ArticlePDF Available

Long-Term Overconsumption of Sugar Starting at Adolescence Produces Persistent Hyperactivity and Neurocognitive Deficits in Adulthood

Frontiers
Frontiers in Neuroscience
Authors:

Abstract and Figures

Sugar has become embedded in modern food and beverages. This has led to overconsumption of sugar in children, adolescents, and adults, with more than 60 countries consuming more than four times (>100 g/person/day) the WHO recommendations (25 g/person/day). Recent evidence suggests that obesity and impulsivity from poor dietary habits leads to further overconsumption of processed food and beverages. The long-term effects on cognitive processes and hyperactivity from sugar overconsumption, beginning at adolescence are not known. Using a well-validated mouse model of sugar consumption, we found that long-term sugar consumption, at a level that significantly augments weight gain, elicits an abnormal hyperlocomotor response to novelty and alters both episodic and spatial memory. Our results are similar to those reported in attention deficit and hyperactivity disorders. The deficits in hippocampal-dependent learning and memory were accompanied by altered hippocampal neurogenesis, with an overall decrease in the proliferation and differentiation of newborn neurons within the dentate gyrus. This suggests that long-term overconsumption of sugar, as that which occurs in the Western Diet might contribute to an increased risk of developing persistent hyperactivity and neurocognitive deficits in adulthood.
Long-term sucrose consumption increases mouse weights without altering emotional behavior. (A) Experimental design of the anxiety-related, depression-related, general locomotor activity and impulse control behavioral testing in elevated-plus-maze (EPM), novelty suppressed feeding (NSF), marble burying (MB), open field (OF) and forced swimming test (FS). Animals consumed 25% sucrose for 5 weeks prior to each behavioral testing and continued to be exposed to sucrose for a total of 12 weeks. Behavioral tests were initially conducted 2 weeks (2 wk) then continued every 5 days (5d) after 24 h of sugar withdrawal. Animals were assigned into two groups: sugar-withdrawn animals and water control. (B) Mice exhibit stable levels of long-term sucrose intake of 82.2 ± 0.9 g/kg indicated by the red line. (C) There was a significant difference in the weights of animals consuming sucrose, compared to water control, starting after 4 weeks, and continuing throughout the 12 weeks. Data are presented as mean ± SEM; n = 8 mice/group, t-test, ***p < 0.001. (D–F) Long-term sucrose consumption does not alter anxiety-related behavior as seen by no change in the number of entries in the open-arm of the EPM (D), number of marbles buried (E) and number of entries in the center in the open field (F) compared to water controls. Long-term sucrose consumption does not induce depressive-like symptoms as seen by no differences in the immobility time in the forced swimming test (G) compared to water controls. Data are presented as mean ± SEM; n = 8 mice/group.
… 
Long-term sucrose consumption reduces the early phases of hippocampal neurogenesis. (A) Stages of neurogenesis in the dentate gyrus of the hippocampus. In stage 1 (proliferation phase; in yellow), the newly generated cells express glial fibrillary acidic protein (GFAP) and are putative progenitor/stem cells located in the subgranular zone (SGZ). The cells in stage 2 (differentiation phase; in green) will lose their GFAP and start expressing Nestin. This determines their fate in the neuronal lineage. Stage 1 progenitors not only give rise to newborn neurons, they can also turn into glial cells. Glial cells [astrocyte and oligodendrocyte progenitor cell (OPC)] can convert back into newborn neurons here in stage 2 and vice-versa. In stage 3, the immature neurons express doublecortin (DCX; in cobalt blue) and have started to migrate into the granule cell layer (GCL) of the dentate gyrus. As the neuron matures, it will send its dendrites toward the molecular layer (ML) of the dentate gyrus and extend their axonal projections toward the hippocampal CA3 pyramidal cell layer and will start losing their DCX and start expressing postmitotic neuronal marker NeuN and calretinin (stage 4; magenta). As the neuron establishes synaptic contacts from the entorhinal cortex and it sends output to the CA3 and hilus regions of the hippocampus the neuron is classified as in stage 5 (in cherry red). Stage 5 neurons start expressing calbindin and continue expressing NeuN. Unlike the astrocytes and oligodendrocytes that are derived from the neuroectoderm, microglia (in cyan blue) are neuroglia derived by embryonic mesoderm. Abbreviations used: GFAP, glial fibrillary acidic protein; OPC, oligodendrocyte progenitor cell; Olig2, oligodendrocyte lineage transcription factor 2; IBA-1, ionized calcium binding adaptor molecule 1; DCX, doublecortin; NeuN, Fox-3, Rbfox3, or Hexaribonucleotide Binding Protein-3; SGZ, subgranular zone; GCL, granule cell layer; ML, molecular layer [original drawing, created using Biorender, adapted from Lucassen et al. (2010)]. (B) Long-term sugar consumption reduced the density of EdU positive cells (green) in the dentate gyrus of the hippocampus. (C) There was a reduction in the density of EdU⁺ (green)/GFAP⁺ (red) immunoreactive cells indicating a reduction in stage 1 (putative stem cells) neurogenesis. (D) A reduction was also observed in the number of EdU⁺ (green)/Nestin⁺ (red)-immunoreactive cells indicating a reduction in stage 2 (neuronal progenitors). All images are colocalized with DAPI (blue). Data are presented as mean ± SEM; n = 8 mice/group. *p < 0.05. Representative image scale bar is 100 μm and close up representative image scale bar is 10 μm.
… 
This content is subject to copyright.
fnins-15-670430 June 4, 2021 Time: 14:29 # 1
ORIGINAL RESEARCH
published: 07 June 2021
doi: 10.3389/fnins.2021.670430
Edited by:
Vittorio Calabrese,
University of Catania, Italy
Reviewed by:
Nafisa M. Jadavji,
Midwestern University, United States
Yinghua Yu,
Xuzhou Medical University, China
*Correspondence:
Arnauld Belmer
arnauld.belmer@qut.edu.au
Selena E. Bartlett
selena.bartlett@qut.edu.au
These authors share senior
authorship
Specialty section:
This article was submitted to
Neuroenergetics, Nutrition and Brain
Health,
a section of the journal
Frontiers in Neuroscience
Received: 22 February 2021
Accepted: 22 April 2021
Published: 07 June 2021
Citation:
Beecher K, Alvarez Cooper I,
Wang J, Walters SB, Chehrehasa F,
Bartlett SE and Belmer A (2021)
Long-Term Overconsumption of
Sugar Starting at Adolescence
Produces Persistent Hyperactivity and
Neurocognitive Deficits in Adulthood.
Front. Neurosci. 15:670430.
doi: 10.3389/fnins.2021.670430
Long-Term Overconsumption of
Sugar Starting at Adolescence
Produces Persistent Hyperactivity
and Neurocognitive Deficits in
Adulthood
Kate Beecher1, Ignatius Alvarez Cooper2, Joshua Wang1, Shaun B. Walters3,
Fatemeh Chehrehasa2, Selena E. Bartlett1*and Arnauld Belmer1*
1Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Translational Research Institute, Faculty of
Health, Queensland University of Technology, Brisbane, QLD, Australia, 2Addiction Neuroscience and Obesity Laboratory,
School of Biomedical Sciences, Translational Research Institute, Faculty of Health, Queensland University of Technology,
Brisbane, QLD, Australia, 3School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
Sugar has become embedded in modern food and beverages. This has led to
overconsumption of sugar in children, adolescents, and adults, with more than
60 countries consuming more than four times (>100 g/person/day) the WHO
recommendations (25 g/person/day). Recent evidence suggests that obesity and
impulsivity from poor dietary habits leads to further overconsumption of processed
food and beverages. The long-term effects on cognitive processes and hyperactivity
from sugar overconsumption, beginning at adolescence are not known. Using a
well-validated mouse model of sugar consumption, we found that long-term sugar
consumption, at a level that significantly augments weight gain, elicits an abnormal
hyperlocomotor response to novelty and alters both episodic and spatial memory. Our
results are similar to those reported in attention deficit and hyperactivity disorders.
The deficits in hippocampal-dependent learning and memory were accompanied by
altered hippocampal neurogenesis, with an overall decrease in the proliferation and
differentiation of newborn neurons within the dentate gyrus. This suggests that long-
term overconsumption of sugar, as that which occurs in the Western Diet might
contribute to an increased risk of developing persistent hyperactivity and neurocognitive
deficits in adulthood.
Keywords: sucrose, hyperactivity, neurocognitive deficits, neurogenesis, adulthood
INTRODUCTION
The concept of “sugar addiction” and the classification of sugar as a substance of abuse are still
debated. There is, however, increasing evidence of overlap in the brain circuitry and molecular
signaling pathways involved in sugar consumption and drug abuse (for recent review see Jacques
et al., 2019). Humans consume sugar and food to regulate homeostatic energy balance, but also
for pleasure and comfort. This hedonistic desire for palatable food is reward-driven and overeating
may result in maladaptive/negative neuroplasticity that overrides homeostatic regulation (Kenny,
2011). In humans, sugar and sweetness can induce dopamine release, reward and craving that are
Frontiers in Neuroscience | www.frontiersin.org 1June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 2
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
comparable in magnitude to those induced by addictive drugs,
suggesting that sugar changes brain reward signaling and
circuitry similar to other drugs of abuse (Avena and Hoebel,
2003b;Rada et al., 2005;Lenoir et al., 2007;Klenowski et al., 2016;
Shariff et al., 2016, 2017).
High sugar and/or high fat diets have been shown to
precipitate addiction-like psychiatric phenotypes in a number of
rodent studies (Avena et al., 2009;Avena, 2010;Criscitelli and
Avena, 2016). In rats, intermittent consumption of 10% (w/v)
sucrose or 25% (w/v) glucose solution) elicits hallmark signs
of addictive behavior such as binging, tolerance, craving (Rada
et al., 2005), cross-sensitization (Avena and Hoebel, 2003b) and
symptoms of withdrawal (Colantuoni et al., 2002;Avena et al.,
2008) such as anxiety-(Colantuoni et al., 2002;Avena et al., 2008;
Parylak et al., 2012;Eudave et al., 2018;Gueye et al., 2018;Xu and
Reichelt, 2018) and depressive-like behaviors (Vollmayr et al.,
2004;Iemolo et al., 2012;Harrell et al., 2015;Santos et al., 2018).
In addition, sugar consumption has been shown to increase
reward seeking, impulsivity to feed and compulsivity in rats
willing to endure noxious stimuli such as extreme cold, heat and
foot-shock to procure sugar and highly palatable foods (Cabanac
and Johnson, 1983;Avena et al., 2005;Foo and Mason, 2005;
Oswald et al., 2011). Interestingly, rats are also more resilient
to foot shock punishments when seeking for palatable food
compared to methamphetamine (Krasnova et al., 2014).
Increasing evidence shows that unrestricted consumption
of high-sugar food and beverages within the Western Diet
might be linked to the increased obesity epidemic (Stanhope,
2016;Johnson et al., 2017;Freeman et al., 2018;Yoshida and
Simoes, 2018;Sigala and Stanhope, 2021). A strong association
between attention-deficits/hyperactivity disorders (ADHD) and
overweight/obesity have further been revealed (Altfas, 2002;
Strimas et al., 2008;Cortese et al., 2016;Cortese, 2019).
Taken together, these data suggest that sugar-induced obesity
may participate to the developing pathogenesis of ADHD-
like symptoms in western countries. In children, high sugar
consumption correlates with hyperactivity (Kim and Chang,
2011) and in adults, with inattention and impulsivity (Li et al.,
2020). However, some inconsistencies remain regarding the
potential correlation (Yu et al., 2016;Farsad-Naeimi et al., 2020)
or not (Del-Ponte et al., 2019) with ADHD (Johnson et al.,
2011;Paglia, 2019). In rodents, high-sucrose consumption also
impairs neurocognitive functions such as spatial learning, object
recognition, behavioral inhibition and fear-memory (Kendig,
2014;Reichelt et al., 2015;Kruse et al., 2019;Spoelma and Boakes,
2021). Interestingly, high sucrose intake during pregnancy
elicits ADHD-like behavioral phenotypes in mice offspring, with
increased locomotor activity, reduced attention/learning and
impulsivity (Choi et al., 2015). Anxiety, depression, and cognitive
deficits are strongly associated with impaired hippocampal
neurogenesis in animal models, although evidence for a causative
relationship is often lacking. Indeed, anxiety and spatial memory
deficits elicited by long-term consumption of sucrose are
accompanied by alterations in hippocampal neurogenesis and
physiology (Molteni et al., 2002;Stranahan et al., 2008;van
der Borght et al., 2011;Lemos et al., 2016;Reichelt et al.,
2016). While drugs of abuse such as ethanol are known to
negatively affect neurogenesis, the effect of high levels of sugar
consumption requires further characterization since link between
neurogenesis to anxiety and depression has not been fully
explored (Xu and Reichelt, 2018).
Sucrose became embedded in modern food and beverages, and
the aforementioned studies suggest that sugar overconsumption
satisfies all criteria for the classification of sugar as a drug of
abuse, with its chronic abuse proposed to produce overweight,
locomotor, emotional and cognitive impairments. However, it
remains unclear whether a lifetime of chronic overconsumption
of sucrose, starting at adolescence, affects locomotor behavior,
emotions and cognition through adulthood. Therefore, we used
a mouse model of long-term intake of sucrose to determine
the effects on locomotion, anxiety, memory, and hippocampal
neurogenesis. Our results show for the first time that long-
term consumption of sucrose leads to significant weight gain
and produces persistent hyperactivity and learning impairments,
correlated to reduced hippocampal neurogenesis in adult mice.
These results suggest that long-term sugar intake in the Western
Diet might play a role in the pathogenesis of attention deficits and
hyperactivity-related disorders.
MATERIALS AND METHODS
Animals and Housing
Five-week-old male C57BL/6J mice (ARC, WA, Australia) were
individually housed under reverse-light cycle conditions (lights
off at 9:00 am) in a climate-controlled room with ad libitum
access to food (standard mouse chow) and water. Following
one week of habituation to the housing conditions, mice were
offered sucrose or water during the drinking sessions. All
procedures were approved by The University of Queensland
and The Queensland University of Technology Animal Ethics
Committees under approval QUT/053/18 and complied with
the policies and regulations regarding animal experimentation
and other ethical matters, in accordance with the Queensland
Government Animal Research Act 2001, associated Animal
Care and Protection Regulations (2002 and 2008), as well as
the Australian Code for the Care and Use of Animals for
Scientific Purposes, 8th Edition (National Health and Medical
Research Council, 2013).
Sucrose Consumption
All mice had food and acidified-filtered water available at all
times. The sucrose solution was freshly prepared weekly and was
presented in 50 ml plastic falcon tubes fitted with rubber stoppers
and a 6.35 cm stainless-steel sipper tube with double ball bearings.
Mouse weights were measured daily to calculate the adjusted
g/kg intake of sucrose. Two groups of mice (n= 23 mice/group)
had ad libitum access to 25% sucrose or water for 12 weeks.
Briefly, mice were given access to one bottle of 25% (w/v) sucrose
solution and one bottle of water available at all times, or two
bottles of water (controls) available at all times. Sucrose and
water containing bottles were weighed daily. Two other groups
of mice (n= 23 mice/group) were trained in the in a restricted
access model of sugar consumption (Drinking-In-the-Dark) for
Frontiers in Neuroscience | www.frontiersin.org 2June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 3
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
12 weeks as previously described (Rhodes et al., 2005;Patkar et al.,
2017, 2019;Belmer et al., 2018). Briefly, mice were given access
to one bottle of 25% (w/v) sucrose for a 2 h period Monday to
Friday. Drinking sessions started 3 h into the dark cycle. Sucrose
containing bottles were weighed prior to presentation, as well as
2 h after presentation.
Behavioral Testing
Following 5 weeks of sucrose consumption, one set of animals
undertook five behavioral tests were conducted over 6 weeks
(n= 8 per group; Figure 1A). These behavioral tests were
conducted during dark cycle and were withdrawn from sucrose
for 24 h. Elevated-plus-maze (EPM), a behavioral test used to
observe anxiety-related behavior, was performed in an apparatus
comprising of four arms, 2 open arms and 2 closed arms
(35 cm ×5 cm), elevated 50 cm above the floor. The closed
arms were fenced with 40 cm high walls. The experiment went
for 5 min, with initial mouse placement in the center, facing the
open arm. The number of entries and time spent in each arm
was recorded using ANY-maze tracking software (Stoelting, IL,
United States) (Walf and Frye, 2007;Belmer et al., 2018).
Novelty suppressed feeding (NSF) test measures the novelty
induced anxiety/impulse control by assessing the latency to
approach and eat familiar food in a novel/aversive environment.
Animals were food-deprived for 16 h before being placed in an
open area (L = 40 cm, W = 36 cm, H = 18 cm) with new bedding
and a piece of familiar chow in the middle of the area. Latency to
feed was measured in seconds before the animal eats the food, by
two experimenters blind to the diet (Bevilacqua et al., 2010).
Marble burying (MB) is used to test anxiety and obsessive-
compulsive disorder-like behavior. MB was performed in novel
individual plastic cages (21 cm ×38 cm ×14 cm) containing
5 cm thick sawdust bedding. Ten glass marbles (diameter 10–
12 mm) were evenly spaced in 2 rows of 5 marbles on the bedding.
After 20 min, the number of unburied marbles was averaged
from counting by two experimenters. A marble covered at least
two-third (2/3) of its size by saw dust will be considered as
“buried”(Deacon, 2006;Belmer et al., 2018).
Open-field (OF) test is used to measure exploratory behavior,
general locomotor behavior and anxiety. OF was performed in
an open arena of 30 cm ×30 cm ×40 cm. The floor was
divided into 16 equal squares (7 cm ×7 cm) and a central
region of 10 cm ×10 cm was considered as the center, outside
this central region was considered as the periphery. Mice were
initially placed in one corner, and allowed to explore freely for
10 min. The number of entries and the time spent in the center or
periphery were recorded using the ANY-maze software (Bailey
and Crawley, 2009;Belmer et al., 2018). General locomotor
activity was assessed using the open field apparatus and recorded
using ANY-maze software.
The forced swimming test (FS) is commonly used to test the
efficacy of antidepressants and by extension, to assess depressive-
like behaviors. The FS test was conducted in a cylindrical glass
container measuring 50 cm in height and a diameter of 20 cm.
The immobility time was recorded using ANY-maze tracking
software (Yankelevitch-Yahav et al., 2015).
Memory Assessment
Using a separate group of animals on the same drinking protocol,
two recognition/spatial memory tests were conducted (n= 15 per
group, Figure 3A). After 8 weeks of 25% sucrose consumption,
recognition (episodic) memory was assessed using novel object
recognition (NOR) test. NOR protocol ran over 4 days and was
performed in the open field apparatus. On the first day, mice were
habituated to the open field apparatus for 10 min. The second
and third day, mice were presented with two identical objects for
10 min. No animals were excluded from analysis based on their
object exploration times. On the last day, under 24 h withdrawal
to 25% sucrose solution, one of the two familiar objects was
replaced with a novel object (Figure 3D) and the interactions
with the objects, including metrics such as object exploration time
and latency to reach the novel and familiar objects were recorded
on ANY-maze software (Leger et al., 2013).
The same animals continued to drink 25% sucrose for 2 more
weeks (Week 10) before spatial recognition was measured using
the Morris Water Maze. This test evaluates the animal’s ability to
escape a stressful situation in a large pool of water. The heated
pool (22C; 150 cm diameter) was divided into 4 quadrants
with a designated visual cue in each quadrant (Figure 3G).
We used a 4-day protocol (Flores-Ramirez et al., 2019) starting
with a pre-training/habituation day where the location of the
escape platform was introduced with the platform being visible
1 cm above the water surface. Each animal was placed on the
platform for 10 s before being released into the water facing the
platform, less than 10 cm away. Once the platform was reached,
place mouse onto the platform for 10 s then released again at a
greater distance (20 cm). This was repeated for a third time with
the release greater again (30 cm). If platform was not reached,
experimenter gently guided the mouse to the platform and the
animal was released in the water again until the mouse swam
to the platform unaided. On day 2 (training day), the platform
was partially covered by water and odorless non-toxic paint was
added so the platform was not visible. Each animal had 8 trials
of 60 s duration to swim to the platform, being released from
each quadrant while facing the wall twice in a random order.
Between trials the mouse had a 45 s break before the next trial
commenced in a warmed holding cage. On day 3 (testing day),
performed under 24 h withdrawal from 25% sucrose solution,
each animal was released from the furthest quadrant to reach
the platform and were removed from the pool if they did not
reach the platform in 60 s. The time spent to reach platform was
recorded manually having up to 1 min to reach the platform.
Testing videos was recorded using ANY-maze. Day 4 involved
probing the anima’s memory, with the platform being removed
and the amount of time spent in the quadrant or in platform area
recorded using ANY-maze. Heating pads and lamps were used
and readily available across all four days.
Neurogenesis
Following 12 weeks of sucrose drinking, a total of three
intraperitoneal injections of the cell proliferation marker, 5-
ethynyl-20-deoxyuridine (EdU; 50 mg/kg) were administered
over 2 weeks (days 0, 7, and 15) as previously described
Frontiers in Neuroscience | www.frontiersin.org 3June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 4
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
(Belmer et al., 2018;Patkar et al., 2019). This dose has been
reported to label all actively dividing precursors in the mouse
subgranular zone (Mandyam et al., 2007). One week after
the last EdU injection, animals were deeply anesthetized with
sodium pentobarbital (100 mg/kg, Lethobarb, Virbac, Australia)
and transcardially perfused with 4% paraformaldehyde. Brains
were harvested and postfixed in the same fixative overnight
at 4C. Thirty micron-thick coronal vibratome sections were
collected and kept floating in ice-cold 0.1M phosphate buffer
saline (PBS). Sections containing the hippocampus were selected
for immunohistochemistry. The sections were permeabilized
in 1% Triton X100, 0.1% Tween20 in PBS for 1 h at room
temperature and incubated with EdU Click-iTTM EdU Alexa
FluorTM 488 Imaging Kit as per supplier recommendation
(Thermo-Fisher Scientific, C10637). When required, antigen
retrieval (0.05% Tween-20 in 10 mM sodium citrate, pH 6, 5–
15 min at 80C) was performed. After three washes in PBS,
sections were incubated in blocking solution (2% normal goat
serum, 0.3% Triton X100 and 0.05% Tween-20) for 1 h at
room temperature. The sections were then probed for markers
of each stage of neurogenesis (Figure 3A) and incubated with
primary antibodies, diluted in blocking solution, for 24 h at
room temperature, washed 3 times in blocking solution and
then incubated with corresponding secondary antibodies, for
2 h at room temperature (Supplementary Table 1). When
biotinylated secondaries were used, sections were incubated in
streptavidin-CY3 for 30 min at room temperature. Sections
were mounted in Prolong gold antifade mountant with DAPI
(Thermofisher Scientific).
Imaging and Analysis
Four coronal sections of whole dentate gyri per animals
were imaged on the Leica DMi8 SP8 Laser Point Scanning
confocal microscope using a 40 ×objective (NA 0.85), x0.5
numerical zoom and 0.5 z-step. Consecutive sections were
used across all staining groups. Images were deconvolved
using Huygens professional v16.10 (Scientific Volume
Imaging, Netherlands) and converted in.tif for subsequent
quantification in Neurolucida 360 (MBF Bioscience). Early stages
of neurogenesis were counted: stage 1: EdU+/GFAP+/Nestin;
stage 2: EdU+/Nestin+/GFAPand stage 3: EdU+/DCX+as
well as glial cell types: astrocytes (EdU+/GFAP+), microglia
(EdU+/IBA-1+) and oligodendrocytes (EdU+/Olig 2+), see
Figure 4A. Quantification was performed by an experimenter
blind to the treatment, averaged per animal and plot as
mean ±SEM for each group. Density of counted cells was
normalized to the volume of granular cell layer sampled in each
group as previous described in Belmer et al. (2018) and Patkar
et al. (2019).
Statistics
Comparisons between groups were statistically analyzed using
t-test, one-way or two-way ANOVA, as appropriate, followed by
a Bonferroni-multiple comparison post hoc test using GraphPad
Prism 8 (Graph Pad Software Co., CA, United States). Pvalues
<0.05 were considered significant. All values are expressed as
the mean ±SEM.
RESULTS
Long-Term Sucrose Intake Increases
Weight Gain
We assessed the effects of long-term unrestricted access to
sucrose on body weight over 12 weeks (60 exposures days).
After 12 weeks of access to 25% sucrose, mice exhibited
stable levels of sucrose intake around 80–90 g/kg/day (mean
82.2 ±0.9 g/kg/24 h indicated by the red line; Figure 1B).
A significant increase in overall weight was observed, starting
around 4 weeks and increasing throughout the 12 weeks of
exposure (Figure 1C, Mixed effect repeated measure two-way
ANOVA, F(58,1276 )= 9.265, p= 0.0001) until reaching 10.6%
overweight (Bonferroni multiple comparison: 33.22 ±0.85 g vs
30.03 ±0.59 g, p<0.0002) compared to water controls.
Long-Term Sucrose Intake Does Not
Produce Anxiety- and Depressive-Like
Behavior
Sugar-withdrawn rats consistently exhibit both anxious and
depressive-like symptoms (Vollmayr et al., 2004;Avena et al.,
2008;Iemolo et al., 2012;Parylak et al., 2012;Harrell et al.,
2015;Eudave et al., 2018;Santos et al., 2018). One study in
C57BL/6 J mice consuming high levels of 10% sucrose (around
72 g/kg/24 h) for 4 weeks, found increased anxiety- (EPM) and
depressive-like behavior (tail suspension test) after one week of
withdrawal (Kim et al., 2018). To evaluate the effect of higher
level of sugar intake (around 85 g/kg/24 h) after 6 weeks of
exposure on emotional behavior, we assessed withdrawal-induced
anxious and depressive-like behaviors in the EPM, MB, OF, and
FS tests, 24 h after the last drinking session of the week. The
number of open arm entries within 5 min was similar in the EPM,
between sucrose and water control animals suggesting no change
in anxiety-related behavior (Figure 1D, ns, p= 0.5690, t-test).
The MB test also yielded a similar result with water- and sugar-
exposed animals burying a similar amount of marbles (Figure 1E,
ns, p= 0.9062, t-test). The OF test showed no difference in
the number of entries into the center between sucrose and
water exposed animals, hence confirming the absence of anxiety-
like behavior following long-term unrestricted access to sucrose
(Figure 1F, ns, p= 0.3583, t-test). No change in immobility time
in the FS was evident in sucrose consuming animals suggesting
no depressive-like symptoms (Figure 1G, ns, p= 0.6654, t-test).
Long-Term Sucrose Intake Produces
Hyperactivity/Hyperlocomotion
There is growing evidence showing that addiction and substance
dependence strongly rely on increased arousal, hyperactivity,
impulsivity and compulsion following cessation (Crews and
Boettiger, 2009). To understand how sucrose overconsumption
affects hyperactivity and compulsion, we assessed general
locomotor activity using OF. To evaluate impulse control we
used the NSF, conflict based anxiety test that has previously been
shown to reflect impulsivity-like behavior (Bevilacqua et al., 2010;
Angoa-Pérez et al., 2014;Piggott et al., 2020).
Frontiers in Neuroscience | www.frontiersin.org 4June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 5
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
FIGURE 1 | Long-term sucrose consumption increases mouse weights without altering emotional behavior. (A) Experimental design of the anxiety-related,
depression-related, general locomotor activity and impulse control behavioral testing in elevated-plus-maze (EPM), novelty suppressed feeding (NSF), marble burying
(MB), open field (OF) and forced swimming test (FS). Animals consumed 25% sucrose for 5 weeks prior to each behavioral testing and continued to be exposed to
sucrose for a total of 12 weeks. Behavioral tests were initially conducted 2 weeks (2 wk) then continued every 5 days (5d) after 24 h of sugar withdrawal. Animals
were assigned into two groups: sugar-withdrawn animals and water control. (B) Mice exhibit stable levels of long-term sucrose intake of 82.2 ±0.9 g/kg indicated
by the red line. (C) There was a significant difference in the weights of animals consuming sucrose, compared to water control, starting after 4 weeks, and continuing
throughout the 12 weeks. Data are presented as mean ±SEM; n= 8 mice/group, t-test, ***p<0.001. (D–F) Long-term sucrose consumption does not alter
anxiety-related behavior as seen by no change in the number of entries in the open-arm of the EPM (D), number of marbles buried (E) and number of entries in the
center in the open field (F) compared to water controls. Long-term sucrose consumption does not induce depressive-like symptoms as seen by no differences in the
immobility time in the forced swimming test (G) compared to water controls. Data are presented as mean ±SEM; n= 8 mice/group.
Frontiers in Neuroscience | www.frontiersin.org 5June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 6
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
Sucrose consuming mice displayed a higher general
locomotor activity as observed by an increase in total distance
traveled (Figures 2B,C) compared to water control animals
(Figures 2A,C,p<0.0001, t-test). This hyperlocomotion
was accompanied by hyperactivity as evidenced by increased
average speed in center of the open-field apparatus (Figure 2D,
p= 0.0008, t-test), total average speed (Figure 2E,p<0.0001,
t-test) in the OF apparatus, and reduced latency to feed in the
NSF, compared to water controls (Figure 2F,p= 0.0005, t-test).
Combined with hyperactivity, this lack of novelty-induced
feeding inhibition could be interpreted as a reduction of impulse
control to anxiogenic environment and further suggests that
chronic sucrose consumption produces both hyperactivity and
impulsivity (Bevilacqua et al., 2010;Angoa-Pérez et al., 2014;
Piggott et al., 2020).
Long-Term Sucrose Intake Alters Both
Episodic and Spatial Memory
Since hyperactivity correlates with memory impairments (Ortega
et al., 2020), we assessed the consequences of chronic sucrose
consumption and subsequent hyperactivity on learning and two
types of memory, episodic and spatial, using the NOR and MWM
tests, respectively. Although sucrose consuming mice learn to
discriminate between old and new objects, the proportion of
interaction time with the novel object compared to old object was
significantly lower than water control mice at 5 min (p= 0.048)
and 10 min (p= 0.028) following the presentation of the objects
(Figure 3B, Two-way ANOVA, F (1,84)= 10.35, p= 0.0018),
hence showing a significant reduction of the area under curve
(Figure 3C,t-test, p= 0.0008).
In the MWM, sucrose and water drinking mice showed similar
latency to reach a visible platform, confirming that, albeit sucrose
mice being slightly overweight compared to water controls,
there was no alteration in learning or their swimming behavior
(Figure 3E,t-test, p= 0.63). However, when the platform
was hidden, sucrose-consuming mice took longer than water
consuming mice to reach the platform area (Figure 3F,t-test,
p= 0.025). These results suggest that chronic overconsumption of
sucrose alters both episodic and spatial memory without affecting
the learning process.
Long-Term Sucrose Intake Decreases
Hippocampal Cell Proliferation and
Neurogenesis
Anxiety, depression, and cognitive deficits are strongly associated
with alteration in hippocampal neurogenesis. Although, sucrose
did not elicit anxiety- or depression-like behavior, we assessed
whether sucrose-induced deficits in memorization was associated
with changes in hippocampal neurogenesis (Figure 4A). Mice
chronically consuming a highsucrose diet showed a reduction in
the overall density of dentate gyrus proliferating cells (EdU+)
compared to water controls (Figure 4B,p= 0.0332, t-test). This
decrease in cell proliferation was likely mediated by decreased
neurogenesis, as evidenced by a reduction in the density of both
putative stem cells (stage 1: EdU+/GFAP+/Nestin;Figure 4C,
p= 0.0127, t-test) and neuronal progenitors (newborn neurons,
stage 2: EdU+/GFAP/Nestin+;Figure 4D,p= 0.0106, t-test)
compared to water controls, suggesting that continuous sucrose
intake alters the transition or differentiation of progenitors
into the proliferating phase. No change within differentiated
neuroblasts was observed (stage 3; EdU+/DCX+;p= 0.2309,
t-test, not shown) suggesting sucrose consumption principally
affects the earlier stages of neurogenesis.
Restricting the Availability of Sucrose
Dampens the Neurocognitive Deficits
The WHO’s guidelines recommend restricting the availability of
sugar in the current diet and advocate a four-time reduction
of the daily intake of sugar. Therefore, we investigated the
consequences of restricting access to sucrose solution to only
2 h/day, on weight gain, locomotion, emotion, cognition,
and neurogenesis. Mice showed a daily intake of sucrose of
20.9 ±0.3 g/kg (Figure 5A), about 4 times less than when access
was unrestricted (Figure 5B,t-test, p<0.0001). This lower
daily intake was associated with no change in weight gain over
12 weeks of exposure, compared to water controls (Figure 5C,
Two-way ANOVA, F (1, 11) = 0.3511, p= 0.5655).
We then evaluated if restricting access to sucrose alters
anxiety- and depression-related behavior in the same behavioral
tests. Restricting the access to sucrose to 2 h per day did not elicit
any change in anxiety-like behavior, as evidenced by no difference
in the number of open-arm entries in the EPM (Figure 5D,
t-test, p= 0.95), no difference in the number of marbles buried
(Figure 5E,t-test, p= 0.69) and no difference in the number
of entries in the center of the OF (Figure 5F,t-test, p= 0.11).
No difference was observed in the immobility time in the FS
(Figure 5G,t-test, p= 0.51) suggesting that restricting access to
sucrose does not elicit depressive-like behavior.
Interestingly, restricting access to sucrose still elicited
hyperactivity as shown by significant increases in the total
distance traveled (Figure 5H,t-test, p<0.0001), the speed
in the center (Figure 5I,t-test, p= 0.0002) and total speed
(Figure 5J,t-test, p<0.0001) in the OF, compared to water
control animals. However, there was no change in the latency to
feed compared to water controls (Figure 5K,t-test, p= 0.37).
These results suggest that restricting sucrose availability and
reducing overall daily intake does not affect the inhibitory control
to resist food, despite promoting hyperactivity. This increased
hyperactivity with no deficits in control to resist food was not
accompanied by any alteration in memory, as evidenced by no
change in the proportion of time interacting with a novel object
(Figure 5L,t-test, p= 0.97) or the latency to reach the platform
area in the MWM (Figure 5M,t-test, p= 0.10). This absence of
memory deficits was accompanied by no change in hippocampal
neurogenesis, however, an increased oligodendrogenesis was
observed in the dentate gyrus (Supplementary Figure 1).
DISCUSSION
Sugar became embedded in the food and beverage chains,
leading to overconsumption in children and adolescents (Han
and Powell, 2013;Dereñ et al., 2019). It is therefore important to
Frontiers in Neuroscience | www.frontiersin.org 6June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 7
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
FIGURE 2 | Long-term sucrose consumption produces hyperactivity and reduces the control to resist food. Tracking plot of water- (A) and sucrose-consuming mice
(B) illustrating that long-term sucrose consumption increases the total distances traveled in the open field (C), the speed in the center (D) and total speed (E) in the
open-field compared to water controls. Long-term sucrose consumption also reduced the latency to feed suggesting reduced control to resist food (F). Data are
presented as mean ±SEM; n= 8 mice/group. ***p<0.001, ****p<0.0001.
investigate how long-term intake of sugar, starting at adolescence,
leads to long-term effects into adulthood. The present study
shows there are deleterious effects of long-term sugar intake, on
weight gain, hyperactivity, impulsivity, and deficits in memory
and hippocampal neurogenesis.
While the WHO recommends that the amount of sugar in
sugar-sweetened be reduced by four-fold to decrease the risk of
childhood overweight and obesity (WHO, 2019), the impact of
sugar intake on the rise in obesity rates is still debated (Alexander
Bentley et al., 2020). Indeed, overall sugar consumption has
dropped since the mid-1990’s whereas the obesity rate has
continuously increased. It has been proposed that this rise
in obesity could result from a delayed effect of excess sugar,
suggesting that adult obesity could be driven by high sugar intake
over a life span (Alexander Bentley et al., 2020). There are many
ways that a decrease in overall sugar consumption and an increase
in obesity rates can be viewed. Sugar has hidden properties,
activates the hypothalamus, inhibits ghrelin and leptin, leading to
over-eating other types of foods (Jacques et al., 2019). It has been
shown that both obesity and sweet-taste can be passed on through
epigenetic modification (Öst et al., 2014;Donkin et al., 2016;
Ling and Rönn, 2019). This means that once obesity or sugar
preference has been established, it can be passed on in families
for up to 3 generations going forward.
In line with this, we observed that unrestricted access to sugar,
beginning at adolescence, only starts to affect weight gain into
adulthood, after 4–5 weeks of sucrose consumption. Although
rodent models of diet-induced obesity use body weight as a
measure of obesity, the effects of high-sugar diets on body weight
are inconsistent (Jurdak et al., 2008;Jurdak and Kanarek, 2009).
While we observed a significant increase in weight gain in sucrose
consuming animals, we cannot comment on the potential link
Frontiers in Neuroscience | www.frontiersin.org 7June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 8
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
FIGURE 3 | Long-term sucrose consumption alters episodic and spatial memory. (A) Experimental design of the testing of episodic and spatial memory in the
novel-object recognition (NOR) and Morris-watermaze (MWM) tests. Animals consumed 25% sucrose for 8 weeks prior to behavioral testing and continued to be
exposed to sucrose for a total of 12 weeks. Memory tests were conducted 3 weeks apart. Animals were assigned into two groups: sugar-withdrawn animals and
water controls. (B–D) Long-term sucrose drinking animals interact less with a novel object across the duration of the test, compared to water controls (B), as
confirmed by a reduction in the area-under-curve (AUC) (C). The 2 objects used alternatively in the novel-object-recognition test, which are discernible in shape, size
and color, are pictured in panel (D).(E–G) Long-term sucrose consuming mice showed no alteration in their learning and swimming behavior to reach a visible
platform (E), however, they showed an increased latency to reach the zone where the platform was previously placed (F) compared to water controls. Schematic
drawing of the MWM apparatus used is depicted in panel (G). Data are presented as mean ±SEM; n= 8 mice/group. *p<0.05, ***p<0.001.
between obesity and long-term unrestricted intake of sugar.
Further investigation of metabolic marker expression, such as
adiposity, insulin resistance, leptin, adiponectin, will be needed
to elucidate how long-term sucrose consumption predisposes to
obesity. Interestingly, we found that a 4-times reduction of daily
sucrose intake is able to prevent sugar-induced increase in weight
gain, supporting the WHO’s guideline to reduce the impact of
sugar on the rise of obesity rate.
We showed that long-term sucrose consumption did not
produce any anxiety- or depression-related behavior, although
previous studies have shown that acute and chronic withdrawal
from sucrose can induce anxious and depressive like behaviors
(Colantuoni et al., 2002;Avena et al., 2008;Iemolo et al., 2012;
Parylak et al., 2012;Harrell et al., 2015;Eudave et al., 2018;
Kim et al., 2018;Santos et al., 2018). Anxiety- and depression-
like behaviors are likely present if withdrawal follows extensive
periods of sucrose consumption. Short-term exposure to sucrose
(<1 month) does not lead to increased anxiety-like behavior in
rats (Parylak et al., 2012) while longer exposure (>1 month)
results in increased anxiety-like behavior 24 h after withdrawal
(Colantuoni et al., 2002;Avena et al., 2008). Interestingly, short-
term unrestricted access to 10% sucrose induced depression-
and anxiety-like behavior after one week withdrawal (Kim et al.,
2018). Since the aforementioned studies used different models of
sucrose consumption with concentrations of sucrose solutions
ranging from 7.9 to 35%, we cannot rule out an absence
of anxiety-like behavior in our study due to methodological
or interspecies differences. In addition, many studies incited
a binge-like patterns of sugar (10% sucrose or 25% glucose)
consumption by food-depriving the animals for 12 h before
the sucrose drinking sessions. The food deprivation/restriction
introduced in these studies may have changed motivational
states and reward seeking behavior, adding another level of
psychological stress (Toth and Gardiner, 2000). Strikingly,
Frontiers in Neuroscience | www.frontiersin.org 8June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 9
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
FIGURE 4 | Long-term sucrose consumption reduces the early phases of hippocampal neurogenesis. (A) Stages of neurogenesis in the dentate gyrus of the
hippocampus. In stage 1 (proliferation phase; in yellow), the newly generated cells express glial fibrillary acidic protein (GFAP) and are putative progenitor/stem cells
located in the subgranular zone (SGZ). The cells in stage 2 (differentiation phase; in green) will lose their GFAP and start expressing Nestin. This determines their fate
in the neuronal lineage. Stage 1 progenitors not only give rise to newborn neurons, they can also turn into glial cells. Glial cells [astrocyte and oligodendrocyte
progenitor cell (OPC)] can convert back into newborn neurons here in stage 2 and vice-versa. In stage 3, the immature neurons express doublecortin (DCX; in cobalt
blue) and have started to migrate into the granule cell layer (GCL) of the dentate gyrus. As the neuron matures, it will send its dendrites toward the molecular layer
(ML) of the dentate gyrus and extend their axonal projections toward the hippocampal CA3 pyramidal cell layer and will start losing their DCX and start expressing
postmitotic neuronal marker NeuN and calretinin (stage 4; magenta). As the neuron establishes synaptic contacts from the entorhinal cortex and it sends output to
the CA3 and hilus regions of the hippocampus the neuron is classified as in stage 5 (in cherry red). Stage 5 neurons start expressing calbindin and continue
(Continued)
Frontiers in Neuroscience | www.frontiersin.org 9June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 10
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
FIGURE 4 | Continued
expressing NeuN. Unlike the astrocytes and oligodendrocytes that are derived from the neuroectoderm, microglia (in cyan blue) are neuroglia derived by embryonic
mesoderm. Abbreviations used: GFAP, glial fibrillary acidic protein; OPC, oligodendrocyte progenitor cell; Olig2, oligodendrocyte lineage transcription factor 2; IBA-1,
ionized calcium binding adaptor molecule 1; DCX, doublecortin; NeuN, Fox-3, Rbfox3, or Hexaribonucleotide Binding Protein-3; SGZ, subgranular zone; GCL,
granule cell layer; ML, molecular layer [original drawing, created using Biorender, adapted from Lucassen et al. (2010)]. (B) Long-term sugar consumption reduced
the density of EdU positive cells (green) in the dentate gyrus of the hippocampus. (C) There was a reduction in the density of EdU+(green)/GFAP+(red)
immunoreactive cells indicating a reduction in stage 1 (putative stem cells) neurogenesis. (D) A reduction was also observed in the number of EdU+(green)/Nestin+
(red)-immunoreactive cells indicating a reduction in stage 2 (neuronal progenitors). All images are colocalized with DAPI (blue). Data are presented as mean ±SEM;
n= 8 mice/group. *p<0.05. Representative image scale bar is 100 µm and close up representative image scale bar is 10 µm.
longer sucrose exposure (>3 months) showed no significant
effects on anxiety- and depression-like behavior in rats (Cao
et al., 2007;Chepulis et al., 2009), hence suggesting that long
term (i.e., greater than 1 month but less than 3 months)
consumption of sucrose increases anxiety-like behavior, followed
by a return to baseline levels of anxiety- and depression-related
behavior at 3 months.
Overall, we observed no anxious or depressive behavior in
mice after 6–10 weeks of sucrose consumption. The reliability
of our findings is bolstered by the fact that we used two
different models, restricted and unrestricted. Perhaps sugar
does not produce the same emotional deficits between species.
An explanation could be due to differences metabolism, with
a well-described faster metabolism in mice compared to rats
(Radermacher and Haouzi, 2013). It may also be possible
that higher concentrations of sucrose (greater than 10%)
recruits different/additional neural circuits, involving other
behavioral deficits, such as hyperactivity and/or impulsivity
that mask the behavioral inhibition produced by anxiogenics
cues/environments.
Our results showed that long-term sucrose overconsumption
increases basal locomotor activity, which could be interpreted
as hyperactivity. There are limited studies examining the effect
of sucrose on locomotor activity, with, to the best of our
knowledge, only one study reporting no change in locomotor
activity in rats (Avena and Hoebel, 2003a). Our observation
of sugar-induced hyperactivity prompted the investigation of
the effect of long-term sugar consumption on impulse control
and inhibitory control to resist food. Interestingly, long-
term sucrose consumption reduced impulse control in the
novelty-suppressed feeding, and this was not observed when
sucrose access was restricted. Although novelty-suppressed
feeding test is primarily to assess anxiety-like behaviors,
studies have shown that a reduced latency to feed could be
correlated with augmented food seeking and increased meal
size (Biddinger et al., 2020), increased hunger and reduced-
feeding control after fasting (Burghardt et al., 2016), and
together with hyperlocomotion, increased motor impulsivity
(Bevilacqua et al., 2010;Angoa-Pérez et al., 2014;Piggott
et al., 2020). This suggests that the reduced latency to feed we
observed after long-term sucrose intake could be the result of
impulsivity. However, there remains disparity in the literature
regarding sugar’s effect on impulsivity in rats with studies
suggesting sugar does not (Stein et al., 2015;Wong et al.,
2017) and others supporting sugar does produce impulsivity
(Steele et al., 2017). Therefore, further investigation is required
to identify the mechanism underlying the effect of sugar
on locomotor and impulsive behavior in mice. This could
be explored further using delay discounting test (temporal
discounting) or 5-choice serial reaction time task (visual
attentional processes and impulse control) (Reichelt et al., 2015,
2016;Lemos et al., 2016).
Excessive sucrose consumption in adolescent rats has been
associated with deficits in spatial memory or object recognition
memory (Reichelt et al., 2015, 2016;Lemos et al., 2016), and
this could be principally mediated by the fructose component of
sucrose (Hsu et al., 2015). However, the link between memory
deficits and changes in hippocampal neurogenesis following
long-term sucrose consumption has been relatively unexplored.
In our study, reduction in hippocampal neurogenesis was only
observed when memory deficits were observed, for example,
when access to sucrose was unrestricted.
Indeed, no change in hippocampal neurogenesis was observed
when sucrose access was restricted, and memory not affected. Our
results further suggest that unrestricted sucrose consumption
likely affects neurogenesis by reducing cell proliferation,
generation of putative stem cell and survival/maturation of
newborn neurons.
Reduced cell proliferation followed by reduced production
of neuronal progenitors suggest reduced neurogenesis/turn over
(Cisternas et al., 2015). This result is in accordance with previous
ethanol studies in rodents suggesting sugar consumption is also
intervening at the G1 phase of the cell cycle, changing the
number of cells entering the S phase (Belmer et al., 2018;Patkar
et al., 2019). Interestingly, we did not see an effect on neuroblast
differentiation, suggesting the reduction in the initial number of
putative stem cells able to dedifferentiate and proliferate. This
could be due to the generation of pluripotent transit-amplifying
progenitor cells [TAPs (Potten and Loeffler, 1990)] that remain
in quiescence for long periods before differentiation (Doetsch
et al., 2002). It is possible that sugar reduces the number of TAPs
resulting in the changes observed here. Early-stage neurogenic
deficits have not been observed previously. Our results found
a reduction in putative stem cell and newborn neurons with
no change in differentiated neuroblasts. This absence of effect
of sugar on neuroblast differentiation has also been reported
in rats with unrestricted access to sucrose, therefore confirming
a degree of interspecies similarity. Another explanation of
the reduction in proliferating cells is cell death. Unrestricted
consumption of sugar has been shown to increase apoptosis
(TUNEL) suggesting our reduction in proliferating cells could
be due to neuronal death as a result of sugar overconsumption
Frontiers in Neuroscience | www.frontiersin.org 10 June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 11
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
FIGURE 5 | Restricting access to sucrose consumption results in an absence of increased weight gain with no emotional and cognitive alterations. (A) Mice exhibit
stable levels of long-term sucrose intake of 20.9 ±0.3 g/kg indicated by the red line. This level of sucrose intake is four-fold lower than when sucrose access was
unrestricted (B) and was not associated with any changes in weight gain (C), or anxiety-related behavior as seen by no change in the number of entries in the
open-arm of the EPM (D), the number of marbles buried (E) and the number of entries in the center in the open field (F) compared to water controls. Restricting
long-term sucrose consumption did not induce depressive-like symptoms as seen by no differences in the immobility time in the forced swimming test (G) compared
to water controls. Restricted sucrose consumption increases the total distances traveled in the open field (H), the speed in the center (I) and total speed (J) in the
open-field, however, there was no change in the latency to feed (K), the time spent with a novel object (L) and the latency to reach the zone where the platform was
previously placed (M), compared to water controls. Data are presented as mean ±SEM; n= 8 mice/group. ***p<0.001, ****p<0.0001.
Frontiers in Neuroscience | www.frontiersin.org 11 June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 12
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
(van der Borght et al., 2011). Neuronal maturation and survival
have been reported to be reduced in the dentate gyrus of the
hippocampus after sugar consumption (van der Borght et al.,
2011;Cisternas et al., 2015). However, it is possible their protocol
of cell proliferation labeling (BrdU injection two weeks prior) has
misevaluated neuronal maturation and survival as the maturation
process of neurons can take up 1–2 months in rodents (Zhao
et al., 2008).
A limitation of this investigation is that the results may
not be applicable to female models of sugar overconsumption,
as only male mice were tested. C57/Bl6 male mice have both
increased overall activity and heightened anxiety when compared
to female mice particularly in maternal separation and chronic
stress models (Romeo et al., 2003;Veenema et al., 2007;Niwa
et al., 2011), meaning that subtle differences in the behavior of
male mice are more easily detected compared to female mice.
Female sex hormones also influence appetitive signaling in the
brain (Gao et al., 2007;Santiago et al., 2016), which therefore
increases the methodological complexity of including female
mice in this study, as all data would need to be normalized
to the estrous cycle. Females are widely underrepresented
in preclinical models of addiction (Beery and Zucker, 2011;
Shansky, 2019), with a majority of studies in this field conducted
exclusively on male mice. This is a major ongoing issue with
neuroscience research, and future investigations should explore
if similar behavioral consequences of sugar consumption are
present in female rodents. However, we believe the results of
this study provide foundational knowledge that can be extended
upon to benefit addiction and obesity generally. Together, our
study demonstrates that excessive sugar consumption starting
at adolescence elicits profound locomotor and memory deficits
in adulthood, that may mimic the hyperactivity and cognitive
dysfunctions observed in attention deficits and hyperactivity-like
disorders. More interestingly, our results reveal that restricting
sugar consumption intake, as recommended by the WHO, might
be effective in limiting the negative consequences of sugar on
obesity, and locomotor and cognitive impairments.
DATA AVAILABILITY STATEMENT
The raw data supporting the conclusions of this article will be
made available by the authors, without undue reservation.
ETHICS STATEMENT
The animal study was reviewed and approved by The University
of Queensland and The Queensland University of Technology
Animal Ethics Committees under approval QUT/053/18.
AUTHOR CONTRIBUTIONS
KB, AB, and SEB were responsible for the study concept and
design. KB and IAC carried out the drinking experiments. KB,
AB, and IAC performed behavioral animal experiments, analyzed
the data and interpreted the findings. KB designed and performed
the immunohistochemistry experiments and acquired the images
with the technical advice of SBW at SBMS facility at The
University of Queensland. KB and AB drafted the manuscript and
drafted the figures. KB and JW drafted the drawing in Figure 3.
SEB, IAC, FC, JW, and AB reviewed and edited the manuscript.
All authors have critically reviewed the content and approved
final version for submission.
FUNDING
This work was supported by National Health and Medical
Research Council (NHMRC) (GNT1146417) to SEB.
ACKNOWLEDGMENTS
We are thankful to PACE animal facility manager Lisa Foster
and her staff Miranda Sleath, Rachel Smith, and Annie Villalta-
Burgett for the exquisite care of our animals. We are grateful
to the imaging facility of the Translational Research Institute,
the facility manager Sandrine Roy and the microscopy officer
Ali Ju, and the School of Biomedical Sciences at University of
Queensland, facility manager Shaun B. Walters for the extensive
use of resources.
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found
online at: https://www.frontiersin.org/articles/10.3389/fnins.
2021.670430/full#supplementary-material
Supplementary Figure 1 | Restricting access to sucrose consumption augments
hippocampal oligodendrogenesis. (A,B) Restricted sugar consumption increased
the density of EdU positive cells (green) in the dentate gyrus of the hippocampus.
(C,D) There was no change in the density of stage 1 putative stem cells
[EdU+/GFAP+,(C)] or stage 3 immature neurons [EdU+/DCX+,(D)] suggesting
no change in neurogenesis, however, it appears that the increased density of
EdU+proliferative cells was mediated by an increased density of oligodendrocytes
progenitors [EdU+(green)/Olig+(magenta), (E,F)]. (G) There was no difference in
the density of proliferative microglia in the dentate gyrus, following restricted sugar
consumption, compared to water controls. All images are colocalized with DAPI
(blue). Representative image scale bar is 100 µm and close up representative
image scale bar is 10 µm.
REFERENCES
Alexander Bentley, R., Ruck, D. J., and Fouts, H. N. (2020). U.S. obesity as delayed
effect of excess sugar. Econ. Hum. Biol. 36:100818. doi: 10.1016/j.ehb.2019.
100818
Altfas, J. R. (2002). Prevalence of attention deficit/hyperactivity disorder among
adults in obesity treatment. BMC Psychiatry 2:9. doi: 10.1186/1471-244x-2-9
Angoa-Pérez, M., Kane, M. J., Sykes, C. E., Perrine, S. A., Church, M. W., and Kuhn,
D. M. (2014). Brain serotonin determines maternal behavior and offspring
survival. Genes Brain Behav. 13, 579–591. doi: 10.1111/gbb.12159
Frontiers in Neuroscience | www.frontiersin.org 12 June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 13
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
Avena, N. M. (2010). The study of food addiction using animal models of binge
eating. Appetite 55, 734–737. doi: 10.1016/j.appet.2010.09.010
Avena, N. M., and Hoebel, B. G. (2003a). A diet promoting sugar dependency
causes behavioral cross-sensitization to a low dose of amphetamine.
Neuroscience 122, 17–20. doi: 10.1016/s0306-4522(03)00502-5
Avena, N. M., and Hoebel, B. G. (2003b). Amphetamine-sensitized rats show sugar-
induced hyperactivity (cross-sensitization) and sugar hyperphagia. Pharmacol.
Biochem. Behav. 74, 635–639. doi: 10.1016/s0091-3057(02)01050-x
Avena, N. M., Bocarsly, M. E., Rada, P., Kim, A., and Hoebel, B. G. (2008). After
daily bingeing on a sucrose solution, food deprivation induces anxiety and
accumbens dopamine/acetylcholine imbalance. Physiol. Behav. 94, 309–315.
doi: 10.1016/j.physbeh.2008.01.008
Avena, N. M., Long, K. A., and Hoebel, B. G. (2005). Sugar-dependent rats show
enhanced responding for sugar after abstinence: evidence of a sugar deprivation
effect. Physiol. Behav. 84, 359–362. doi: 10.1016/j.physbeh.2004.12.016
Avena, N. M., Rada, P., and Hoebel, B. G. (2009). Sugar and fat bingeing have
notable differences in addictive-like behavior. J. Nutr. 139, 623–628. doi: 10.
3945/jn.108.097584
Bailey, K. R., and Crawley, J. N. (2009). “Anxiety-related behaviors in mice, in
Methods of Behavior Analysis in Neuroscience, ed. J. J. Buccafusco (Boca Raton,
FL: CRC Press/Taylor & Francis).
Beery, A. K., and Zucker, I. (2011). Sex bias in neuroscience and biomedical
research. Neurosci. Biobehav. Rev. 35, 565–572. doi: 10.1016/j.neubiorev.2010.
07.002
Belmer, A., Patkar, O. L., Lanoue, V., and Bartlett, S. E. (2018). 5-HT1A
receptor-dependent modulation of emotional and neurogenic deficits elicited
by prolonged consumption of alcohol. Sci. Rep. 8:2099.
Bevilacqua, L., Doly, S., Kaprio, J., Yuan, Q., Tikkanen, R., Paunio, T., et al. (2010).
A population-specific HTR2B stop codon predisposes to severe impulsivity.
Nature 468, 1061–1066. doi: 10.1038/nature09629
Biddinger, J. E., Lazarenko, R. M., Scott, M. M., and Simerly, R. (2020). Leptin
suppresses development of GLP-1 inputs to the paraventricular nucleus of the
hypothalamus. eLife 9:e59857.
Burghardt, P., Krolewski, D. M., Dykhuis, K. E., Ching, J., Pinawin, A. M.,
Britton, S. L., et al. (2016). Nucleus accumbens cocaine-amphetamine regulated
transcript mediates food intake during novelty conflict. Physiol. Behav. 158,
76–84. doi: 10.1016/j.physbeh.2016.02.035
Cabanac, M., and Johnson, K. G. (1983). Analysis of a conflict between palatability
and cold exposure in rats. Physiol. Behav. 31, 249–253. doi: 10.1016/0031-
9384(83)90128-2
Cao, D., Lu, H., Lewis, T. L., and Li, L. (2007). Intake of sucrose-sweetened water
induces insulin resistance and exacerbates memory deficits and amyloidosis in a
transgenic mouse model of Alzheimer disease. J. Biol. Chem. 282, 36275–36282.
doi: 10.1074/jbc.m703561200
Chepulis, L. M., Starkey, N. J., Waas, J. R., and Molan, P. C. (2009). The effects of
long-term honey, sucrose or sugar-free diets on memory and anxiety in rats.
Physiol. Behav. 97, 359–368. doi: 10.1016/j.physbeh.2009.03.001
Choi, C. S., Kim, P., Park, J. H., Gonzales, E. L., Kim, K. C., Cho, K. S., et al. (2015).
High sucrose consumption during pregnancy induced ADHD-like behavioral
phenotypes in mice offspring. J. Nutr. Biochem. 26, 1520–1526. doi: 10.1016/j.
jnutbio.2015.07.018
Cisternas, P., Salazar, P., Serrano, F. G., Montecinos-Oliva, C., Arredondo, S. B.,
Varela-Nallar, L., et al. (2015). Fructose consumption reduces hippocampal
synaptic plasticity underlying cognitive performance. Biochim. Biophys. Acta
BBA Mol. Basis Dis. 1852, 2379–2390. doi: 10.1016/j.bbadis.2015.08.016
Colantuoni, C., Rada, P., McCarthy, J., Patten, C., Avena, N. M., Chadeayne,
A., et al. (2002). Evidence that intermittent, excessive sugar intake causes
endogenous opioid dependence. Obes. Res. 10, 478–488. doi: 10.1038/oby.2002.
66
Cortese, S. (2019). The association between ADHD and Obesity: intriguing,
progressively more investigated, but still puzzling. Brain Sci 9, 256. doi: 10.
3390/brainsci9100256
Cortese, S., Moreira-Maia, C. R., St Fleur, D., Morcillo-Peñalver, C., Rohde, L. A.,
and Faraone, S. V. (2016). Association between ADHD and obesity: a systematic
review and meta-analysis. Am. J. Psychiatry 173, 34–43.
Crews, F. T., and Boettiger, C. A. (2009). Impulsivity, frontal lobes and risk for
addiction. Pharmacol. Biochem. Behav. 93, 237–247. doi: 10.1016/j.pbb.2009.
04.018
Criscitelli, K., and Avena, N. M. (2016). The neurobiological and behavioral
overlaps of nicotine and food addiction. Prev. Med. 92, 82–89. doi: 10.1016/
j.ypmed.2016.08.009
Deacon, R. M. J. (2006). Digging and marble burying in mice: simple methods
for in vivo identification of biological impacts. Nat. Protoc. 1, 122–124. doi:
10.1038/nprot.2006.20
Del-Ponte, B., Anselmi, L., Assunção, M. C. F., Tovo-Rodrigues, L., Munhoz,
T. N., Matijasevich, A., et al. (2019). Sugar consumption and attention-
deficit/hyperactivity disorder (ADHD): a birth cohort study. J. Affect. Disord.
243, 290–296.
Dereñ, K., Weghuber, D., Caroli, M., Koletzko, B., Thivel, D., Frelut, M. L., et al.
(2019). Consumption of sugar-sweetened beverages in paediatric age: a position
paper of the european academy of paediatrics and the european childhood
obesity group. Ann. Nutr. Metab. 74, 296–302. doi: 10.1159/000499828
Doetsch, F., Petreanu, L., Caille, I., Garcia-Verdugo, J.-M., and Alvarez-Buylla,
A. E. G. F. (2002). Converts transit-amplifying neurogenic precursors in the
adult brain into multipotent stem cells. Neuron 36, 1021–1034. doi: 10.1016/
s0896-6273(02)01133-9
Donkin, I., Versteyhe, S., Ingerslev, L. R., Qian, K., Mechta, M., Nordkap, L., et al.
(2016). Obesity and bariatric surgery drive epigenetic variation of spermatozoa
in humans. Cell Metab. 23, 369–378. doi: 10.1016/j.cmet.2015.11.004
Eudave, D. M., BeLow, M. N., and Flandreau, E. I. (2018). Effects of high fat or high
sucrose diet on behavioral-response to social defeat stress in mice. Neurobiol.
Stress 9, 1–8. doi: 10.1016/j.ynstr.2018.05.005
Farsad-Naeimi, A., Asjodi, F., Omidian, M., Askari, M., Nouri, M., Pizarro,
A. B., et al. (2020). Sugar consumption, sugar sweetened beverages and
Attention Deficit Hyperactivity Disorder: a systematic review and meta-
analysis. Complement. Ther. Med. 53:102512. doi: 10.1016/j.ctim.2020.102512
Flores-Ramirez, F. J., Parise, L. F., Alipio, J. B., Garcia-Carachure, I., Castillo,
S. A., Rodriguez, M., et al. (2019). Adolescent fluoxetine history impairs spatial
memory in adult male, but not female, C57BL/6 mice. J. Affect. Disord. 249,
347–356. doi: 10.1016/j.jad.2019.02.051
Foo, H., and Mason, P. (2005). Sensory suppression during feeding. Proc. Natl.
Acad. Sci. U.S.A. 102, 16865–16869. doi: 10.1073/pnas.0506226102
Freeman, C. R., Zehra, A., Ramirez, V., Wiers, C. E., Volkow, N. D., and Wang,
G. J. (2018). Impact of sugar on the body, brain, and behavior. Front. Biosci.
Landmark Ed. 23:2255–2266. doi: 10.2741/4704
Gao, Q., Mezei, G., Nie, Y., Rao, Y., Choi, C. S., Bechmann, I., et al. (2007).
Anorectic estrogen mimics leptin’s effect on the rewiring of melanocortin cells
and Stat3 signaling in obese animals. Nat. Med. 13, 89–94. doi: 10.1038/nm1525
Gueye, A. B., Vendruscolo, L. F., de Ávila, C., Le Moine, C., Darnaudéry, M.,
and Cador, M. (2018). Unlimited sucrose consumption during adolescence
generates a depressive-like phenotype in adulthood. Neuropsychopharmacology
43, 2627–2635. doi: 10.1038/s41386- 018-0025-9
Han, E., and Powell, L. M. (2013). Consumption patterns of sugar-sweetened
beverages in the United States. J. Acad. Nutr. Diet. 113, 43–53. doi: 10.1016/
j.jand.2012.09.016
Harrell, C. S., Burgado, J., Kelly, S. D., Johnson, Z. P., and Neigh, G. N.
(2015). High-fructose diet during periadolescent development increases
depressive-like behavior and remodels the hypothalamic transcriptome in male
rats. Psychoneuroendocrinology 62, 252–264. doi: 10.1016/j.psyneuen.2015.08.
025
Hsu, T. M., Konanur, V. R., Taing, L., Usui, R., Kayser, B. D., Goran, M. I.,
et al. (2015). Effects of sucrose and high fructose corn syrup consumption on
spatial memory function and hippocampal neuroinflammation in adolescent
rats. Hippocampus 25, 227–239. doi: 10.1002/hipo.22368
Iemolo, A., Valenza, M., Tozier, L., Knapp, C. M., Kornetsky, C., Steardo, L., et al.
(2012). Withdrawal from chronic, intermittent access to a highly palatable food
induces depressive-like behavior in compulsive eating rats. Behav. Pharmacol.
23, 593–602. doi: 10.1097/fbp.0b013e328357697f
Jacques, A., Chaaya, N., Beecher, K., Ali, S. A., Belmer, A., and Bartlett, S. (2019).
The impact of sugar consumption on stress driven, emotional and addictive
behaviors. Neurosci. Biobehav. Rev. 103, 178–199. doi: 10.1016/j.neubiorev.
2019.05.021
Johnson, R. J., Gold, M. S., Johnson, D. R., Ishimoto, T., Lanaspa, M. A., Zahniser,
N. R., et al. (2011). Attention-deficit/hyperactivity disorder: is it time to
reappraise the role of sugar consumption? Postgrad. Med. 123, 39–49. doi:
10.3810/pgm.2011.09.2458
Frontiers in Neuroscience | www.frontiersin.org 13 June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 14
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
Johnson, R. J., Sánchez-Lozada, L. G., Andrews, P., and Lanaspa, M. A. (2017).
Perspective: a historical and scientific perspective of sugar and its relation with
obesity and diabetes. Adv. Nutr. Bethesda Md 8, 412–422. doi: 10.3945/an.116.
014654
Jurdak, N., and Kanarek, R. B. (2009). Sucrose-induced obesity impairs novel object
recognition learning in young rats. Physiol. Behav. 96, 1–5. doi: 10.1016/j.
physbeh.2008.07.023
Jurdak, N., Lichtenstein, A. H., and Kanarek, R. B. (2008). Diet-induced obesity
and spatial cognition in young male rats. Nutr. Neurosci. 11, 48–54. doi:
10.1179/147683008x301333
Kendig, M. D. (2014). Cognitive and behavioural effects of sugar consumption in
rodents. Rev. Appetite 80, 41–54. doi: 10.1016/j.appet.2014.04.028
Kenny, P. J. (2011). Reward mechanisms in obesity: new insights and future
directions. Neuron 69, 664–679. doi: 10.1016/j.neuron.2011.02.016
Kim, S., Shou, J., Abera, S., and Ziff, E. B. (2018). Sucrose withdrawal induces
depression and anxiety-like behavior by Kir2.1 upregulation in the nucleus
accumbens. Neuropharmacology 130, 10–17. doi: 10.1016/j.neuropharm.2017.
11.041
Kim, Y., and Chang, H. (2011). Correlation between attention deficit hyperactivity
disorder and sugar consumption, quality of diet, and dietary behavior in school
children. Nutr. Res. Pract. 5, 236–245. doi: 10.4162/nrp.2011.5.3.236
Klenowski, P. M., Shariff, M. R., Belmer, A., Fogarty, M. J., Mu, E. W., Bellingham,
M. C., et al. (2016). Prolonged consumption of sucrose in a binge-like manner,
alters the morphology of medium spiny neurons in the nucleus accumbens
shell. Front. Behav. Neurosci 10:54. doi: 10.3389/fnbeh.2016.00054
Krasnova, I. N., Marchant, N. J., Ladenheim, B., McCoy, M. T., Panlilio, L. V.,
Bossert, J. M., et al. (2014). Incubation of methamphetamine and palatable
food craving after punishment-induced abstinence. Neuropsychopharmacology
39, 2008–2016. doi: 10.1038/npp.2014.50
Kruse, M. S., Vadillo, M. J., Miguelez Fernández, A. M. M., Rey, M., Zanutto, B. S.,
and Coirini, H. (2019). Sucrose exposure in juvenile rats produces long-term
changes in fear memory and anxiety-like behavior. Psychoneuroendocrinology
104, 300–307. doi: 10.1016/j.psyneuen.2019.03.016
Leger, M., Quiedeville, A., Bouet, V., Haelewyn, B., Boulouard, M., Schumann-
Bard, P., et al. (2013). Object recognition test in mice. Nat. Protoc. 8, 2531–2537.
Lemos, C., Rial, D., Gonçalves, F. Q., Pires, J., Silva, H. B., Matheus, F. C.,
et al. (2016). High sucrose consumption induces memory impairment in
rats associated with electrophysiological modifications but not with metabolic
changes in the hippocampus. Neuroscience 315, 196–205. doi: 10.1016/j.
neuroscience.2015.12.018
Lenoir, M., Serre, F., Cantin, L., and Ahmed, S. H. (2007). Intense sweetness
surpasses cocaine reward. PLoS One 2:e698. doi: 10.1371/journal.pone.0000698
Li, L., Taylor, M. J., Bälter, K., Kuja-Halkola, R., Chen, Q., Hegvik, T. A., et al.
(2020). Attention-deficit/hyperactivity disorder symptoms and dietary habits in
adulthood: a large population-based twin study in Sweden. Am. J. Med. Genet.
183, 475–485. doi: 10.1002/ajmg.b.32825
Ling, C., and Rönn, T. (2019). Epigenetics in human obesity and type 2 diabetes.
Cell Metab. 29, 1028–1044. doi: 10.1016/j.cmet.2019.03.009
Lucassen, P. J., Meerlo, P., Naylor, A. S., van Dam, A. M., Dayer, A. G., Czéh,
B., et al. (2010). Regulation of adult neurogenesis by stress, sleep disruption,
exercise and inflammation: implications for depression and antidepressant
action. Eur. Neuropsychopharmacol. 20, 1–17. doi: 10.1016/j.euroneuro.2009.
08.003
Mandyam, C. D., Harburg, G. C., and Eisch, A. J. (2007). Determination of key
aspects of precursor cell proliferation, cell cycle length and kinetics in the
adult mouse subgranular zone. Neuroscience 146, 108–122. doi: 10.1016/j.
neuroscience.2006.12.064
Molteni, R., Barnard, R. J., Ying, Z., Roberts, C. K., and Gómez-Pinilla, F. (2002).
A high-fat, refined sugar diet reduces hippocampal brain-derived neurotrophic
factor, neuronal plasticity, and learning. Neuroscience 112, 803–814. doi: 10.
1016/s0306-4522(02)00123-9
Niwa, M., Matsumoto, Y., Mouri, A., Ozaki, N., and Nabeshima, T. (2011).
Vulnerability in early life to changes in the rearing environment plays
a crucial role in the aetiopathology of psychiatric disorders. Int. J.
Neuropsychopharmacol. 14, 459–477. doi: 10.1017/S1461145710001239
Ortega, R., López, V., Carrasco, X., Escobar, M. J., García, A. M., Parra, M. A., et al.
(2020). Neurocognitive mechanisms underlying working memory encoding
and retrieval in Attention-Deficit/Hyperactivity disorder. Sci. Rep. 10:7771.
Öst, A., Lempradl, A., Casas, E., Weigert, M., Tiko, T., Deniz, M., et al. (2014).
Paternal diet defines offspring chromatin state and intergenerational obesity.
Cell 159, 1352–1364. doi: 10.1016/j.cell.2014.11.005
Oswald, K. D., Murdaugh, D. L., King, V. L., and Boggiano, M. M. (2011).
Motivation for palatable food despite consequences in an animal model of binge
eating. Int. J. Eat. Disord. 44, 203–211. doi: 10.1002/eat.20808
Paglia, L. (2019). The sweet danger of added sugars. Eur. J. Paediatr. Dent. 20:89.
Parylak, S. L., Cottone, P., Sabino, V., Rice, K. C., and Zorrilla, E. P. (2012). Effects
of CB1 and CRF1 receptor antagonists on binge-like eating in rats with limited
access to a sweet fat diet: lack of withdrawal-like responses. Physiol. Behav. 107,
231–242. doi: 10.1016/j.physbeh.2012.06.017
Patkar, O. L., Belmer, A., Beecher, K., Jacques, A., and Bartlett, S. E. (2019).
Pindolol rescues anxiety-like behavior and neurogenic maladaptations of long-
term binge alcohol intake in mice. Front. Behav. Neurosci 13:264. doi: 10.3389/
fnbeh.2019.00264
Patkar, O. L., Belmer, A., Holgate, J. Y., Tarren, J. R., Shariff, M. R., Morgan, M.,
et al. (2017). The antihypertensive drug pindolol attenuates long-term but not
short-term binge-like ethanol consumption in mice. Addict. Biol. 22, 679–691
doi: 10.1111/adb.12359
Piggott, V. M., Lloyd, S. C., Perrine, S. A., and Conti, A. C. (2020). Chronic
intermittent ethanol exposure increases ethanol consumption following
traumatic stress exposure in mice. Front. Behav. Neurosci. 14:114. doi: 10.3389/
fnbeh.2020.00114
Potten, C. S., and Loeffler, M. (1990). Stem cells: attributes, cycles, spirals, pitfalls
and uncertainties. Lessons for and from the crypt. Development 110, 1001–1020.
doi: 10.1242/dev.110.4.1001
Rada, P., Avena, N. M., and Hoebel, B. G. (2005). Daily bingeing on sugar
repeatedly releases dopamine in the accumbens shell. Neuroscience 134, 737–
744. doi: 10.1016/j.neuroscience.2005.04.043
Radermacher, P., and Haouzi, P. (2013). A mouse is not a rat is not a man: species-
specific metabolic responses to sepsis - a nail in the coffin of murine models for
critical care research? Intensive Care Med. Exp. 1:26.
Reichelt, A. C., Killcross, S., Hambly, L. D., Morris, M. J., and Westbrook, R. F.
(2015). Impact of adolescent sucrose access on cognitive control, recognition
memory, and parvalbumin immunoreactivity. Learn. Mem. 22, 215–224. doi:
10.1101/lm.038000.114
Reichelt, A. C., Morris, M. J., and Westbrook, R. F. (2016). Daily access to sucrose
impairs aspects of spatial memory tasks reliant on pattern separation and
neural proliferation in rats. Learn. Mem. Cold Spring Harb. N 23, 386–390.
doi: 10.1101/lm.042416.116
Rhodes, J. S., Best, K., Belknap, J. K., Finn, D. A., and Crabbe, J. C. (2005).
Evaluation of a simple model of ethanol drinking to intoxication in C57BL/6J
mice. Physiol. Behav. 84, 53–63. doi: 10.1016/j.physbeh.2004.10.007
Romeo, R. D., Mueller, A., Sisti, H. M., Ogawa, S., McEwen, B. S., and Brake,
W. G. (2003). Anxiety and fear behaviors in adult male and female C57BL/6
mice are modulated by maternal separation. Horm. Behav. 43, 561–567. doi:
10.1016/s0018-506x(03)00063-1
Santiago, A. M., Clegg, D. J., and Routh, V. H. (2016). Ventromedial hypothalamic
glucose sensing and glucose homeostasis vary throughout the estrous cycle.
Physiol. Behav. 167, 248–254. doi: 10.1016/j.physbeh.2016.09.021
Santos, C. J., Ferreira, A. V. M., Oliveira, A. L., Oliveira, M. C., Gomes, J. S.,
and Aguiar, D. C. (2018). Carbohydrate-enriched diet predispose to anxiety
and depression-like behavior after stress in mice. Nutr. Neurosci. 21, 33–39.
doi: 10.1080/1028415x.2016.1213529
Shansky, R. M. (2019). Are hormones a “female problem” for animal research?
Science 364, 825–826. doi: 10.1126/science.aaw7570
Shariff, M., Klenowski, P., Morgan, M., Patkar, O., Mu, E., Bellingham, M.,
et al. (2017). Binge-like sucrose consumption reduces the dendritic length and
complexity of principal neurons in the adolescent rat basolateral amygdala.
PLoS One 12:e0183063. doi: 10.1371/journal.pone.0183063
Shariff, M., Quik, M., Holgate, J., Morgan, M., Patkar, O. L., Tam, V., et al. (2016).
Neuronal nicotinic acetylcholine receptor modulators reduce sugar intake.
PLoS One 11:e0150270. doi: 10.1371/journal.pone.0150270
Sigala, D. M., and Stanhope, K. L. (2021). An exploration of the role of sugar-
sweetened beverage in promoting obesity and health disparities. Curr. Obes.
Rep. 10, 39–52. doi: 10.1007/s13679-020- 00421-x
Spoelma, M. J., and Boakes, R. A. (2021). Sugar consumption and behavioural
inhibition in the rat. Appetite 159:105043. doi: 10.1016/j.appet.2020.105043
Frontiers in Neuroscience | www.frontiersin.org 14 June 2021 | Volume 15 | Article 670430
fnins-15-670430 June 4, 2021 Time: 14:29 # 15
Beecher et al. Sucrose Consumption and Neurocognitive Deficits
Stanhope, K. L. (2016). Sugar consumption, metabolic disease and obesity: the state
of the controversy. Crit. Rev. Clin. Lab. Sci. 53, 52–67. doi: 10.3109/10408363.
2015.1084990
Steele, C. C., Pirkle, J. R. A., and Kirkpatrick, K. (2017). Diet-induced impulsivity:
effects of a high-fat and a high-sugar diet on impulsive choice in rats. PLoS One
12:e0180510. doi: 10.1371/journal.pone.0180510
Stein, J. S., Renda, C. R., Barker, S. M., Liston, K. J., Shahan, T. A., and Madden,
G. J. (2015). Impulsive choice predicts anxiety-like behavior, but not alcohol
or sucrose consumption, in male long-evans rats. Alcohol. Clin. Exp. Res. 39,
932–940. doi: 10.1111/acer.12713
Stranahan, A. M., Norman, E. D., Lee, K., Cutler, R. G., Telljohann, R. S.,
Egan, J. M., et al. (2008). Diet-induced insulin resistance impairs hippocampal
synaptic plasticity and cognition in middle-aged rats. Hippocampus 18, 1085–
1088. doi: 10.1002/hipo.20470
Strimas, R., Davis, C., Patte, K., Curtis, C., Reid, C., and McCool, C. (2008).
Symptoms of attention-deficit/hyperactivity disorder, overeating, and body
mass index in men. Eat. Behav. 9, 516–518. doi: 10.1016/j.eatbeh.2008.07.005
Toth, L. A., and Gardiner, T. W. (2000). Food and water restriction protocols:
physiologicaland behavioral considerations. Contemp. Top Lab. Anim. Sci. 39,
9–17.
van der Borght, K., Köhnke, R., Göransson, N., Deierborg, T., Brundin, P.,
Erlanson-Albertsson, C., et al. (2011). Reduced neurogenesis in the rat
hippocampus following high fructose consumption. Regul. Pept. 167, 26–30.
doi: 10.1016/j.regpep.2010.11.002
Veenema, A. H., Bredewold, R., and Neumann, I. D. (2007). Opposite effects
of maternal separation on intermale and maternal aggression in C57BL/6
mice: link to hypothalamic vasopressin and oxytocin immunoreactivity.
Psychoneuroendocrinology 32, 437–450. doi: 10.1016/j.psyneuen.2007.02.008
Vollmayr, B., Bachteler, D., Vengeliene, V., Gass, P., Spanagel, R., and Henn,
F. (2004). Rats with congenital learned helplessness respond less to sucrose
but show no deficits in activity or learning. Behav. Brain Res. 150, 217–221.
doi: 10.1016/s0166-4328(03)00259- 6
Walf, A. A., and Frye, C. A. (2007). The use of the elevated plus maze as an
assay of anxiety-related behavior in rodents. Nat. Protoc. 2, 322–328. doi:
10.1038/nprot.2007.44
WHO (2019). Reducing Consumption of Sugar-Sweetened Beverages to Reduce the
Risk of Childhood Overweight and Obesity. Geneva: WHO.
Wong, A., Dogra, V. R., and Reichelt, A. C. (2017). High-sucrose diets in male rats
disrupt aspects of decision making tasks, motivation and spatial memory, but
not impulsivity measured by operant delay-discounting. Behav. Brain Res. 327,
144–154. doi: 10.1016/j.bbr.2017.03.029
Xu, T. J., and Reichelt, A. C. (2018). Sucrose or sucrose and caffeine
differentially impact memory and anxiety-like behaviours, and alter
hippocampal parvalbumin and doublecortin. Neuropharmacology 137,
24–32. doi: 10.1016/j.neuropharm.2018.04.012
Yankelevitch-Yahav, R., Franko, M., Huly, A., and Doron, R. (2015). The forced
swim test as a model of depressive-like behavior. J. Vis. Exp. JoVE 97, 52587.
doi: 10.3791/52587
Yoshida, Y., and Simoes, E. J. (2018). Sugar-sweetened beverage, obesity, and type
2 diabetes in children and adolescents: policies, taxation, and programs. Curr.
Diab. Rep. 18:31.
Yu, C.-J., Du, J. C., Chiou, H. C., Feng, C. C., Chung, M. Y., Yang, W., et al.
(2016). Sugar-sweetened beverage consumption is adversely associated with
childhood attention deficit/hyperactivity disorder. Int. J. Environ. Res. Public.
Health 13:678. doi: 10.3390/ijerph13070678
Zhao, C., Deng, W., and Gage, F. H. (2008). Mechanisms and functional
implications of adult neurogenesis. Cell 132, 645–660. doi: 10.1016/j.cell.2008.
01.033
Conflict of Interest: The authors declare that the research was conducted in the
absence of any commercial or financial relationships that could be construed as a
potential conflict of interest.
Copyright © 2021 Beecher,Alvarez Cooper, Wang, Walters, Chehrehasa, Bartlett and
Belmer. This is an open-access article distributed under the terms of the Creative
Commons Attribution License (CC BY). The use, distribution or reproduction in
other forums is permitted, provided the original author(s) and the copyright owner(s)
are credited and that the original publication in this journal is cited, in accordance
with accepted academic practice. No use, distribution or reproduction is permitted
which does not comply with these terms.
Frontiers in Neuroscience | www.frontiersin.org 15 June 2021 | Volume 15 | Article 670430
... Evidence in animal models indicates that overconsumption of free sugars can lead to molecular changes and cognitive impairment [25], particularly in hippocampal-dependent memory [12,26]. Long-term cohort studies and cross-sectional studies in humans have found significant associations between the consumption of added sugars, specifically sugar sweetened beverages (SSBs), and reduced cognitive function, poor memory performance, and higher risk of cognitive impairment [27,28]. ...
... While the Western diet is understood to have deleterious effects on physical, psychiatric, and neurophysiological function, the proportion of risk of disease or impairment attributed to any individual macronutrients is still debated [9,10]. A growing body of evidence from human and animal studies suggests that free and added sugars by themselves may be a significant modifiable risk factor for cognitive impairment [9,[11][12][13][14]. Free sugars refer to sugars that are found in honey, syrups, and juices or added in food preparation glucose were included. ...
... The 12 cross-sectional and cohort studies identified in the review found that chronic overconsumption of sugar negatively correlated with measures of global cognitive function, executive function, and memory. These results are similar to the findings of numerous animal studies that investigated the short-and long-term impacts of sugar consumption, finding alterations in neurophysiology and related impairment in cognitive function, specifically memory [12,25,26,120,121]. The present findings also identified two papers of mother-infant pairs that showed correlations between maternal dietary sugar intake and impairment in infant cognitive function. ...
Article
Full-text available
A relationship between excessive sugar consumption and cognitive function has been described in animal models, but the specific effects of sugars in humans remains unclear. This systematic review and meta-analysis aimed to evaluate the current knowledge, research characteristics, and quality of evidence of studies investigating the impacts of free and added sugars on human cognition in healthy participants. The review identified 77 studies (65 experimental trials, n = 3831; 9 cross-sectional studies, n = 11,456; and 3 cohort studies, n = 2059). All cohort studies and eight of the nine cross-sectional studies found significant positive correlations between added sugar consumption and risk of cognitive impairment. Four studies identified reduced risk of cognitive impairment associated with natural fructose-containing foods. The majority of randomised control trials assessed short-term glucose facilitation effects on cognitive outcomes. The results from these studies suggest the need for a tightly regulated blood glucose level, dependent on individualised physiological factors, for optimal cognitive function. A meta-analysis of a subset of studies that assessed the impact of glucose on recall found improvements in immediate free recall compared to controls (p = 0.002). The findings highlight the potentially detrimental effect of excessive, long-term, or prenatal added sugar consumption on cognitive function. Further research is needed to examine the specific effects of free and added sugars on cognitive function.
... The overconsumption of dietary fructose has been associated with the rise of chronic inflammatory diseases, such as obesity, diabetes, cardiovascular disease, and cancer (58-60). Furthermore, epidemiological studies have now revealed that high fructose consumption can also induce brain disturbances and negatively affect the development of neural system (61,62). Early studies that investigated the relationship between the effects of dietary fructose and neural functions have shown that microglia do express fructose transporter (GLUT5) (18) and that fructose metabolism in the brain was stimulated under hyperglycemic condition (50). ...
Article
Full-text available
The overconsumption of dietary fructose has been proposed as a major culprit for the rise of many metabolic diseases in recent years, yet the relationship between a high fructose diet and neurological dysfunction remains to be explored. Although fructose metabolism mainly takes place in the liver and intestine, recent studies have shown that a hyperglycemic condition could induce fructose metabolism in the brain. Notably, microglia, which are tissue-resident macrophages (Mφs) that confer innate immunity in the brain, also express fructose transporters (GLUT5) and are capable of utilizing fructose as a carbon fuel. Together, these studies suggest the possibility that a high fructose diet can regulate the activation and inflammatory response of microglia by metabolic reprogramming, thereby altering the susceptibility of developing neurological dysfunction. In this review, the recent advances in the understanding of microglia metabolism and how it supports its functions will be summarized. The results from both in vivo and in vitro studies that have investigated the mechanistic link between fructose-induced metabolic reprogramming of microglia and its function will then be reviewed. Finally, areas of controversies and their associated implications, as well as directions that warrant future research will be highlighted.
Article
Full-text available
The Carter Center has estimated that the addiction crisis in the United States (US), if continues to worsen at the same rate, may cost the country approximately 16 trillion dollars by 2030. In recent years, the well-being of youth has been compromised by not only the coronavirus disease 2019 pandemic but also the alarming global opioid crisis, particularly in the US. Each year, deadly opioid drugs claim hundreds of thousands of lives, contributing to an ever-rising death toll. In addition, maternal usage of opioids and other drugs during pregnancy could compromise the neurodevelopment of children. A high rate of DNA polymorphic antecedents compounds the occurrence of epigenetic insults involving methylation of specific essential genes related to normal brain function. These genetic antecedent insults affect healthy DNA and mRNA transcription, leading to a loss of proteins required for normal brain development and function in youth. Myelination in the frontal cortex, a process known to extend until the late 20s, delays the development of proficient executive function and decision-making abilities. Understanding this delay in brain development, along with the presence of potential high-risk antecedent polymorphic variants or alleles and generational epigenetics, provides a clear rationale for embracing the Brain Research Commission’s suggestion to mimic fitness programs with an adaptable brain health check (BHC). Implementing the BHC within the educational systems in the US and other countries could serve as an effective initiative for proactive therapies aimed at reducing juvenile mental health problems and eventually criminal activities, addiction, and other behaviors associated with reward deficiency syndrome.
Article
The systemic effects of the artificial sweetener sorbitol on older adult individuals have not been elucidated. We assessed the effects of sorbitol consumption on cognitive and gingival health in a mouse model. Aged mice were fed 5% sorbitol for 3 months before their behavior was assessed, and brain and gingival tissues were collected. Long‐term sorbitol consumption inhibited gingival tissue aging in aged mice. However, it caused cognitive decline and decreased brain‐derived neurotrophic factor (BDNF) in the hippocampus. Sorbitol consumption did not affect homeostatic function; however, it may exert effects within the brain, particularly in the hippocampus.
Article
High Fructose Corn Syrup (HFCS) and Fructose (FR) are widely used sweeteners in many foods and beverages. This study aimed at investigating the cytotoxic effects of HFCS (5%-30%) and FR (62.5-2000 μg/mL) using MTT assay in Human Hepatocellular Carcinoma (HepG2) cells, and genotoxic effects of using Chromosome Aberrations (CAs), Sister Chromatid Exchanges (SCEs), Micronuclei (MN) and comet assays in human lymphocytes. HFCS significantly reduced the cell viability in HepG2 cells at between 7.5% and 30% for 24 and 48 h. 30% HFCS caused a very significant toxic effect. FR had a cytotoxic effect in HepG2 cells at all treatments. However, as fructose concentration decreased, the cell viability decreased. HFCS (10%-20%) and FR (250-2000 μg/mL) decreased the mitotic index at higher concentrations. IC50 value was found to be a 15% for 48 h. IC50 value of FR was detected as 62.5 μg/mL for 24 h and 48 h. HFCS significantly increased CAs frequency at 15% and 20%. FR significantly increased the frequency of CAs at 250, 1000, and 2000 μg/mL for 48 h. Both sweeteners increased the frequency of SCEs at all concentrations. HFCS (15% and 20%) and FR (250, 1000, and 2000 μg/mL) induced MN frequency at higher concentrations. HFCS caused DNA damage in comet assay at 10% -30%. FR increased tail intensity and moment at 125-2000 μg/mL and tail length at 62.5, 250 and 500 μg/mL. Therefore, HFCS and FR are clearly seen to be cytotoxic and genotoxic, especially at higher concentrations.
Article
BACKGROUND: Research has shown that excessive sugar-sweetened beverage (SSB) consumption has been associated with being overweight and obese and the military is not immune to this disorder. Being overweight/obese is one of the characteristics that comprise a condition known as Metabolic Syndrome (MetS). The global prevalence of MetS in the military population is estimated to be 21%, which varies depending upon the armed forces type and specialty. The purpose of this study is to identify the subset of the aviation population that may develop MetS at some future point and to put forth suggestions on how best to combat this and maintain operational readiness. METHODS: Data were collected by means of an anonymous 44-item survey administered and completed by 302 students enrolled in Aviation Preflight Indoctrination at the Naval Air Station Base in Pensacola, FL. RESULTS: Results indicated that 70.86% of students reported SSB consumption, with 95.3% preferring caffeinated to decaffeinated SSBs. 11.7% of personnel met the “at risk criteria” of consuming SSBs four times per week or greater (four 12-oz cans or more per week). DISCUSSION: Findings from this study indicate that approximately 11.7% of aviation pilots or flight officers are “at risk” of developing MetS. Given that it takes up to 4 yr to train a new fighter pilot, this could become a significant readiness issue on par with the impact seen with pilots leaving the services through natural attrition.
Article
Excessive consumption of sugary drinks negatively impacts the developing brain, producing long‐lasting behavioral and metabolic disorders. Here, we study whether treatment with the antihyperglycemic agent metformin prevents some of the anxiety and memory alterations produced by chronic sucrose consumption. Male Sprague–Dawley rats had unrestricted access to water (control group) and a bottle containing a 10% sucrose solution (sucrose group, SUC) for 35 days. In parallel, a group of animals from SUC received metformin (25 mg/kg or 50 mg/kg, orally; MET 25 and MET 50 groups, respectively). After 2 weeks of metformin treatment, the animals weighed less than controls. SUC and MET 50 groups compensated for the caloric intake from the sugary solution by consuming less chow. In contrast, total energy intake in MET 25 was higher than the rest of the groups, but they still weighed less than control and SUC groups, suggesting that at this concentration, metformin delays body growth. The animals were then tested for the open field (OF), elevated plus maze (EPM) and novel object location (NOL) tests. In the OF, SUC animals spent more time in the central zone of the arena, evidenced by an increased number of entries and the distance traveled there. In the EPM, SUC animals spent more time in the open arms and less time in the central square. Metformin treatment prevented the decreased anxiety observed in SUC animals in the OF and EPM. In the NOL test, SUC animals showed less interest in novelty and metformin treatment did not improve this alteration. The preference for open spaces in the OF and EPM were associated with increased serum triglycerides (TG) and malondialdehyde levels in the medial prefrontal cortex (mPFC) and the hippocampus (HIP), while poor memory performance was associated with high basal blood glucose levels. In conclusion, the decreased anxiety‐like behavior produced by chronic sucrose consumption was prevented by metformin treatment, through a mechanism that probably involves normalization of TG levels and decreased oxidative stress in mPFC and HIP.
Article
Adolescence, a critical period of developmental period, is marked by neurobiological changes influenced by environmental factors. Here, we show how exposure to sucrose, which is ubiquitously available in modern diets, results in changes in behavioural response to cocaine as an adult. Rats were given daily access to either 10% sucrose or water during the adolescent period (PND28–42). Following this period, rats are left undisturbed until they reach adulthood. In adulthood, rats were tested for (i) acquisition of a low dose of cocaine, (ii) progressive ratio (PR) test, and (iii) resistance to punished cocaine taking. Sucrose exposure resulted in significant alterations in all behavioural measures. To determine the neurobiological mechanisms leading to such behavioural adaptations, we find that adolescent sucrose exposure results in an upregulation of the transcription factor Smad3 in the nucleus accumbens (NAc) when compared with water‐exposed controls. Transiently blocking the active form of this transcription factor (HSV‐dnSmad3) during adolescence mitigated the enhanced cocaine vulnerability‐like behaviours observed in adulthood. These findings suggest that prior exposure to sucrose during adolescence can heighten the reinforcing effects of cocaine. Furthermore, they identify the TGF‐beta pathway and Smad3 as playing a key role in mediating enduring and long‐lasting adaptations that contribute to sucrose‐induced susceptibility to cocaine. Taken together, these results have important implications for development and suggest that adolescent sucrose exposure may persistently enhance the susceptibility to substance abuse.
Article
Full-text available
Purpose of Review The mechanistic role of sugar-sweetened beverage (SSB) in the etiology of obesity is undetermined. We address whether, compared to other foods, does consumption of SSB (1) automatically lead to failure to compensate for the energy it contains? (2) fail to elicit homeostatic hormone responses? (3) promote hedonic eating through activation of the brain’s reward pathways? We followed the evidence to address: (4) Would restriction of targeted marketing of SSB and other unhealthy foods to vulnerable populations decrease their prevalence of obesity? Recent Findings The data are lacking to demonstrate that SSB consumption promotes body weight gain compared with isocaloric consumption of other beverages or foods and that this is linked to its failure to elicit adequate homeostatic hormone responses. However, more recent data have linked body weight gain to reward activation in the brain to palatable food cues and suggest that sweet tastes and SSB consumption heightens the reward response to food cues. Studies investigating the specificity of these responses have not been conducted. Nevertheless, the current data provide a biological basis to the body of evidence demonstrating that the targeted marketing (real life palatable food cues) of SSB and other unhealthy foods to vulnerable populations, including children and people of color and low socioeconomic status, is increasing their risk for obesity. Summary While the mechanisms for the association between SSB consumption and body weight gain cannot be identified, current scientific evidence strongly suggests that proactive environmental measures to reduce exposure to palatable food cues in the form of targeting marketing will decrease the risk of obesity in vulnerable populations.
Article
Full-text available
The nucleus of the solitary tract (NTS) is critical for the central integration of signals from visceral organs and contains preproglucagon (PPG) neurons, which express leptin receptors in the mouse and send direct projections to the paraventricular nucleus of the hypothalamus (PVH). Here, we visualized projections of PPG neurons in leptin-deficient Lep ob/ob mice and found that projections from PPG neurons are elevated compared with controls, and PPG projections were normalized by targeted rescue of leptin receptors in LepRb TB/TB mice, which lack functional neuronal leptin receptors. Moreover, Lep ob/ob and LepRb TB/TB mice displayed increased levels of neuronal activation in the PVH following vagal stimulation, and whole-cell patch recordings of GLP-1 receptor-expressing PVH neurons revealed enhanced excitatory neurotransmission, suggesting that leptin acts cell autonomously to suppress representation of excitatory afferents from PPG neurons, thereby diminishing the impact of visceral sensory information on GLP-1 receptor-expressing neurons in the PVH.
Article
Full-text available
The metabolic effects of sugary drinks have been extensively studied, whereas the effects on psychological processes have received relatively limited attention. Several studies have found that high-sugar diets can produce impaired performance by rats on tests assessing spatial learning and memory. In contrast, despite claims that weakened inhibitory control underlies many sugar-induced deficits, evidence supporting this proposal has been limited. The aim of the present study was to assess the impact of high-sugar diets on response inhibition, as measured by rats’ performance on a differential reinforcement of low rates schedule (DRL) in Experiments 1 and 2 and on a Pavlovian discrimination reversal task in Experiment 3. In all three experiments a 30-day diet stage, in which Sugar groups were given unrestricted access to 10% sucrose solution and Control groups had access to water only, was followed by behavioural tests. In Experiment 1 the Sugar group performed poorly on a spatial memory task, but no difference was detected between the performances of the two groups in the DRL test. In Experiment 2 longer DRL training was given and post-diet performance was assessed both before and after access to sugar was withdrawn. Null results were obtained under both conditions. In Experiment 3 rats’ performance on a discrimination learned prior to the diet intervention was not affected by the high-sugar diet, but neither was performance once the discrimination was reversed. The implications of these results for understanding of sugar-induced psychological deficits are discussed.
Article
Full-text available
Associations between adult attention-deficit/hyperactivity disorder (ADHD) symptoms and dietary habits have not been well established and the underlying mechanisms remain unclear. We explored these associations using a Swedish population-based twin study with 17,999 individuals aged 20-47 years. We estimated correlations between inattention and hyperactivity/impulsivity with dietary habits and fitted twin models to determine the genetic and environmental contributions. Dietary habits were defined as (a) consumption of food groups, (b) consumption of food items rich in particular macro-nutrients, and (c) healthy and unhealthy dietary patterns. At the phenotypic level, inattention was positively correlated with seafood, high-fat, high-sugar, high-protein food consumptions, and unhealthy dietary pattern, with correlation coefficients ranging from 0.03 (95%CI: 0.01, 0.05) to 0.13 (95% CI: 0.11, 0.15). Inattention was negatively correlated with fruits, vegetables consumptions and healthy dietary pattern, with correlation coefficients ranging from −0.06 (95%CI: −0.08, −0.04) to −0.07 (95%CI: −0.09, −0.05). Hyperactivity/impulsivity and dietary habits showed similar but weaker patterns compared to inattention. All associations remained stable across age, sex and socioeconomic status. Nonshared environmental effects contributed substantially to the correlations of inattention (56-60%) and hyperactivity/impulsivity (63-80%) with dietary habits. The highest and lowest genetic correlations were between inattention and high-sugar food (r A = .16, 95% CI: 0.07, 0.25), and between hyperactivity/impulsivity and unhealthy dietary pattern (r A = .05, 95% CI: −0.05, 0.14), respectively. We found phenotypic and etiological overlap between ADHD and dietary habits, although these associations were weak. Our findings contribute to a better understanding of common etiological pathways between ADHD symptoms and various dietary habits. K E Y W O R D S
Article
Full-text available
Background Attention-deficit/hyperactivity disorder (ADHD) is a significant neurobehavioral disorder in children and adolescence which may be affected by diet. Objective To evaluate the possible relationship between sugar consumption and the development of symptoms of ADHD. Methods In March 2020, an exhaustive systematic literature search was conducted using Google Scholar, PubMed, and Scopus. In this meta-analysis of observational studies, odds ratios, relative risks, hazard ratios, and their 95% confidence intervals, which was reported for ADHD regarding SSBS, soft drink consumption, and dietary sugars, were used to calculate ORs and standard errors. At first, a fixed-effects model was used to drive the overall effect sizes using log ORs and SEs. If there was any significant between-studies heterogeneity, the random-effects model was conducted. Cochran’s Q test and I² were used to measure potential sources of heterogeneity across studies. The Newcastle-Ottawa scale was used to assess the quality of the included articles. Results Seven studies, two cross-sectional, two case-control, and three prospective with a total of 25,945 individuals were eligible to include in the current meta-analysis. The association between sugar and soft drink consumption and the risk of ADHD symptoms were provided based on the random-effects model (pooled effect size: 1.22, 95%CI: 1.04-1.42, P = 0.01) (I² = 81.9%, P heterogeneity< 0.0001). Conclusion This meta-analysis indicated a positive relationship between overall sugar and sugar-sweetened beverages consumption and symptoms of ADHD; however, there was heterogeneity among included studies. Future well-designed studies that can account for confounds are necessary to confirm the effect of sugar on ADHD.
Article
Full-text available
Individuals with post-traumatic stress disorder (PTSD) often use alcohol to cope with their distress. This aberrant use of alcohol often develops into alcohol use disorder (AUD) leading to high rates of PTSD-AUD co-occurrence. Individuals with comorbid PTSD-AUD have more intense alcohol cravings and increased relapse rates during withdrawal than those with AUD alone. Also, individuals with PTSD or AUD alone often show similar psychological behaviors, such as impulsivity and anhedonia. Extensive clinical studies on the behavioral effects of PTSD-AUD comorbidity, namely alcohol use, have been performed. However, these effects have not been well studied or mechanistically explored in animal models. Therefore, the present study evaluated the effects of traumatic stress comorbid with alcohol exposures on ethanol intake, impulsivity, and anhedonia in mice. Adult male C57Bl/6 mice were first exposed to either mouse single-prolonged stress (mSPS), an animal model that has been validated for characteristics akin to PTSD symptoms, or control conditions. Baseline two-bottle choice ethanol consumption and preference tests were conducted after a 7-day isolation period, as part of the mSPS exposure. Next, mice were exposed to air or chronic intermittent ethanol (CIE), a vapor-induced ethanol dependence and withdrawal model, for 4 weeks. Two-bottle choice ethanol drinking was used to measure dependence-induced ethanol consumption and preference during periods intervening CIE cycles. The novelty suppressed feeding (NSF) test was used to evaluate impulsivity and anhedonia behaviors 48 h after mSPS and/or repeated CIE exposure. Results showed that, compared to control conditions, mSPS did not affect baseline ethanol consumption and preference. However, mSPS-CIE mice increased Post-CIE ethanol consumption compared to Control-Air mice. Mice exposed to mSPS had a shorter latency to feed during the NSF, whereas CIE-exposed mice consumed less palatable food reward in their home cage after the NSF. These results demonstrate that mice exposed to both mSPS and CIE are more vulnerable to ethanol withdrawal effects, and those exposed to mSPS have increased impulsivity, while CIE exposure increases anhedonia. Future studies to examine the relationship between behavioral outcomes and the molecular mechanisms in the brain after PTSD-AUD are warranted.
Article
Full-text available
Working memory (WM) impairments in ADHD have been consistently reported along with deficits in attentional control. Yet, it is not clear which specific WM processes are affected in this condition. A deficient coupling between attention and WM has been reported. Nevertheless, most studies focus on the capacity to retain information rather than on the attention-dependent stages of encoding and retrieval. The current study uses a visual short-term memory binding task, measuring both behavioral and electrophysiological responses to characterize WM encoding, binding and retrieval comparing ADHD and non-ADHD matched adolescents. ADHD exhibited poorer accuracy and larger reaction times than non-ADHD on all conditions but especially when a change across encoding and test displays occurred. Binding manipulation affected equally both groups. Encoding P3 was larger in the non-ADHD group. Retrieval P3 discriminated change only in the non-ADHD group. Binding-dependent ERP modulations did not reveal group differences. Encoding and retrieval P3 were significantly correlated only in non-ADHD. These results suggest that while binding processes seem to be intact in ADHD, attention-related encoding and retrieval processes are compromised, resulting in a failure in the prioritization of relevant information. This new evidence can also inform recent theories of binding in visual WM.
Article
Full-text available
Long-term binge alcohol consumption alters the signaling of numerous neurotransmitters in the brain including noradrenaline (NE) and serotonin (5-HT). Alterations in the signaling of these neuronal pathways result in dysfunctional emotional states like anxiety and depression which are typically seen during alcohol withdrawal. Interestingly, studies have demonstrated that the development of alcohol-induced negative affective states is linked to disrupted neurogenesis in the dentate gyrus (DG) region of the hippocampus in alcohol-dependent animals. We have previously shown that modulation of NE and 5-HT activity by pharmacological targeting of β-adrenoreceptors (β-ARs) and 5-HT1A/1B receptors with pindolol reduces consumption in long-term alcohol-consuming mice. Since these receptors are also involved in emotional homeostasis and hippocampal neurogenesis, we investigated the effects of pindolol administration on emotional and neurogenic deficits in mice consuming long-term alcohol (18 weeks). We report that acute administration of pindolol (32 mg/kg) reduces anxiety-like behavior in mice at 24 h withdrawal in the marble-burying test (MBT) and the elevated plus-maze (EPM). We also show that chronic (2 weeks) pindolol treatment (32 mg/kg/day) attenuates alcohol-induced impairments in the density of immature neurons (DCX+) but not newborn cells (BrdU+) in the hippocampal DG. Pindolol treatment also restores the normal proportion of newborn proliferating cells (BrdU+/Ki67+/DCX−), newborn proliferating immature neurons (BrdU+/Ki67+/DCX+) and newborn non-proliferating immature neurons (BrdU+/Ki67−/DCX+) following long-term alcohol intake. These results suggest that pindolol, through its unique pharmacology may rescue some but not all deficits of long-term alcohol abuse on the brain, adding further value to its properties as a strong pharmaceutical option for alcohol use disorders (AUDs).
Article
Full-text available
This narrative review is aimed at presenting the most recent evidence on the association between attention-deficit/hyperactivity disorder (ADHD) and obesity. The review is informed by previous relevant systematic reviews and a search in Pubmed and PsycINFO up to 1 August 2019. Although the association between ADHD and obesity would seem, at first, paradoxical, in the past two decades there has been an increasing number of studies on this topic. The present review shows that there is meta-analytic evidence supporting a significant association between these two conditions, at least in adults. Growing evidence is also being published on the genetic and environmental factors underlying the association. However, the cause–effects paths, as well as the exact mechanisms explaining the association, remain unclear. Additionally, empirical evidence guiding the management/treatment of patients with the two conditions is still limited. Therefore, after almost 20 years from the first report of a link between ADHD and obesity, this association continues to be puzzling.
Article
We consider some of the problems involved in current discussions on stem cells in adult mammalian tissues. The present concepts involve a number of pitfalls, weaknesses and logical, semantic and classification problems. This indicates the necessity for new and well-defined concepts that are amenable to experimental analysis. One of the major difficulties in considering stem cells is that they are defined in terms of their functional capabilities which can only be assessed by testing the abilities of the cells, which itself may alter their characteristics during the assay procedure: a situation similar to the uncertainty principle in physics. The terms that describe stem cell functions are often not well defined and are used loosely, which can lead to confusion. If such context-dependent interactions exist between the manipulation and measurement process and the challenged stem cells, the question of, for example, the number of stem cells, in a tissue has to be posed in a new way. Rather than obtaining a single number one might end up with various different numbers under different circumstances, all being complementary. This might suggest that stemness is not a property but a spectrum of capabilities from which to choose. This concept might facilitate a reconciliation between the different and sometimes opposing experimental results. Given certain experimental evidence, we have attempted to provide a novel concept to describe structured cell populations in tissues involving stem cells, transit cells and mature cells. It is based on the primary assumption that the proliferation and differentiation/maturation processes are in principle independent entities in the sense that each may proceed without necessarily affecting the other. Stem cells may divide without maturation while cells approaching functional competence may mature but do not divide. In contrast, transit cells divide and mature showing intermediate properties between stem cells and mature functional cells. The need to describe this transition process and the variable coupling between proliferation and maturation leads us to formulate a spiral model of cell and tissue organisation. This concept is illustrated for the intestinal epithelium. It is concluded that the small intestinal crypts contain 4–16 actual stem cells in steady state but up to 30–40 potential stem cells (clonogenic cells) which may take over stem cell properties following perturbations. This implies that transit cells can under certain circumstances behave like actual stem cells while they undergo maturation under other conditions. There is also evidence that the proliferation and differentiation/maturation processes are subject to controls that ultimately lead to a change in the spiral trajectories.(ABSTRACT TRUNCATED AT 400 WORDS)