the functioning of the hypothalamus [178,179] and brain regions of the ventral system (i.e., the nucleus accumbens, and the insula) [180]. ...
Article
Full-text available
Binge-eating disorder (BED) is a highly prevalent disorder. Subthreshold BED conditions (sBED) are even more frequent in youth, but their significance regarding BED etiology and long-term prognosis is unclear. A better understanding of brain findings associated with BED and sBED, in the context of critical periods for neurodevelopment, is relevant to answer such questions. The present narrative review starts from the knowledge of the development of emotional self-regulation in youth, and the brain circuits supporting emotion-regulation and eating behaviour. Next, neuroimaging studies with sBED and BED samples will be reviewed, and their brain-circuitry overlap will be examined. Deficits in inhibition control systems are observed to precede, and hyperactivity of reward regions to characterize, sBED, with overlapping findings in BED. The imbalance between reward/inhibition systems, and the implication of interoception/homeostatic processing brain systems should be further examined. Recent knowledge of the potential impact that the high consumption of ultra-processed foods in paediatric samples may have on these sBED/BED-associated brain systems is then discussed. There is a need to identify, early on, those sBED individuals at risk of developing BED at neurodevelopmental stages when there is a great possibility of prevention. However, more neuroimaging studies with sBED/BED pediatric samples are needed.
... In fact, previous research conducted in the US found that only one in four adults correctly identified packaged foods containing NNS (Sylvetsky et al., 2014). However, it is important that consumers are able to identify NNS in foods, particularly as exposure to these sweeteners during early development may influence sweet taste preferences, increasing the motivation to consume sweet foods (Yunker et al., 2020). In a recent paper from Chile, all children included in the study (n = 250) had consumed at least one NNS during the previous month of life (Martínez et al., 2020). ...
Article
This study investigated the use of added sugars and non-nutritive sweeteners (NNS) in 4,805 packaged foods (including non-alcoholic beverages) sold in a major supermarket in Brazil. The ingredient lists of all products were searched for the presence of added sugars and NNS. The added sugar content was estimated and compared between products containing added sugar, with and without NNS. Using criteria set by the new Brazilian food label rules, we determined whether foods with added sugars and NNS were classified as “high in added sugar”. Most products (66.7%) contained at least one type of sweetener (added sugars or NNS). Of the products containing NNS (n = 597), 81.5% also contained added sugar. Products containing both NNS and added sugar ingredients showed a lower overall added sugar content than products containing added sugars only. However, for a few food categories (chocolates, dairy and non-dairy dessert mixes and candies), the added sugar content did not differ significantly between products with and without NNS. Sixty-three percent of products containing both added sugar and NNS were classified as “high in added sugar”.
... On the contrary, preliminary evidence suggests that LNCSs may not appropriately regulate the brain network involved in appetite and reward to process sweet taste. This has been suggested to prompt an extension of the meal episodes to match the expected energy needs through induced variations in the expected signals received by the brain [148,149]. Findings in healthy samples support that the sweet taste in the absence of nutritive carbohydrates may not lead to hypothalamic changes that are typically linked to satiation [150,151]. The study of van Opstal and colleagues [152] expanded this initial evidence by showing that unlike glucose and fructose sweetened fat/protein milkshakes, the ingestion of those sweetened with sucralose and allulose had no effect on the functioning of several brain areas, including the insula, and reward-based regions such as the ventral tegmental area and the nucleus accumbens. ...
Article
Full-text available
Ultra-processed foods and drinks (UPF) are formulation of ingredients, mostly of exclusive industrial use, that result from a series of industrial processes. They usually have a low nutrient but high energy density, with a high content of saturated and trans fats, and added sugars. In addition, they have characteristic organoleptic properties, and usually contain sophisticated additives, including artificial sweeteners, to intensify their sensory qualities and imitate the appearance of minimally processed foods. In addition, recent research has warned about the presence of chemicals (e.g., bisphenol) and neo-formed contaminants in these products. UPF production and consumption growth have been spectacular in the last decades, being specially consumed in children and adolescents. UPF features have been associated with a range of adverse health effects such as overeating, the promotion of inflammatory and oxidative stress processes, gut dysbiosis, and metabolic dysfunction including problems in glucose regulation. The evidence that these UPF-related adverse health effects may have on the neural network implicated in eating behavior are discussed, including the potential impact on serotonergic and dopaminergic neurotransmission, brain integrity and function. We end this review by placing UPF in the context of current food environments, by suggesting that an increased exposure to these products through different channels, such as marketing, may contribute to the automatic recruitment of the brain regions associated with food consumption and choice, with a detrimental effect on inhibitory-related prefrontal cortices. While further research is essential, preliminary evidence point to UPF consumption as a potential detrimental factor for brain health and eating behavior.
... Overall, the existing literature shows mixed results on the effects of NNS on appetite, glucose metabolism, and body weight, [2][3][4][5][6] with no clear consensus on whether NNSs are beneficial or harmful for health. 7,8 Prior work [9][10][11][12][13][14] provides evidence that brain areas involved in regulation of taste, reward, and homeostasis may respond differently to NNSs compared with nutritive sugars, yet a number of questions still remain. Of note, the majority of previous studies in humans examining brain responses to NNS compared with nutritive sweeteners have been largely limited to studies of individuals with normal weight [9][10][11][12][15][16][17] and exclusively male cohorts. ...
Article
Full-text available
Importance Nonnutritive sweeteners (NNSs) are used as an alternative to nutritive sweeteners to quench desire for sweets while reducing caloric intake. However, studies have shown mixed results concerning the effects of NNSs on appetite, and the associations between sex and obesity with reward and appetitive responses to NNS compared with nutritive sugar are unknown. Objective To examine neural reactivity to different types of high-calorie food cues (ie, sweet and savory), metabolic responses, and eating behavior following consumption of sucralose (NNS) vs sucrose (nutritive sugar) among healthy young adults. Design, Setting, and Participants In a randomized, within-participant, crossover trial including 3 separate visits, participants underwent a functional magnetic resonance imaging task measuring blood oxygen level–dependent signal in response to visual cues. For each study visit, participants arrived at the Dornsife Cognitive Neuroimaging Center of University of Southern California at approximately 8:00 am after a 12-hour overnight fast. Blood was sampled at baseline and 10, 35, and 120 minutes after participants received a drink containing sucrose, sucralose, or water to measure plasma glucose, insulin, glucagon-like peptide(7-36), acyl-ghrelin, total peptide YY, and leptin. Participants were then presented with an ad libitum meal. Participants were right-handed, nonsmokers, weight-stable for at least 3 months before the study visits, nondieters, not taking medication, and with no history of eating disorders, illicit drug use, or medical diagnoses. Data analysis was performed from March 2020 to March 2021. Interventions Participants ingested 300-mL drinks containing either sucrose (75 g), sucralose (individually sweetness matched), or water (as a control). Main Outcomes and Measures Primary outcomes of interest were the effects of body mass index (BMI) status and sex on blood oxygen level–dependent signal to high-calorie food cues, endocrine, and feeding responses following sucralose vs sucrose consumption. Secondary outcomes included neural, endocrine, and feeding responses following sucrose vs water and sucralose vs water (control) consumption, and cue-induced appetite ratings following sucralose vs sucrose (and vs water). Results A total of 76 participants were randomized, but 2 dropped out, leaving 74 adults (43 women [58%]; mean [SD] age, 23.40 [3.96] years; BMI range, 19.18-40.27) who completed the study. In this crossover design, 73 participants each received water (drink 1) and sucrose (drink 2), and 72 participants received water (drink 1), sucrose (drink 2), and sucralose (drink 3). Sucrose vs sucralose was associated with greater production of circulating glucose, insulin, and glucagon-like peptide–1 and suppression of acyl-ghrelin, but no differences were found for peptide YY or leptin. BMI status by drink interactions were observed in the medial frontal cortex (MFC; P for interaction < .001) and orbitofrontal cortex (OFC; P for interaction = .002). Individuals with obesity (MFC, β, 0.60; 95% CI, 0.38 to 0.83; P < .001; OFC, β, 0.27; 95% CI, 0.11 to 0.43; P = .002), but not those with overweight (MFC, β, 0.02; 95% CI, –0.19 to 0.23; P = .87; OFC, β, –0.06; 95% CI, –0.21 to 0.09; P = .41) or healthy weight (MFC, β, –0.13; 95% CI, –0.34 to 0.07; P = .21; OFC, β, –0.08; 95% CI, –0.23 to 0.06; P = .16), exhibited greater responsivity in the MFC and OFC to savory food cues after sucralose vs sucrose. Sex by drink interactions were observed in the MFC (P for interaction = .03) and OFC (P for interaction = .03) after consumption of sucralose vs sucrose. Female participants had greater MFC and OFC responses to food cues (MFC high-calorie vs low-calorie cues, β, 0.21; 95% CI, 0.05 to 0.37; P = .01; MFC sweet vs nonfood cues, β, 0.22; 95% CI, 0.02 to 0.42; P = .03; OFC food vs nonfood cues, β, 0.12; 95% CI, 0.02 to 0.22; P = .03; and OFC sweet vs nonfood cues, β, 0.15; 95% CI, 0.03 to 0.27; P = .01), but male participants’ responses did not differ (MFC high-calorie vs low-calorie cues, β, 0.01; 95% CI, –0.19 to 0.21; P = .90; MFC sweet vs nonfood cues, β, −0.04; 95% CI, –0.26 to 0.18; P = .69; OFC food vs nonfood cues, β, −0.08; 95% CI, –0.24 to 0.08; P = .32; OFC sweet vs nonfood cues, β, –0.11; 95% CI, –0.31 to 0.09; P = .31). A sex by drink interaction on total calories consumed during the buffet meal was observed (P for interaction = .03). Female participants consumed greater total calories (β, 1.73; 95% CI, 0.38 to 3.08; P = .01), whereas caloric intake did not differ in male participants (β, 0.68; 95% CI, –0.99 to 2.35; P = .42) after sucralose vs sucrose ingestion. Conclusions and Relevance These findings suggest that female individuals and those with obesity may be particularly sensitive to disparate neural responsivity elicited by sucralose compared with sucrose consumption. Trial Registration ClinicalTrials.gov Identifier: NCT02945475
Article
Full-text available
It is widely believed that exposure to sweetened foods and beverages stimulates the liking and desire for sweetness. Here we provide an updated review of the empirical evidence from human research examining whether exposure to sweet foods or beverages influences subsequent general liking for sweetness (“sweet tooth”), based on the conclusions of existing systematic reviews and more recent research identified from a structured search of literature. Prior reviews have concluded that the evidence for a relationship between sweet taste exposure and measures of sweet taste liking is equivocal, and more recent primary research generally does not support the view that exposure drives increased liking for sweetness, in adults or children. In intervention trials using a range of designs, acute exposure to sweetness usually has the opposite effect (reducing subsequent liking and desire for sweet taste), while sustained exposures have no significant effects, or inconsistent effects. Recent longitudinal observational studies in infants and children also report no significant associations of exposures to sweet foods and beverages with measures of sweet taste preferences. Overall, while it is widely assumed that exposure to sweetness stimulates a greater liking and desire for sweetness, this is not borne out by the balance of empirical evidence. While new research may provide a more robust evidence base, there are also a number of methodological, biological and behavioural considerations that may underpin the apparent absence of a positive relationship between sweetness exposure and liking.
Article
Full-text available
Conceptualising ultra-processed foods high in carbohydrates and fats as addictive substances can contribute to efforts to improve health, argue Ashley Gearhardt and colleagues
Article
Children and adolescents are the highest consumers of added sugars, particularly from sugar-sweetened beverages (SSB). Regular consumption of SSB early in life induces a variety of negative consequences on health that can last into adulthood. Low-calorie sweeteners (LCS) are increasingly used as an alternative to added sugars because they provide a sweet sensation without adding calories to the diet. However, the long-term effects of early-life consumption of LCS are not well understood. Considering LCS engage at least one of the same taste receptors as sugars and potentially modulate cellular mechanisms of glucose transport and metabolism, it is especially important to understand how early-life LCS consumption impacts intake of and regulatory responses to caloric sugars. In our recent study, we found that habitual intake of LCS during the juvenile-adolescence period significantly changed how rats responded to sugar later in life. Here, we review evidence that LCS and sugars are sensed via common and distinct gustatory pathways, and then discuss the implications this has for shaping sugar-associated appetitive, consummatory, and physiological responses. Ultimately, the review highlights the diverse gaps in knowledge that will be necessary to fill to understand the consequences of regular LCS consumption during important phases of development.
Article
Full-text available
Sugar is crucial as an essential nutrient for humans as well as for providing texture, sweetness and so on to food. But with the rise in people’s pursuit of health, it is becoming increasingly clear that excessive consumption of sugar can locate a load on the body. It has been that excessive sugar is associated with many diseases, such as dental caries, obesity, diabetes, and coronary heart disease. Therefore, researchers and industries are trying to reduce or substitute sugar in food without affecting the sensory evaluation. Substituting sugar with sweeteners is alternatively becoming the most traditional way to minimize its use. So far, the sweeteners such as stevia and xylitol have been are commercially applied. Several studies have shown that technological innovation can partially compensate for the loss in sweetness as a result of sugar reduction, such as cross-modal interactions that stimulate sweetness with aroma, nanofiltration that filters disaccharides and above, enzyme-catalyzed sugar hydrolysis, and microbial fermentation that turns sugar into sugar alcohol. This review summarizes these studies to enhance the safety and quality of sugar-reduced products, and will provide some theoretical frameworks for the food industry to reduce sugar in foods, meet consumers’ needs, and promote human health.
Article
Background: There is growing evidence that an addictive-eating phenotype may exist. There is significant debate regarding whether highly processed foods (HPFs; foods with refined carbohydrates and/or added fats) are addictive. The lack of scientifically grounded criteria to evaluate the addictive nature of HPFs has hindered the resolution of this debate. Analysis: The most recent scientific debate regarding a substance's addictive potential centered around tobacco. In 1988, the Surgeon General issued a report identifying tobacco products as addictive based on three primary scientific criteria: their ability to (1) cause highly controlled or compulsive use, (2) cause psychoactive (i.e. mood-altering) effects via their effect on the brain and (3) reinforce behavior. Scientific advances have now identified the ability of tobacco products to (4) trigger strong urges or craving as another important indicator of addictive potential. Here, we propose that these four criteria provide scientifically valid benchmarks that can be used to evaluate the addictiveness of HPFs. Then, we review the evidence regarding whether HPFs meet each criterion. Finally, we consider the implications of labeling HPFs as addictive. Conclusion: Highly processed foods (HPFs) can meet the criteria to be labeled as addictive substances using the standards set for tobacco products. The addictive potential of HPFs may be a key factor contributing to the high public health costs associated with a food environment dominated by cheap, accessible and heavily marketed HPFs.
Article
Full-text available
Here, we tested the hypothesis that sucralose differentially affects metabolic responses to labeled oral glucose tolerance tests (OGTTs) in participants with normal weight and obesity. Participants (10 with normal weight and 11 with obesity) without diabetes underwent three dual-tracer OGTTs preceded, in a randomized order, by consuming sucralose or water, or by tasting and expectorating sucralose (e.g., sham-fed; sweetness control). Indices of β-cell function and insulin sensitivity (SI) were estimated using oral minimal models of glucose, insulin, and C-peptide kinetics. Compared with water, sucralose ingested (but not sham-fed) resulted in a 30 ± 10% increased glucose area under the curve in both weight groups. In contrast, the insulin response to sucralose ingestion differed depending on the presence of obesity: decreased within 20–40 min of the OGTT in normal-weight participants but increased within 90–120 min in participants with obesity. Sham-fed sucralose similarly decreased insulin concentrations within 60 min of the OGTT in both weight groups. Sucralose ingested (but not sham-fed) increased SI in normal-weight participants by 52 ± 20% but did not affect SI in participants with obesity. Sucralose did not affect glucose rates of appearance or β-cell function in either weight group. Our data underscore a physiological role for taste perception in postprandial glucose responses, suggesting sweeteners should be consumed in moderation.
Article
Full-text available
Introduction: The brain plays an important regulatory role in directing energy homeostasis and eating behavior. The increased ingestion of sugars and sweeteners over the last decades makes investigating the effects of these substances on the regulatory function of the brain of particular interest. We investigated whole brain functional response to the ingestion of nutrient shakes sweetened with either the nutritive natural sugars glucose and fructose, the low- nutritive natural sugar replacement allulose or the non-nutritive artificial sweetener sucralose. Methods: Twenty healthy, normal weight, adult males underwent functional MRI on four separate visits. In a double-blind randomized study setup, participants received shakes sweetened with glucose, fructose, allulose or sucralose. Resting state functional MRI was performed before and after ingestion. Changes in Blood Oxygen Level Dependent (BOLD) signal, functional network connectivity and voxel based connectivity by Eigenvector Centrality Mapping (ECM) were measured. Results: Glucose and fructose led to significant decreased BOLD signal in the cingulate cortex, insula and the basal ganglia. Glucose led to a significant increase in eigen vector centrality throughout the brain and a significant decrease in eigen vector centrality in the midbrain. Sucralose and allulose had no effect on BOLD signal or network connectivity but sucralose did lead to a significant increase in eigen vector centrality values in the cingulate cortex, central gyri and temporal lobe. Discussion: Taken together our findings show that even in a shake containing fat and protein, the type of sweetener can affect brain responses and might thus affect reward and satiety responses and feeding behavior. The sweet taste without the corresponding energy content of the non-nutritive sweeteners appeared to have only small effects on the brain. Indicating that the while ingestion of nutritive sugars could have a strong effect on feeding behavior, both in a satiety aspect as well as rewarding aspects, non-nutritive sweeteners appear to not have these effects. Trial registration: This study is registered at clinicaltrials.gov under number NCT02745730.
Article
Full-text available
Background: Recently, the absence of metabolic effects from nonnutritive sweeteners has been questioned. Objective: The aim of this study was to evaluate the effects of sucralose consumption on glucose metabolism variables. Design: We performed a randomized controlled trial involving healthy subjects without comorbidities and with a low habitual consumption of nonnutritive sweeteners (n = 33/group). Methods: The intervention consisted of sucralose consumption as 15% of Acceptable Daily Intake every day for 14 d using commercial sachets. The control group followed the same procedures without any intervention. The glucose metabolism variables (insulin sensitivity, acute insulin response to glucose, disposition index, and glucose effectiveness) were evaluated by using a 3-h modified intravenous-glucose-tolerance test before and after the intervention period. Results: Individuals assigned to sucralose consumption showed a significant decrease in insulin sensitivity with a median (IQR) percentage change of -17.7% (-29.3% to -1.0%) in comparison to -2.8% (-30.7% to 40.6%) in the control group (P= 0.04). An increased acute insulin response to glucose from 577 mU · L-1· min (350-1040 mU · L-1· min) to 671 mU · L-1· min (376-1010 mU · L-1· min) (P = 0.04) was observed in the sucralose group for participants with adequate adherence. Conclusions: Sucralose may have effects on glucose metabolism, and our study complements findings previously reported in other trials. Further studies are needed to confirm the decrease in insulin sensitivity and to explore the mechanisms for these metabolic alterations. This trial was registered at www.clinicaltrials.gov as NCT02589002.
Article
Full-text available
Objective: To estimate the prevalence of diagnosed total diabetes, type 1 diabetes, and type 2 diabetes in the US general population and the proportions of each among US adults with a diagnosis of diabetes. Design: Nationwide, population based, cross sectional survey. Setting: National Health Interview Survey, 2016 and 2017. Participants: Adults aged 20 years or older (n=58 186), as a nationally representative sample of the civilian, non-institutionalized US population. Main outcome measures: Prevalence of diagnosed diabetes, type 1 diabetes, and type 2 diabetes in the US general population, and the proportions of each subtype in participants with a diagnosis of diabetes. Results: Among the 58 186 included adults, 6317 had received a diagnosis of diabetes. The weighted prevalence of diagnosed diabetes, type 1 diabetes, and type 2 diabetes among US adults was 9.7% (95% confidence interval 9.4% to 10.0%), 0.5% (0.5% to 0.6%), and 8.5% (8.2% to 8.8%), respectively. Type 1 diabetes was more prevalent among adults with lower education level, and type 2 diabetes was more prevalent among older adults, men, and those with lower educational level, lower family income level, and higher body mass index (BMI). Among adults with a diagnosis of diabetes, the weighted percentage of type 1 and type 2 diabetes was 5.6% (4.9% to 6.4%) and 91.2% (90.4% to 92.1%), respectively. The percentage of type 1 diabetes was higher among younger adults (age 20-44 years), non-Hispanic white people, those with higher education level, and those with lower BMI, whereas the percentage of type 2 diabetes was higher among older adults (age ≥65 years), non-Hispanic Asians, those with lower education level, and those with higher BMI. Conclusion: This study provided benchmark estimates on the national prevalence of diagnosed type 1 diabetes (0.5%) and type 2 diabetes (8.5%) among US adults. Among US adults with diagnosed diabetes, type 1 and type 2 diabetes accounted for 5.6% and 91.2%, respectively.
Article
Background: This study aims to determine the effect of pure forms of sucralose and aspartame, in doses reflective of common consumption, on glucose metabolism. Methods: Healthy participants consumed pure forms of a non-nutritive sweetener (NNS) mixed with water that were standardized to doses of 14% (0.425 g) of the acceptable daily intake (ADI) for aspartame and 20% (0.136 g) of the ADI for sucralose every day for two weeks. Blood samples were collected and analysed for glucose, insulin, active glucagon-like peptide-1 (GLP-1), and leptin. Results: Seventeen participants (10 females and 7 males; age 24 ± 6.8 years; BMI 22.9 ± 2.5 kg/m2) participated in the study. The total area under the curve (AUC) values of glucose, insulin, active GLP-1 and leptin were similar for the aspartame and sucralose treatment groups compared to the baseline values in healthy participants. There was no change in insulin sensitivity after NNS treatment compared to the baseline values. Conclusions: These findings suggest that daily repeated consumption of pure sucralose or aspartame for 2 weeks had no effect on glucose metabolism among normoglycaemic adults. However, these results need to be tested in studies with longer durations. Novelty: • Daily consumption of pure aspartame or sucralose for 2 weeks had no effect on glucose metabolism. • Daily consumption of pure aspartame or sucralose for 2 weeks had no effect on insulin sensitivity among healthy adults.
Article
Objective: Exposure in utero to maternal obesity or gestational diabetes mellitus (GDM) is linked to a high risk for obesity in offspring. Animal studies suggest that these exposures disrupt the development of the hypothalamus, a brain region that regulates body weight, predisposing offspring to develop obesity. This study tested the hypothesis in humans that in utero exposure to maternal obesity and/or GDM is associated with alterations in the hypothalamic response to glucose and the altered hypothalamic response would predict greater increases in child adiposity 1 year later. Research design and methods: Participants were 91 children aged 7-11 years with and without in utero exposure to GDM. Maternal prepregnancy BMI and GDM exposures were determined from electronic medical records. Arterial spin labeling MRI was used to determine the child's hypothalamic blood flow response to oral glucose. Anthropometric measures were acquired in all children at their initial visit and again 1 year later in a subset of 44 children. Results: Children exposed to GDM diagnosed at ≤26 weeks' gestation had increased hypothalamic blood flow (a marker of hypothalamic activation) in response to glucose when compared with unexposed children, and results remained after adjustments for child age, sex, BMI, and maternal prepregnancy BMI. Maternal prepregnancy BMI was positively associated with the child's hypothalamic response to glucose. Greater hypothalamic response to glucose predicted greater increases in child's BMI 1 year later. Conclusions: Increased glucose-linked hypothalamic activation during childhood represents a possible mechanism by which exposure to maternal metabolic disorders during fetal development increases future risk for obesity.
Article
Background: Low-calorie sweeteners (LCSs) provide sweetness with little or no energy. However, each LCS's unique chemical structure has potential to elicit different sensory, physiological, and behavioral responses that affect body weight. Objective: The purpose of this trial was to compare the effects of consumption of 4 LCSs and sucrose on body weight, ingestive behaviors, and glucose tolerance over a 12-wk intervention in adults (18-60 y old) with overweight or obesity (body mass index 25-40 kg/m2). Methods: In a parallel-arm design, 154 participants were randomly assigned to consume 1.25-1.75 L of beverage sweetened with sucrose (n = 39), aspartame (n = 30), saccharin (n = 29), sucralose (n = 28), or rebaudioside A (rebA) (n = 28) daily for 12 wk. The beverages contained 400-560 kcal/d (sucrose treatments) or <5 kcal/d (LCS treatments). Anthropometric indexes, energy intake, energy expenditure, appetite, and glucose tolerance were measured at baseline. Body weight was measured every 2 wk with energy intake, expenditure, and appetite assessed every 4 wk. Twenty-four-hour urine collections were completed every 4 wk to determine study compliance via para-aminobenzoic acid excretion. Results: Of the participants enrolled in the trial, 123 completed the 12-wk intervention. Sucrose and saccharin consumption led to increased body weight across the 12-wk intervention (Δweight = +1.85 ± 0.36 kg and +1.18 ± 0.36 kg, respectively; P ≤ 0.02) and did not differ from each other. There was no significant change in body weight with consumption of the other LCS treatments compared with baseline, but change in body weight for sucralose was negative and significantly lower compared with all other LCSs at week 12 (weight difference ≥ 1.37 ± 0.52 kg, P ≤ 0.008). Energy intake decreased with sucralose consumption (P = 0.02) and ingestive frequency was lower for sucralose than for saccharin (P = 0.045). Glucose tolerance was not significantly affected by any of the sweetener treatments. Conclusions: Sucrose and saccharin consumption significantly increase body weight compared with aspartame, rebA, and sucralose, whereas weight change was directionally negative and lower for sucralose compared with saccharin, aspartame, and rebA consumption. LCSs should be categorized as distinct entities because of their differing effects on body weight. This trial was registered at clinicaltrials.gov as NCT02928653.
Article
Objective To examine associations between consumption of low‐calorie sweetened beverages (LCSBs), sugar, and total energy intake in children in the United States. Methods We used 24‐hour dietary recalls from 7026 children enrolled in the National Health and Nutrition Examination Survey (NHANES) 2011 to 2016 to assess energy and macronutrient intake among LCSB (≥4 oz LCSB, <4 oz SB), SB (≥4 oz SB, <4 oz LCSB), and LCSB + SB consumers (≥4 oz each) compared with water consumers (≥4 oz water, <4 oz LCSB and SBs). Sample weights and complex survey procedures were used for all analyses. Results Adjusting for body mass index (BMI) percentile, LCSB, SB, and LCSB + SB consumption was associated with 196, 312, and 450 more total calories and 15, 39, and 46 more calories from added sugar, respectively, compared with water consumers (P < .05 for all pairwise comparisons). No differences in energy intake were observed between LCSB and SB consumers. Conclusions These findings challenge the utility of LCSB for weight management in children and adolescents.
Article
In the United States, 32% of beverages consumed by adults and 19% of beverages consumed by children in 2007 to 2010 contained low-calorie sweeteners (LCSs). Among all foods and beverages containing LCSs, beverages represent the largest proportion of LCS consumption worldwide. The term LCS includes the 6 high-intensity sweeteners currently approved by the US Food and Drug Administration and 2 additional high-intensity sweeteners for which the US Food and Drug Administration has issued no objection letters. Because of a lack of data on specific LCSs, this advisory does not distinguish among these LCSs. Furthermore, the advisory does not address foods sweetened with LCSs. This advisory reviews evidence from observational studies and clinical trials assessing the cardiometabolic outcomes of LCS beverages. It summarizes the positions of government agencies and other health organizations on LCS beverages and identifies research needs on the effects of LCS beverages on energy balance and cardiometabolic health. The use of LCS beverages may be an effective strategy to help control energy intake and promote weight loss. Nonetheless, there is a dearth of evidence on the potential adverse effects of LCS beverages relative to potential benefits. On the basis of the available evidence, the writing group concluded that, at this time, it is prudent to advise against prolonged consumption of LCS beverages by children. (Although water is the optimal beverage choice, children with diabetes mellitus who consume a balanced diet and closely monitor their blood glucose may be able to prevent excessive glucose excursions by substituting LCS beverages for sugar-sweetened beverages [SSBs] when needed.) For adults who are habitually high consumers of SSBs, the writing group concluded that LCS beverages may be a useful replacement strategy to reduce intake of SSBs. This approach may be particularly helpful for persons who are habituated to a sweet-tasting beverage and for whom water, at least initially, is an undesirable option. Encouragingly, self-reported consumption of both SSBs and LCS beverages has been declining in the United States, suggesting that it is feasible to reduce SSB intake without necessarily substituting LCS beverages for SSBs. Thus, the use of other alternatives to SSBs, with a focus on water (plain, carbonated, and unsweetened flavored), should be encouraged.
Article
Objective: The brain is essential in regulating intake of food and beverages by balancing energy homeostasis, which is regulated by the hypothalamus, with reward perception, which is regulated by the ventral tegmental area (VTA). The aim of this study was to investigate the effects of ingestion of glucose, fructose, sucrose, and sucralose (a non-caloric artificial sweetener) on the magnitude and trajectory of the hypothalamic and the VTA blood oxygen level-dependent (BOLD) responses. Method: In five visits, 16 healthy men between 18 to 25 y of age with a body mass index between 20 and 23 kg/m2 drank five interventions in a randomized order while a functional magnetic resonance imaging scan was taken. The interventions consisted of 50 g of glucose, fructose, or sucrose, or 0.33 g of sucralose dissolved in 300 mL tap water. The control condition consisted of 300 mL of plain tap water. BOLD signals were determined in the hypothalamus and the VTA within a manually drawn region of interest. Differences in changes in BOLD signal between stimuli were analyzed using mixed models. Results: Compared with the control condition, a decrease in BOLD signal in the hypothalamus was found after ingestion of glucose (P = 0.0003), and a lesser but delayed BOLD response was found after ingestion of sucrose (P = 0.006) and fructose (P = 0.003). Sucralose led to a smaller and transient response from the hypothalamus (P = 0.026). In the VTA, sucralose led to a very similar response to the water control condition, leading to an increase in VTA BOLD activity that continued over the measured time period. The natural sugars appeared to only lead to a transient increase in VTA activity. Conclusions: Glucose induces a deactivation in the hypothalamus immediately after ingestion and continued over the next 12 min, which is correlated with satiety signaling by the brain. Fructose and sucrose are both associated with a delayed and lesser response from the hypothalamus, likely because the sugars first have to be metabolized by the body. Sucralose leads to the smallest and most transient decrease in BOLD in the hypothalamus and leads to a similar response as plain water in the VTA, which indicates that sucralose might not have a similar satiating effect on the brain as the natural sugars.