Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

Contents lists available at ScienceDirect

Neuroscience and Biobehavioral Reviews


journal homepage: www.elsevier.com/locate/neubiorev

The impact of sugar consumption on stress driven, emotional and addictive T


behaviors
Angela Jacquesa,b, Nicholas Chaayaa,b, Kate Beechera,b, Syed Aoun Alia,b, Arnauld Belmera,b,1,
Selena Bartletta,b, ,1

a
School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
b
Institute of Health and Biomedical Innovation at Translational Research Institute, Brisbane, Australia

ARTICLE INFO ABSTRACT

Keywords: In 2016 the World Health Organization reported 39% of the world’s adult population (over 18 y) was over-
Sucrose consumption weight, with western countries such as Australia and the United States of America at 64.5% and 67.9% re-
Stress spectively. Overconsumption of high fat/sugar containing food and beverages contribute to the development of
Anxiety obesity. Neural plasticity that occurs as a result of long term sugar consumption has been shown to reduce
Depression
impulse control and therefore lower the ability to resist the high fat/sugar foods contributing to the obesity
Fear
epidemic. There is significant overlap between the neural pathways involved in emotions that guide behavioural
Obesity
Addiction responses to survival situations with those regulating overconsumption of highly palatable food. This suggests
Emotion that having a clearer understanding of the role of stress and emotions in the development of obesity will lead to
Behaviour the development of novel therapeutic strategies. Sucrose consumption activates the mesocorticolimbic system in
a manner synonymous with substances of abuse. There is overwhelming evidence to support the hypothesis that
sucrose consumption results in pathophysiological consequences such as morphological neuronal changes, al-
tered emotional processing and modified behaviour in rodent and human models. In this comprehensive review,
we examined > 300 studies investigating the interaction between sugar consumption, stress and emotions.
Preclinical and clinical trials investigating highly palatable foods and stress, anxiety, depression and fear are
reviewed. Importantly, the synergy between sugar consumption and neurobiology is addressed. This review
summarizes the neurochemical changes and neural adaptations ö including changes in the dopaminergic system
ö that influence emotion and behaviour following sugar consumption.

1. Introduction health (Peet, 2004). For example, a diet higher in refined sugar has
been shown to predict a worsening of schizophrenic behaviour over a
A sedentary lifestyle combined with a high caloric diet plays a sig- two year period (Peet, 2004). Despite the many psychological, physical
nificant role in obesity (Organization, W.H., 2017). In 2016 the World and neurological burdens of sugar overconsumption and consequent
Health Organization reported more than 1.9 billion adults were over- obesity, there are no therapies directed at reducing sugar consumption,
weight (Organization, W.H., 2016). World obesity has essentially tri- and few therapies capable of successfully treating obesity (for review
pled since 1975 (Organization, W.H., 2016). Excessive sugar con- see (Kanoski and Davidson, 2011)).
sumption has been shown to be one of the leading contributors to Obesity, arising from overconsumption of rewarding foods such as
weight gain (Malik et al., 2013). Furthermore, a diet high in sugars has those with a high sugar content, may result in negative consequences,
been linked to cognitive impairments, negative neuroplasticity changes such as a loss of self-control and subsequent poor decision-making
such as hippocampal dysfunction (Kanoski and Davidson, 2011; Noble (Beilharz et al., 2014). This loss of control poses a significant challenge
et al., 2017) and emotional disorders such as anxiety and depression for overweight individuals attempting to lose weight. The desire to eat,
(Kim et al., 2018). High sugar intake increases the risk of cancer, oxi- is regulated by brain regions known as feeding centers, located in the
dative stress, inflammation, and obesity (Makarem et al., 2018), as well arcuate nucleus of the hypothalamus. Importantly, these regions are
as impacting cognitive function (Barnes and Joyner, 2012) and mental interconnected with the limbic system and cerebral cortex (specifically


Corresponding author at: School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia.
E-mail address: selena.bartlett@qut.edu.au (S. Bartlett).
1
These authors share equal senior authorship.

https://doi.org/10.1016/j.neubiorev.2019.05.021
Received 7 November 2018; Received in revised form 14 March 2019; Accepted 19 May 2019
Available online 21 May 2019
0149-7634/ © 2019 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/BY/4.0/).
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

Fig. 1. Reward pathway encompassing the mesocorticolimbic distribution of dopaminergic neurons. The neural regions of the reward pathway include the prefrontal
cortex (PFC), amygdala (AMG), ventral tegmental area (VTA) and nucleus accumbens (NAc). Each anatomical region modulates individual behaviours and con-
tributes to general behaviours through cross-connectivity. Regions shown and behaviours listed are consistent between human and rodent brains (Belin and Dalley,
2012).

the hippocampus and amygdala), which are responsible for the mod- regulated by the hypothalamic-pituitary-adrenal (HPA) axis. Interest-
ulation of emotions (Sweeney and Yang, 2017; Liu et al., 2014; ingly, activity of the HPA axis has been shown to be reduced through
Stunkard et al., 2003; Simon et al., 2006; Ulrich-Lai et al., 2010; Ahima the consumption of sugar containing foods (Ulrich-Lai et al., 2011).
and Antwi, 2008). Following consumption, hormones are released to reduce the feelings of
stress, which also increase the desire for comfort foods, thus perpetu-
1.1. Sugar, artificial sweeteners and obesity ating emotional eating habits (Ulrich-Lai et al., 2011; Ursano et al.,
2009).
Sugar became embedded in the food chain in the late 1960s and The objective of this review is to summarize research that examines
replaced fats to mask bitterness and make food more palatable (Bakke how the consumption of sugar leads to changes in neurobiological brain
et al., 2018; Moss, 2013). In the 1970’s a shift toward increased sugar- function that alters emotional states and subsequent behaviours. We
sweetened beverages became apparent (Wolf et al., 2008). Our early will examine findings from studies at the intersection between the
ancestors obtained sugar from either fruit, limited by seasons, or honey, consumption of sucrose and changes in the performance of tasks with a
protected by bees. In the last half century, sugar consumption has tri- stressful or emotional component and review neurobiological and
pled worldwide, partially due to the hidden use of added sugars in neurochemical mechanisms involved in addiction, stress, fear, anxiety
processed food (Lustig et al., 2012). The first artificial sweetener (sac- and depression to determine whether there are overlapping neural
charin) was introduced in 1879 with low production costs during mechanisms. Lastly, we will determine whether there are novel phar-
wartime, increasing its popularity (Weihrauch and Diehl, 2004). In the macotherapeutics and/or interventions that target these brain circuits
1950s as sugar became readily available, the use of sweeteners shifted or neurochemical pathways to improve the current approaches for the
to so-called ‘diet products’ with low caloric content. treatment of obesity.
The development of obesity relies on both the hedonic, sweet taste
of food in conjunction with the negative emotional properties of food 2. Common neuronal pathways for sucrose consumption,
consumption (Meye and Adan, 2014; Yu and Kim, 2012; Berthoud addiction, emotions and obesity
et al., 2017). Hedonic reactions to a 10% sucrose solution (and as low as
3.4%) were tested and found to be significantly higher in adolescent Addiction is characterized by a difficulty to control habitual beha-
rats when compared to adult rats (Wilmouth and Spear, 2009). viour even in the face of negative consequences (Lindgren et al., 2018).
Early addiction research focused on drugs of abuse such as alcohol,
1.2. Sugar and emotions morphine and nicotine. This has since been extended to include gam-
bling, eating and more recently, sugar consumption (Comings et al.,
Emotional eating has been show to stem from the desire to mitigate 2001; Weeks, 1962; Jellinek, 1952; Benowitz, 1988; Gearhardt et al.,
the effects of stress (Van Strien et al., 1986) and stress is partially 2009; Klenowski et al., 2016; Benton, 2010). Addiction to substances of

179
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

abuse relies on the drug binding to specific protein targets which elicits thus play a role in driving the compulsive over-consumption of food
certain physiological and behavioural responses unique to that drug (Derman and Ferrario, 2017; Berridge et al., 2010). Similarly, con-
(Lee and Messing, 2008). Psychoactive drugs commonly result in re- sumption of a 10% sucrose solution resulted in alterations in synaptic
warding sensations that lead to repeated use and, depending on genetic strength via AMPA receptor trafficking, which modulates the compul-
susceptibility, environmental factors and subsequent addiction (Lee and sive tendencies characteristic of any drug-seeking (Peng et al., 2011).
Messing, 2008). Stress has long been associated with both the motiva- Drug induced neuroplasticity has been observed in the NAc through
tion to use rewarding substances and the result of not attaining those alterations in dendritic morphology and altered gene expression
substances (for review see (Sinha, 2008)). The negative symptoms (Huang et al., 2018; Legastelois et al., 2014; Li et al., 2004). Similar to
produced through withdrawal are common to all forms of addictive psychoactive substances, sugar binging has been shown to cause re-
substances including highly palatable food. These include prolonged peated increases in dopamine release and altered expression of NAc
sensitization to the substance of choice and associative learning where Delta FosB as demonstrated in rats with an increase in Delta Fos B
environmental cues become associated with the pleasure derived from expression after consumption of a high sugar diet (Avena et al., 2006;
the substance (Hebebrand et al., 2014). These associative memories, Wallace et al., 2008). Dopamine release increases as a direct effect of
combined with intense cravings, increase the incidence of relapse even chronic drug use and results in postsynaptic changes of Delta FosB and
after sustained abstinence. These commonalities may be due to sub- CREB accumulation (Nestler and Aghajanian, 1997). The intracellular
stances of addiction utilizing the same circuitry within the brain’s build-up of Delta FosB can alter the gene production of receptors which
mesocorticolimbic system (Fig. 1) (Volkow et al., 2004; Koob and Le may result in reinstatement during withdrawal (Nestler and
Moal, 2001; Baron et al., 2018). Aghajanian, 1997).This data suggest that neuroadaptations in the brain
Food consumption is necessary to regulate homeostatic energy reward pathway in obese subjects may contribute to the progression of
balance. However, humans also consume food for pleasure or comfort. compulsive eating. The NAc is considered to be the main region to
The hedonistic desire for palatable food is considered reward-related undergo neuroadaptation after sugar consumption, but changes have
and may result in maladaptive or negative neuroplasticity that can been noted in many brain regions encompassing the mesocorticolimbic
override homeostatic regulation and result in overeating behaviours system (De Jong et al., 2016).
(Kenny, 2011). Reward is delivered through stimuli that produce
pleasurable or enjoyable experiences, contrasting with addiction, which 2.2. Amygdala
involves compulsive and sometimes painful behaviour, yet both share
similar neuroadaptive responses and overlapping neuronal pathways The AMG plays a key role in negative reinforcement, the progres-
(Adinoff, 2004). The reward or mesocorticolimbic pathway plays an sion towards addiction and extensively in the learned associations that
influential role in what we choose to eat as demonstrated by studies lead to relapse (See et al., 2003). Neuroplastic changes occur in the
where rats appear willing to endure noxious stimuli such as extreme AMG which facilitate the level of dependence to a substance of abuse to
cold, heat and foot-shock to procure highly palatable foods over stan- move from impulsivity to compulsion (Koob, 2009a). This was de-
dard rat chow (Cabanac and Johnson, 1983; Foo and Mason, 2005; monstrated by long term sucrose consumption in rats which produced
Oswald et al., 2011). maladaptive alterations in the apical dendritic morphology of AMG
The reward pathway consists of the prefrontal cortex (PFC), principle neurons (Shariff et al., 2017). Optogenetic stimulation of
amygdala (AMG), ventral-tegmental area (VTA) and nucleus ac- central AMG neurons was paired with a sucrose reward to test reward
cumbens (NAc) and in accord with drugs of abuse (Sinha, 2018; Alsiö incentive in rats (Robinson et al., 2014). Stimulation increased in-
et al., 2012), is thought to be stimulated by the overconsumption of centive to choose the sucrose reward over a similar sweet alternative
sugar, thus contributing to the development of obesity (Leigh and and amplified the degree of effort the rats were willing to exert to ob-
Morris, 2016; Murray et al., 2016; Shariff et al., 2016). The reward tain the reward (Robinson et al., 2014). These findings suggest neuro-
pathway is highly associated with the efflux of dopamine which reg- plastic changes in the AMG may occur due to sucrose consumption and
ulates the motivational state of wanting or craving that substance or increased addictive-like compulsive behaviour. The AMG and PFC share
behaviour (Adinoff, 2004). Once overstimulated, the pathway becomes a role in motivation, associative learning (including negative re-
primed to require that particular stimuli when presented with con- inforcement) (Koob, 2009a), compulsive behaviour and deficits in ex-
textual cues or emotional stress (Adinoff, 2004). The VTA induces this ecutive functioning (Zehra et al., 2018). Relapsing into addictive be-
sensitization while the NAc modulates its expression through dopami- haviours is most likely due to dysregulation of motivational processes.
nergic control (Huang et al., 2018; Xiong et al., 2018; Zhang et al., Negative reinforcement, or substance abuse to alleviate an hedonic,
2018a). anxious, irritable or dysphoric state, is characteristic of addiction
withdrawal (Koob, 2018).
2.1. Nucleus accumbens
2.3. Prefrontal cortex
The NAc consists of two sub regions (core and shell), each con-
taining specific neuronal populations fundamental in processes of mo- The orbitofrontal cortex and anterior cingulate are associated with
tivation (Yang et al., 2018; Reker et al., 2018), aversion (Rosas et al., compulsive cravings for drugs of abuse (Adinoff, 2004). Interestingly,
2018), fear related avoidance (Moscarello and Maren, 2018), re- neurons in the orbitofrontal cortex of rats were shown to encode this
inforcement learning (Floresco et al., 2018; Anderson et al., 2018), same compulsion to seek a sucrose reward solution (15%) (Moorman
pleasure seeking, addiction (Kim et al., 2018; Lam and Jadavji, 2018; and Aston-Jones, 2014). The PFC is the brain region responsible for
Barrientos et al., 2018) and behavioural sensitization (Nona and executive functions such as planning and decision making. Addiction
Nobrega, 2018). To examine motivation and the role played by α- and periods of withdrawal from substances of abuse are generally ac-
amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) re- companied by a loss of executive function, which is due to dysfunc-
ceptors in the NAc, intra-NAc infusions of AMPAR antagonists were tional neurocircuitry in the medial PFC (Molnar et al., 2018). Sucrose
given to obesity prone and obesity resistant male rats (Derman and and fructose consumption have also been linked to metabolic and
Ferrario, 2017). Compared with controls, findings showed the obesity- electrophysiological changes in the hippocampus (Hsu et al., 2015;
prone animals exhibited cue-triggered food seeking and the behaviour Lemos et al., 2016), the thalamus (Jastreboff et al., 2016) and the hy-
was mediated by increased surface expression of AMPA receptors in the pothalamus (Berthoud et al., 2017; Ulrich-Lai et al., 2011); brain re-
core of the NAc. This data suggests cue-triggered food seeking may gions involved in the associative learning of cues that lead to receiving
elicit greater motivational feedback in individuals prone to obesity, and rewards (Atkins et al., 1998; Anderson and Petrovich, 2018).

180
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

3. Physiological and neural substrates of sugar consumption To determine separation of neural networks engaged in choosing to
eat based on palatability or nutritional status, striatal dopamine levels
Feeding centers linked to limbic regions relaying emotional in- were measured in sugar consuming mice (Tellez et al., 2016). Only
formation influence our behaviour towards food (Liu et al., 2014). The when the sweet solution contained energy was the dorsal striatum,
current view of what drives us to eat takes into account the drive to basal ganglia descending pathway recruited. Energy content drives the
maintain an internal balance between energy expenditure and con- release of dopamine in the NAc, however, sweetness modulated this
sumption. However, the influence of external cues that promise im- efflux, inhibiting dopamine release as palatability decreased (Tellez
mediate reward may negate this balance (Stroebe et al., 2008; Lowe and et al., 2016). Alternatively, suppression of the sweeteners nutritional
Butryn, 2007). Sucrose added to food provides this immediate reward, value inhibited dopamine release in the dorsal striatum, demonstrating
with the human desire for sucrose or any sweet taste being comparable recruitment of different circuitry when separating energy requirements
to the degree of yearning and reward produced by drugs of addiction from pleasurable taste (Tellez et al., 2016).
(Ahmed et al., 2013). Sugar or sucrose (table sugar) is a disaccharide
composed of the monosaccharides glucose and fructose (Smith, 1960; 3.1. The effect of sugar on the neurobiology of food consumption
Southgate, 1995). Unlike most drugs of addiction (excluding alcohol),
sugar does not cross the blood brain barrier to bind to molecular sub- The hedonic value of food, the reward associated with its con-
strates/receptors on the cell surface and subsequently alter neural sumption and the learned external cues which trigger the desire to feed
plasticity. Recent research has observed astrocytes, a type of glial cell are modulated via bidirectional circuitry between the reward pathway,
that helps maintain the blood-brain barrier appear to sense and uptake hippocampus and orexigenic cells (which induce appetite and stimulate
sugar to regulate neuronal signaling related to appetite (García-Cáceres food intake) in the lateral hypothalamus (Petrovich and Gallagher,
et al., 2016). However, as sugar is a food, ingestion begins in the mouth. 2003; Petrovich et al., 2012; Kastin, 2013) (see Fig. 2) and for a com-
Sugar is initially sensed by heterodimeric G protein-coupled re- prehensive review see (Murray et al., 2016). Studies activating the
ceptors on taste cells located in the mouth and gut (Brown and Rother, lateral hypothalamus show glutamatergic inhibition of feeding
2012). Once in the small intestine, sucrose is broken down into glucose (Jennings et al., 2013) and GABAergic stimulation of feeding (Jennings
and fructose; which are metabolized by separate and disparate me- et al., 2015). Furthermore, a subset of these GABAergic neurons project
chanisms (Southgate, 1995). Glucose increases the absorption of fruc- from the VTA to the hypothalamus and express galanin (a neuropep-
tose from the gut, whereas fructose acts as a catalyst for the uptake and tide) which enhances motivation for sucrose consumption (Qualls-
storage of glucose by the liver (Laughlin, 2014). Fructose is absorbed Creekmore et al., 2017; Bocarsly, 2018).
into the bloodstream at a slower rate and persists for longer (Teff et al., Lateral hypothalamic orexin neurons gain input regarding food in-
2004). As opposed to glucose, fructose is not well absorbed by pan- take from the arcuate nucleus of the hypothalamus via endogenous
creatic beta cells and therefore stimulates very little insulin secretion melanocortin receptor antagonists; neuropeptide Y (NPY) and agouti-
(Kyriazis et al., 2012). Insulin increases satiety and subdues the reward related peptide-expressing neurons (AgRP). Further, metabolic home-
value derived from food, suggesting fructose may play a more complex ostasis is sensed by the lateral hypothalamus through surrounding
role in the development of obesity than glucose consumption (Grant glucose, ghrelin and leptin levels, and this drives food seeking beha-
et al., 1980). In addition to these observed features fructose can in- viours (Yamanaka et al., 2000).
crease the rate of carbohydrate oxidation after a meal, also decreasing Hypothalamic orexinergic and anorexinergic pathways are regu-
satiety levels (Laughlin, 2014). lated by NPY/AgRP and POMC/CART peptides and are affected in
Glucose and fructose have the ability to cross the blood-brain-bar- different ways by the consumption of sugar (Murray et al., 2016). The
rier. However, as the gut and liver rapidly break down fructose, the orexigenic pathway is influenced by two specific neuronal populations
blood concentration levels are generally low, resulting in only small within the arcuate nucleus, the first population expresses NPY and
amounts available to cross the barrier (Gonzalez and Betts, 2018). Once AgRP and stimulates food intake (Benoit et al., 2008; Yamanaka et al.,
across the barrier glucose signaling mechanisms are activated. As de- 2000). Variations in both NPY and AgRP were associated with rats
monstrated in animal studies (Asnaghi et al., 2003; Kikkawa et al., consuming more chow after being provided with a 30% sucrose solu-
1987), emerging arguments suggest fructose is produced from glucose tion (Gaysinskaya et al., 2011). Reduced expression of these peptides
in the brain via the polyol pathway (glucose → aldose reductase → was noticed following sucrose consumption, which then increased
sorbitol → sorbitol dehydrogenase → fructose), driven by hypergly- thirty to sixty minutes later, prior to feeding (Gaysinskaya et al., 2011).
cemia (Hwang et al., 2017). Glucose consumption increases functional The link between p53 (a gene encoding a protein involved in reg-
connectivity between the hypothalamus, thalamus and striatum where ulating the cell cycle) in AgRP neurons (which regulate ghrelin-induced
fructose does not affect striatal connectivity (Cha et al., 2008). Regional appetite) and obesity, has been demonstrated in mice, with its over-
cerebral blood flow patterns also differ post ingestion of these mono- expression resulting in excessive weight loss (Quiñones et al., 2018).
saccharides, with reduced flow in appetite and reward regions of the Under normal circumstances, a lack of available nutrients drives AgRP
brain after glucose consumption inclusive of the hypothalamus, tha- neurons to initiate feeding behaviour. In mice, it has been shown that
lamus, insula, anterior cingulate, and striatum. Alternate to this, fruc- the desire to feed does necessarily involve AgRP neuronal activation
tose reduces flow in the thalamus, hippocampus, posterior cingulate when the food has been enriched with sugar and fat (Denis et al., 2015).
cortex, fusiform, and visual cortex (Page et al., 2013). These differences In this case dopamine signaling initiates the desire to eat and feeding
may assist in clarifying the role of sucrose consumption in obesity and behaviour becomes driven by reward, through the neural circuits in-
diabetes mellitus. Both human and rat studies have shown that fructose volved in emotion, as opposed to the orexigenic pathway induced by
and not glucose consumption results in conditions such as metabolic metabolic need. This appears to be the case when eating for comfort
syndrome and contributes to obesity (Nakagawa et al., 2006). (Denis et al., 2015). Further examinations directly link sucrose con-
On the neuroanatomical level, regulation of energy homeostasis is sumption with the orexigenic pathway. In two separate studies sucrose
dependent on the hypothalamus (Benoit et al., 2008), however, the intake was found to be increased following NPY infusion into the lateral
NAc, part of the reward-pathway, is pivotal in the pathophysiology of ventricle or AgRP administration into the NAc shell (Badia‐Elder et al.,
sugar consumption (Geiger et al., 2008). Table 1 depicts the findings of 2003; Pandit et al., 2015). The effects of AgRP administration into the
several studies examining the effect of sugar consumption on brain NAc shell is, however, halted by pre-treatment of α-flupenthixol, a non-
regions involved in the reward pathway, defining the molecular me- selective dopamine receptor antagonist Pandit et al., 2015). Further-
chanisms resultant of the neural adaptations and the subsequent be- more, chemogenetic and optogenetic manipulation of AgRP neurons
havioural changes demonstrated by the animals. activity modulates the emotional valence of feeding. Specifically,

181
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

Table 1
The effects of sugar consumption on the reward pathway.
Sugar consumption causes neural adaptation to brain regions in Sugar consumption alters activation of Sugar consumption alters behaviour
the reward pathway molecular substrates

Ventral tegmental area ↑ Dopamine (VTA-NAc) ↑ Sensitisation (Wyvell and Berridge, 2001; Sharpe et al.,
Nucleus accumbens 2016)
Amygdala ↑ Reward seeking (Spangler et al., 2004; Gosnell, 1987)
Prefrontal cortex (Avena and Hoebel, 2003)
↓ Dopamine ↑ Anxiety (Kim et al., 2018)
↑ Kir2.1 ↑ Depression (Avena et al., 2008)
↓CREB (NAc) (Leão et al., 2012)
↑ Serotonin ↓ Motivation (Mathes et al., 2013)
↓ α6β2 nAChRs ↑ Anticipation of reward
↑ α4β2 nAChRs ↑ Motivation (Shariff et al., 2016)
↑ AMPAR (NAc core) ↑ Motivational feedback
↑ Cue-triggered food seeking (Derman and Ferrario, 2017)
IL-6↑ Leptin ↑ Anxiety
↑ IL-6 ↑Fear
↑ TNF-α ↑ Depression (Santos et al., 2018)
↓ Protein oxidation (PFC) ↓Executive function
↑ Anxiety (Chepulis et al., 2009)
↑ Delta FosB (NAc) ↑ Reward seeking (Wallace et al., 2008)
↑ Delta FosB (PFC) ↓ Fear extinction memory retention (Baker and Reichelt,
2016)

Fig. 2. Regulation of feeding behaviour and food intake by central and peripheral appetite-regulating hormones and peptides. Hypothalamic orexigenic and an-
orexigenic pathways and their regulation by NPY/AgRP and POMC/CART peptides, respectively, are depicted in the right-hand side panel. Peripheral appetite-
stimulating (green) and –inhibiting (red) hormones and peptides crossing the blood-brain barrier (BBB), and the organs they originate from are also represented. The
effect of these hormones/peptides on the stimulation or inhibition of the orexigenic (dashed green arrows) and anorexigenic (dashed red arrows) is depicted as (+) or
(-), respectively.

182
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

Table 2
The effects of sugar consumption on the orexigenic pathway.
Sugar consumption causes neural adaptation to brain regions connected Sugar consumption alters activation of Sugar consumption alters behaviour
to the orexigenic pathway molecular substrates

Hypothalamus ↑ Dopamine ↓Satiety (Hoebel et al., 2009)


Hippocampus ↓Acetylcholine
↓ Corticosterone ↓Stress (Kinzig et al., 2008)
↓CRH mRNA
↑ Orexin / hypocretin ↑Cue-induced over-consumption (Wise et al., 1995)
↓BDNF ↓Spatial learning (Molteni et al., 2002)
↑AgRP ↑Stimulation to feed (Badia‐Elder et al., 2003; Pandit
et al., 2015)
↑5-HT2CRs ↓Incentive motivation (Valencia-Torres et al., 2017)
↑Mu-1 binding ↑Overconsumption (Colantuoni et al., 2001)
↑TNF-α ↓Neurogenesis (Van der Borght et al., 2011)
↑NPY ↑Desire to feed
↑Learned cue associations (Badia‐Elder et al., 2003)

arcuate nucleus AgRP neurons were shown to regulate the emotional (Bocarsly, 2018). High fructose intake results in lower insulin levels,
aspects of feeding involved with anxiety, fear-like behaviour and ag- decreased levels of leptin and increased concentrations of ghrelin when
gression (Sweeney and Yang, 2017; Burnett et al., 2016; Joly-Amado compared to meals high in glucose (Teff et al., 2004). Alteration to
et al., 2014; Padilla et al., 2016). levels of these satiety hormones (which result in the feeling of ‘fullness’
The second, or anorexigenic pathway contains proopiomelanocortin following food consumption) may be a precursor to overeating. These
(POMC), a pre-cursor to melanocortin receptor agonist α-MS, and co- results concur with those found after intracerebroventricular injection
caine- and amphetamine-regulated transcript (CART). This pathway is of concentrated fructose or glucose into the hypothalamus of rats ex-
used to inhibit the consumption of food (Benoit et al., 2008; Lau and posed to 2-deoxy-Dglucose (2DG) (Miller et al., 2002). DG is an ana-
Herzog, 2014). Disruption of the genes encoding POMC, CART or the logue of glucose that cannot be metabolized and is known to cause
melanocortin-4 receptor may lead to obesity (Graham et al., 1997). increased food intake in rats by interfering with the process of glyco-
Mice fed a high fat diet and then provided with sucrose-sweetened lysis (Fiorentini and Müller, 1975). Rats provided with fructose injec-
water showed a down-regulation of POMC mRNA expression in the tions showed enhanced food intake both in the presence and absence of
hypothalamus (Soto et al., 2015). Prolonged limited access to sucrose 2DG, where glucose suppressed 2DG induced food intake suggesting a
lead to decreased activity of the anorexigenic oxytocin system (asso- role for brain glucoreceptors in the control of food consumption (Miller
ciated with satiety and termination of feeding), in the hypothalamus of et al., 2002). Glucoreceptors monitor blood glucose levels and are lo-
rats (Mitra et al., 2010). Together, these findings suggest sugar plays a cated within the hypothalamus. They have been shown to be stimulated
role in limiting the activation of this pathway, thus increasing the desire by low blood sugar levels to increase feeding behaviour (Ritter et al.,
to feed. 1981).
Neural pathways connecting feeding centres of the brain to the Dopaminergic neurons projecting from the VTA to the NAc are in-
limbic system have been identified through optogenetic activation of hibited by leptin and insulin and stimulated by ghrelin (Palmiter,
melanin-concentrating hormone neurons during the intake of an arti- 2007). Overconsumption of sugar has been shown to increase dopamine
ficial sweetener (sucralose) (Domingos et al., 2013). Sucralose was D1 receptor binding in the NAc core and shell, decrease dopamine D2
found to increase striatal dopamine levels which transposed the pre- binding in the dorsal striatum and increase binding to dopamine
ference to sugar normally shown by mice into a preference for the transporters in the midbrain (Colantuoni et al., 2001; Bello et al.,
sweetener (Domingos et al., 2013). Further to this, Domingos and col- 2002). Increases in dopamine delays the release of acetylcholine during
leagues showed that melanin-concentrating hormone neurons pro- feeding, which postpones satiety and paves the way for over-
jecting to reward areas are necessary for the rewarding effects of su- consumption (Hoebel et al., 2009). Alternatively, opioid receptor
crose (Domingos et al., 2013). binding (mu-1) was increased in the NAc shell, hippocampus, locus
Furthermore, the serotonergic system is indicated in the regulation coeruleus and cingulate cortex after rats were permitted intermittent
of hedonic feeding, with increases of serotonin causing decreased food binging on a 25% glucose solution (Colantuoni et al., 2001). Together
intake and decreased serotonin increasing motivation to feed (Halford these findings suggest the overconsumption of sugar may sensitize do-
et al., 2010). The suggested mechanism of action for serotonin in pamine D1 and opioid mu-1 receptors in a similar manner to drugs of
feeding behaviour is via AgRP and POMC neurons in the arcuate nu- dependence. Table 2 depicts studies examining the effect of sugar
cleus (Sohn et al., 2013). The serotonin transporter-linked polymorphic consumption on brain regions, molecular mechanisms and behavioural
region (5-HTTLPR) regulates vulnerability to stress, which is increased changes involved in the orexigenic pathway.
in cases of pathological fear, and influences energy intake, suggesting
its role on stress related overeating (Capello and Markus, 2014). Neural
adaptations resulting from sugar dependence in rodents include al- 3.2. Compulsive sucrose seeking
terations in dopamine and opioid receptor binding in the mesolimbic
cortex, changed expression of encephalin mRNA and modified NAc Since the 1980’s it has been known that neurons from the lateral
release of dopamine and acetylcholine (Avena et al., 2008). hypothalamus encode reward-associated cues (Nakamura et al., 1987)
and both feeding (Schwartzbaum, 1988) and drinking (Tabuchi et al.,
2002) behaviour. In 2015 an attempt was made to define the neural
3.1.1. Leptin and ghrelin circuits specific to compulsive sucrose seeking (Nieh et al., 2015).
There are several alternate neuronal populations, pathways and Photoinhibition was employed to show a selective pathway from the
brain nuclei which drive overeating behaviour and sucrose consump- lateral hypothalamus to the VTA reduced compulsive sucrose-seeking in
tion. Along with hypothalamic orexin neurons, MCH producing neurons mice without affecting normal feeding behaviour (Nieh et al., 2015).
and leptin-receptor cells projecting from the lateral hypothalamus to The study found a bidirectional circuit of both inhibitory and excitatory
the VTA also influence hedonic feeding and reward seeking behaviour projections from the lateral hypothalamus to dopaminergic and

183
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

GABAergic neurons in the VTA (Nieh et al., 2015). It is possible that this consumption of sucrose but not the sweetener (Aspartame) resulted in
circuit increases the focus and intensity of sucrose motivation, high- reduced reduced stress-induced cortisol (Tryon et al., 2015). Sucrose
lighting it as a potential therapeutic target for the compulsive over- and not Aspartame also produced greater activity in the left hippo-
eating of sugar. campus (Tryon et al., 2015) suggesting the HPA negative feedback loop
As obesity occurs due to excess caloric intake, the incentive value of may assist in generating a tendency towards sugar consumption in
the food consumed must be considered in parallel to the satiation of people dealing with stress. The following section reviews the complex
hunger. 5-hydroxytryptamine 2C receptors (5-HT2CRs) play a role in relationship between the HPA axis, food intake, energy stores and
incentive motivation via hypothalamic-VTA feeding circuits and have chronic stress.
therefore been considered a target for obesity treatments (Valencia- Any discerned acute stressor may cause engagement of the HPA axis
Torres et al., 2017). Lorcaserin, a 5-HT2CR agonist was administered to and result in an emotional response eliciting fear, anxiety or defensive
mice and shown to reduce both standard food intake and the desire for behaviours designed to maximize chances of survival (Ulrich-Lai et al.,
chocolate pellets with a corresponding increase in c-fos expression in 2010). The relationship between stress, cortisol levels and sugar con-
VTA 5-HT2CR GABAergic neurons (not dopaminergic neurons) sumption is far from understood, however, it appears that high sugar
(Valencia-Torres et al., 2017), validating their role in the inhibition of diets play a significant role in HPA axis regulation and binge-like eating
motivational behaviour. Similar findings arose from observations of associated with stress. When ingested, foods high in sugar release
sucrose drinking where leptin administered to the VTA was found to neuropeptides, elevating mood and reinforcing desire or selective pre-
reduce food intake and knockdown of the leptin receptors increased ference towards greater amounts of high caloric food (Ulrich-Lai et al.,
sucrose seeking (Clifton, 2017). Interestingly, mediation of the orexi- 2011). Sucrose and the sweetener saccharin have been shown to
genic pathway between the VTA and NAc may affect behavioural dampen the HPA axis response and this effect is possibly why highly
changes towards reward seeking for drugs of abuse, including with- palatable foods are consumed to assuage unpleasant emotions (Ulrich-
drawal and relapse behaviour (Kastin, 2013; Carr and Kalivas, 2006). Lai et al., 2011). Sugar consumption regulates stress-like behaviour via
Decreased dopamine levels and simultaneous increased acetylcho- the HPA axis however, the precise mechanisms of action are yet to be
line levels are associated with behavioural signs of withdrawal from elucidated.
drugs of abuse (Rada et al., 2001; Rada and Hoebel, 2005). Rats pro- Contrarily, examples of chronic stress reveal elevated levels of
vided intermittent sugar access show similar imbalances to initiate glucocorticoids with increased risk of developing stress related illnesses
withdrawal after provision of the opioid receptor antagonist naloxone such as depression. Long term stress appears to alter brain function
(Colantuoni et al., 2002). An animal will crave a substance of abuse it through changes in negative feedback loops from energy stores and
has been deprived, as shown by elevated operant responding even when glucocorticoid modulation of neural circuits (for review see (Dallman
unrewarded and increased responding to associated drug cues et al., 2003)). As glucocorticoid levels are tightly controlled by the HPA
(Bienkowski et al., 2004; Lu et al., 2004). Rats deprived of sugar after axis, one avenue of investigation has been to examine levels of mRNA
glucose overconsumption (i.e. permitted 25% glucose for 30 min per expression of 11βHSD-1(a mediator of glucocorticoid metabolism in the
day for 28 days and glucose access in their home cages for an additional liver) in cases of early life stress (Maniam et al., 2015). Interested in the
11.5 h per day) responded significantly more in operant chambers role of sugar intake on lipid homeostasis, researchers found a 53% in-
compared to controls suggesting that sensitization of the dopaminergic crease in transcriptional levels of 11βHSD-1 in animals permitted
system and associative learning leads to increased motivation to seek chronic sugar (25% sucrose solution) consumption in contrast to con-
sugar (Avena et al., 2005). The neural adaptations required to cause the trols revealing a correlation between sucrose consumption, stress and
behavioural change appear long lasting as sugar consumption ceased 2 increased glucocorticoid metabolism (Maniam et al., 2015). Ad-
weeks prior to testing. Similar results were shown in rat studies of al- ditionally, when humans are exposed to stress at a young age, they
cohol consumption (Heyser et al., 1997). become more likely to develop anxiety and depression associated with
dysregulation of the HPA axis (Heim and Nemeroff, 2001). Increased
3.3. Sucrose consumption and the hypothalamic-pituitary-adrenal axis levels of cortisol (the resultant product of the HPA axis in humans) have
also been positively correlated with higher visceral fat deposits and
Long term stress, depending on its severity, appears to correlate insulin resistance (Misra et al., 2008). Similarly high cortisol levels
with a preference for high sugar foods, suggesting its contribution to the (22% higher) were reported in overweight/obese adolescents after 2 or
progression of obesity. Chronic stress may develop through an accu- more sugar-sweetened beverages per day, suggesting significant in-
mulation of physical (traumatic), chemical (dietary), physiologic creases in stress hormone were not only due to visceral fat deposits but
(painful), psychologic (fear) or social (lifestyle) stressors (Powers and also increased sugar consumption (Shearrer et al., 2016). One suggested
Howley, 2007). Extended periods of stress result in hyper activation of mechanism may include morphological alteration of the adrenal glands
the hypothalamic-pituitary-adrenal (HPA) axis, a mammalian stress as shown through consumption of high sugar content beverages which
response system involving the endocrine and central nervous systems increased the risk of metabolic syndromes such as diabetes and lead to
(Smith and Vale, 2006). Hyper activation of the HPA axis leads to in- dysfunction of the adrenal glands (Díaz-Aguila et al., 2016). Young
creased release of corticosteroids (steroid hormones produced in the adult rats fed a 30% sucrose solution for 12 weeks showed increased
adrenal cortex) see Fig. 3 (Smith and Vale, 2006; BjÖrntorp, 2000). visceral fat deposits and insulin resistance (Díaz-Aguila et al., 2016).
Activation of the HPA axis leads to increased adrenocorticotropic Interestingly, the adrenal glands showed histomorphological changes in
hormone (ACTH) levels and increased glucocorticoids, which affect the the adrenal cortex and medulla due to sucrose consumption, suggesting
utilization of energy stores (Ulrich-Lai et al., 2010). This is of particular hyperplasia and indicators of metabolic syndrome (Díaz-Aguila et al.,
importance, as people regularly report choosing to consume sweet 2016).
tasting food (of higher caloric content) due to its ability to enhance
their mood and relieve negative emotional states (Smith and Vale, 3.4. Developmental neuroadaptation
2006). In an observation of rats undergoing acute stress, the provision
of a sucrose solution significantly reduced levels of both ACTH and In rodents prenatal stress can decrease glucocorticoid and miner-
corticosterone secretion (Ulrich-Lai et al., 2010). Furthermore, rats fed alocorticoid receptor levels in the hippocampus thus decreasing re-
with a sweetener (Saccharin), also showed lower HPA axis responses to ceptor availability for the feedback inhibition of corticosterone, which
acute stress suggesting the hedonic nature of sugar may be responsible may explain why novel stressors cause an increased and longer lasting
for the reduced stress response (Ulrich-Lai et al., 2010). corticosterone response (Henry et al., 1994). Pregnant rats underwent
Contrastingly, a study involving nineteen women found restraint stress during the third week of gestation (Henry et al., 1994).

184
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

Fig. 3. Hypothalamic-pituitary-adrenal axis.


Stress causes the release of corticotrophin-re-
leasing hormone and vasopressin from the hy-
pothalamus. These hormones are transported
to the anterior pituitary. Adrenocorticotropic
hormone (ACTH) is then released from the pi-
tuitary gland which stimulates the adrenal
glands to release cortisol, catecholamines and
aldosterone into the bloodstream. Cortisol re-
leased from the adrenal glands increases a de-
sire for palatable foods such as sugar and tar-
gets systemic organs, including the brain
where it exerts a feedback inhibition on the
release of CRH by the hypothalamus and ACTH
by the pituitary gland, via corticoid receptors
(mostly glucocorticoid receptors (GRs), and to
a lesser extent mineralocorticoid receptors).

The HPA axis and hippocampal corticosteroid receptors in the male contributes to increased craving and relapse (Robinson and Berridge,
offspring were investigated. Plasma corticosterone was found to be 2001).
significantly higher in the prenatally-stressed rats compared to controls Adolescent rats permitted sucrose consumption (5% sucrose solu-
and the receptor subtypes (hippocampal type I and type II corticos- tion) for sixteen days were later tested for motivation to seek either
teroid receptors) were decreased in the hippocampus suggesting long saccharin, maltodextrin or cocaine (Vendruscolo et al., 2010). The adult
term changes in the HPA axis may occur following prenatal stress rats showed a reduction in motivation to procure saccharin and mal-
(Henry et al., 1994). Numerous studies have examined maternal stress todextrin, however this was not the case for cocaine (Vendruscolo et al.,
and corticosteroids, for a comprehensive review see (Welberg and 2010). Further to these findings, rats presented with a choice between a
Seckl, 2001). Fewer studies involve these neuroadaptations and diet, saccharin solution and intravenous cocaine demonstrated the reward
therefore more direct investigation of high sugar consumption in mo- provided by the sweetness surpassed the desire for cocaine (Lenoir
thers may be significant in the study of childhood obesity. et al., 2007); an intense debate remains as to whether sugar is in fact
Furthermore, evidence exists for the pre-programming of HPA axis addictive (Benton, 2010; Corsica and Pelchat, 2010; Wilson, 2010).
hyper activation during prenatal periods (Welberg and Seckl, 2001). In It has been hypothesized the addictive-like behaviours present after
adolescents deemed to be at higher risk of becoming obese (by evalu- long-term sucrose consumption arise from the highly palatable nature
ating their parent’s body mass index), a higher functional magnetic of sugar and not the neurochemical effects of sugar itself (for reviews
resonance imaging (fMRI) signal was observed in brain regions asso- see (Ahmed et al., 2013; Hoebel et al., 2009; Westwater et al., 2016;
ciated with reward learning, processing and motivation after the con- Avena et al., 2009; Criscitelli and Avena, 2017; Kendig, 2014)). The
sumption of high sugar milkshakes as compared to high fat milkshakes purpose of this review is not to debate the argument, but to examine the
(Shearrer et al., 2018a). Although fat provides greater energy and role of sugar in emotional dysfunction.
therefore contributes to obesity by providing excess calories, sugar is
more often associated with modulation of habitual overeating and 4. Common neurochemistry underlie consumptive behaviours and
therefore the addiction-like behaviour associated with obesity (Shearrer emotions
et al., 2018a).
Overconsumption during adolescence leads to long-lasting changes Neuroimaging studies and animal models investigating the me-
in the dopaminergic reward system and may cause stimuli-induced chanisms involved in the progression to obesity have revealed neuro-
sensitization that is observed in adulthood (Naneix et al., 2018; biological correlates participating in the neuroadaptations of obesity
Robinson and Berridge, 2008). Stimuli-induced sensitization refers to and sugar consumption (Lindgren et al., 2018; Novelle and Diéguez,
an increased effect of the stimuli, following repeated exposures eg. 2018). Preclinical rodent models of consumption are useful for in-
repeated exposure to a loud noise may create a sensitization to noise, vestigating neural regions and pathways underlying consumptive be-
generating an enhanced response, the same being true for repeated haviours (Holgate et al., 2017; Bonin et al., 2018) with intermittent
exposures to a drug of abuse. In drug addiction, sensitization causes access to sucrose shown to have an effect on opioid, cholinergic and,
changes in dopamine transmission and delta FosB expression which importantly, dopaminergic receptors (Shariff et al., 2016; Avena et al.,

185
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

2008; Colantuoni et al., 2002; Pratt and Kelley, 2004). 4.2. Acetylcholine

Neuronal nicotinic acetylcholine receptors (nAChRs) act as biolo-


4.1. Opioids gical targets for ethanol, nicotine (Bito‐Onon et al., 2011) and sucrose
(Shariff et al., 2016; Löf et al., 2010) and modulate the neuro-
Opioid receptors are expressed throughout the limbic system and transmission of GABA, glutamate, dopamine, serotonin, acetylcholine
play key roles in the regulation of fear, happiness, anger, arousal, and noradrenaline (Wu, 2009). Homeostatic dysregulation of acet-
motivation and reward related feeding (Nummenmaa and Tuominen, ylcholine in the limbic system has been shown to modulate the moti-
2017; Levine and Billington, 2004). Opioid-induced behaviour includes vation and reward seeking behaviours characteristic of addiction and
the modulation of pain, drug addiction and control of the autonomic relapse (Hoebel et al., 2007; Aosaki et al., 2010; Rahman et al., 2015),
nervous system, which includes emotions, cognitive processes and neurodegeneration (Alzheimer’s disease, mild cognitive impairment)
stress coping (Benarroch, 2012; Belzeaux et al., 2018). Opioids influ- and mental illness (anxiety, depression and schizophrenia) (Wu, 2009;
ence the way we process rewards through interaction with the dopa- Posadas et al., 2013). Within the NAc, cholinergic interneurons regulate
mine system (Bray and Bouchard, 2014). The way an animal behaves the expression and release of enkephalin (Kelley et al., 2005) and many
towards food is also modulated by endogenous opioid neuro- rat studies support the theory that they inhibit feeding behaviou (Avena
transmitters, which are neural substrates that create susceptibility to et al., 2006; Hoebel et al., 2007; Mark et al., 2006). As increased ser-
reward (Wise, 1996; Kavaliers and Hirst, 1987). Alterations in hedonic otonin reduces the motivation to feed, a study using rats to perform a
processing are demonstrated to be highly dependent on endogenous progressive ratio task and paroxetine (a selective serotonin reuptake
opioid receptors in the NAc suggesting these changes may result in the inhibitor) found that increased serotonergic activity decreased the ap-
development of reward-related disorders such as obesity (Naneix et al., petitive-based responses to the hedonic taste of sucrose and the aversive
2016). taste of quinine (Mathes et al., 2013). Withdrawal from morphine in-
Opioid peptides (β-endorphin, enkephalins, and dynorphins), and creases acetylcholine levels in the NAc, while dopamine simultaneously
activation of their receptor types, μ (mu), δ (delta) and κ (kappa) in the remains low, leading to the assertion that this mechanism is involved in
NAc cause inhibition at both pre and post synapses (Benarroch, 2012). the unpleasant aspects associated with withdrawal (Avena et al., 2008;
Opioid agonists modulate pleasure, reward and reinforcement through Rada et al., 2001). Arecholine (a muscarinic agonist) inhibits feeding
activation of mu and delta receptor ligands via the mesolimbic dopa- but can be blocked by pirenzapine (a muscarinic acetylcholine 1 re-
mine system whereas the dysphoria associated with withdrawal relies ceptor antagonist), suggesting a role for acetylcholine in food intake
on kappa receptors (Nummenmaa and Tuominen, 2017; Benarroch, (Avena et al., 2006).
2012; Herz, 1997). Mu receptor binding in the NAc is significantly in- In 2010, Lof and associates utilized rats to demonstrate the role of
creased after cocaine (Bailey et al., 2005), morphine (Vigano et al., nAChRs in the conditioned reinforcement of lever pressing for a sucrose
2003) and sucrose (Colantuoni et al., 2001). reward (Löf et al., 2010). Using a selective antagonist at α7 nAChRs
In the endeavor to define specific peptide and receptor involvement (methyllycaconitine) they showed reduced lever pressing for the su-
in the motivation and reward seeking behaviour characteristic of he- crose solution down to the level of the control, suggesting mediation of
donistic feeding, several animal studies have combined opioid antago- the amount of influence the cue had on the desirability of the sucrose
nists and agonists with sucrose consumption (Hayward et al., 2006; (Löf et al., 2010). More recently our laboratory elucidated the effects of
Beczkowska et al., 1992; Ruegg et al., 1997). Opioid receptor antago- varenicline (an FDA-approved drug to reduce nicotine cravings) on
nists (naltrexone and naloxone), used to treat drug and alcohol de- sucrose consumption in rats (Shariff et al., 2016). Varenicline, a partial
pendence, were shown to use kappa and mu2 binding sites to inhibit agonist at α4β2* and antagonist α6β2* nAChRs subtypes, significantly
sucrose intake, thus demonstrating the role of endogenous opioids in reduced sucrose consumption in both short and long term binge like
regulation of hedonic rewards in rats (Beczkowska et al., 1992). (intermittent access) drinking. While α4β2* nAChR binding sites were
Substance P and the NK1 receptor system, interact with opioid re- increased, α6β2* nAChRs were significantly decreased in the NAc as a
ceptor systems to regulate reward related behaviours, suggesting its result of both short-term and long-term sucrose consumption (Shariff
role in addictive behaviour (Steensland et al., 2010). Substance P is also et al., 2016). By modulating dopamine release in the NAc (Feduccia
a neuropeptide that affects the orexigenic pathway, via the NK1 re- et al., 2014) the control of sucrose consumption by nicotinic receptors
ceptor (Karagiannides et al., 2008). Its presence in the brain assists the and their subtypes may provide promising therapeutic strategies for
regulation of feeding, however, it is also located in the stomach and obesity (Cocores and Gold, 2008).
small intestine suggesting it may be a potential therapeutic target for
obesity. Administration of a substance P antagonist (CJ 012,255) was 4.3. Dopamine
shown to prevent weight gain in obese mice following a 2 w high fat
diet while treatment with CJ 012,255 in obese mice resulted in a loss of Sucrose is considered a primary or natural reinforcer that does not
weight loss and reduction improved insulin sensitivity, partially due to require a learning process to be considered desirable (Corsini, 1999).
reduced food intake (Karagiannides et al., 2008). Primary reinforcers are known to trigger dopaminergic activation in the
Using a two-bottle choice drinking paradigm, knockout mouse VTA (Wise, 1996; Olds and Olds, 1963; Moore et al., 2018), which act
models (lacking either one or two opioid peptides) were used to identify on the NAc via the medial forebrain bundles (Yu and Kim, 2012). Ac-
opioid receptor ligands modulating sucrose preference (Hayward et al., tivation of dopaminergic neurons in the VTA modulates processes of
2006). Enkephalin and dynorphin were found to modulate preference memory, learning and motivation (Yang et al., 2018; Adcock et al.,
to sucrose but were not considered necessary for its consumption, 2006; Otmakhova et al., 2013; Wise, 2004) as well as the intense
supporting the role these peptides play in the motivation to consume emotions associated with love, sexual desire, orgasm (Ulrich et al.,
unnecessary calories208. A decrease in enkephalin mRNA expression 2018; Holstege et al., 2003; Acevedo et al., 2012), fear, stress, anxiety
was observed in the NAc after rats were provided with intermittent (Bouchet et al., 2018; Zhang et al., 2018b; Moghaddam, 2018) and
access to sugar (Spangler et al., 2004). The link between opioids, do- psychosis (De Nijs et al., 2018; Knolle et al., 2018). Rat models of
pamine and motivation for hedonic rewards is further reinforced with obesity display low levels of dopamine and impairment in the release of
data showing rats preference for sweet taste after the introduction of dopamine (Geiger et al., 2008).
morphine to the NAc (Berridge, 1996; Pecina and Berridge, 1995). The mesocorticolimbic dopaminergic system has been implicated in
modulation of addictive reward seeking behaviour, (for review see (Liu,
2018)). Drugs of abuse such as alcohol, nicotine, cocaine and

186
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

amphetamines that cause increased extracellular dopamine and sub- shown opioid agonists such as morphine to increase the impulsivity to
sequent heightened pleasure levels in humans (Volkow et al., 2002; feed when injected systemically or directly into the NAc, para-
Robinson and Berridge, 1993) also cause increases in dopamine release ventricular nucleus, AMG, hypothalamus and tegmentum (Gosnell,
in the NAc in rats (Wise et al., 1995; Di Chiara and Imperato, 1988). 1987; Stanley et al., 1988; Woods and Leibowitz, 1985).
Commonly abused (opiates, ethanol, cocaine, amphetamine and nico- Further investigations have demonstrated how sugar access can lead
tine) given to rats resulted in increased extracellular dopamine in the to cross-sensitization of dopamine-altering drugs. Intermittent access to
NAc and the dorsal caudate nucleus Di Chiara and Imperato, 1988). sugar has been found to cross-sensitize with amphetamines up to 8 days
Similar results were found in rats permitted intermittent access to sugar post sugar ingestion (Avena and Hoebel, 2003) Female rats provided
with subsequent binge drinking resulting in increased dopamine release with either a 10% sucrose solution or a rotation of sucrose solution and
in the NAc (Avena et al., 2006; Rada et al., 2005). withdrawal, displayed behavioural cross-sensitization to a small dose of
Supporting both the hedonic potency of sucrose and its reliance on amphetamine (Avena and Hoebel, 2003; Avena et al., 2008). Similar
dopaminergic regulation, orosensory stimulation in sucrose sham-fed results for sucrose and cocaine (Gosnell, 2005) and sucrose and quin-
rats resulted in increased dopamine release within the NAc, thus in- pirole (a dopamine agonist) have also been found (Foley et al., 2006).
creasing reward seeking behaviour (Hajnal et al., 2004; Schneider, The rats maintained on the cyclic sucrose solution responded hyper-
1989). Compelling evidence shows drugs not routinely abused, such as actively to amphetamine in comparison to controls, suggesting the
imipramine, atropine and diphenhydramine do not display rewarding binge-like sugar consumption leads to increased amphetamine sensi-
properties and do not alter synaptic dopamine levels (Di Chiara and tivity, which may be due to neuroadaptations of the dopamine system
Imperato, 1988). Studies show an attenuated dopamine system in (Avena and Hoebel, 2003). Compelling evidence suggests chronic stress
obesity prone rats in the NAc, PFC and dorsal striatum and suggest the leads to similar cross-sensitization (Leão et al., 2012; Koob, 2009b). As
reduction in hedonic response available to these animals is related to globally the consumption of sugar is on the rise an interesting avenue of
hyperphagia (increased appetite) and resultant obesity (Geiger et al., investigation may be to examine the effect of sucrose consumption on
2008). These findings were similar to those of a human study showing stress derived cross sensitization.
reduced levels of dopamine D2 receptors correlated with increased
body mass index in 10 obese individuals (Wang et al., 2001). 4.3.2. Dopamine and impulsivity
Dopamine is released in response to food cues, making it essential
4.3.1. Dopamine sensitization for the motivational prompt to consume food (Flagel et al., 2011;
In 1993 Robinson and Berridge postulated the incentive-sensitiza- Salamone and Correa, 2012). As dopamine neurons project from the
tion theory, which suggested that repeated exposure to rewarding VTA to the NAc, caudate, putamen, PFC, hippocampus and AMG, they
substances sensitizes the dopaminergic system resulting in exaggerated modulate a wide variety of behaviours and emotions (de Jong, 2015)
“cue-triggered wanting” which can transform into a compulsion to seek including impulsivity (Wade et al., 2000). In relation to eating disorders
out an associated reward (Wyvell and Berridge, 2001; Robinson and such as obesity, there appears to be an impulsive tendency to over-
Berridge, 1993). Cue-reward learning depicts the learned association consume sweet foods in response to cravings for carbohydrates and
between the rewarding effects of substances of abuse and environ- sugars. When a subject is withdrawn from an addictive substance, im-
mental cues (Bianchi et al., 2018; Tye et al., 2008). This learning pulsivity increases and can be quantified using a behavioural test called
contributes to obsessive overuse and the tendency to relapse after long differential-reinforcement-of-low-rate-schedule performance (DRL)
periods of abstinence (Bianchi et al., 2018; Ziauddeen et al., 2012). (Mangabeira et al., 2015). The test requires the subject to withhold a
Repeated or prolonged substance abuse can cause modifications to response for a set period of time before they are permitted to respond
neurotransmitter release and alterations in synaptic strengths and thereby earn a reward (Kirshenbaum et al., 2008). Deprivation
(Gerdeman et al., 2003). For example, rats provided with intermittent after long term sugar consumption in rats resulted in impairment of
access to sucrose display signs of dopamine sensitization through al- DRL performance, confirming its resemblance with drug addiction and
tered dopamine receptor function (Sharpe et al., 2016). suggesting an increase in impulsive behaviour following sugar depri-
Behavioural sensitization or hyper-locomotion is a motor response vation (Mangabeira et al., 2015). The suggested mechanism of action
that increases incrementally with repeated exposures to a drug. It is for this behaviour is the dopaminergic system as dopamine receptor
reflective of a hyposensitized (or attenuated) reward pathway and is antagonists (pimozide (Wise, 2006), raclopride (Maldonado et al.,
said to contribute to the craving or compulsive seeking which char- 2006), SCH 23390 (Verty et al., 2004)) can attenuate the desirability of
acterizes addictive behaviour (Nona and Nobrega, 2018; Robinson and highly palatable foods in rats (Mangabeira et al., 2015; Wise, 2006).
Berridge, 2001). In both human and rodent studies, behavioural sen- The progression towards obesity is linked with a decline in neural
sitization has been observed as dose-dependent (Jing et al., 2018; responses to reward similarly induced by cocaine and heroin, which
Huber et al., 2018) and shows considerable individual variation moderate the transition from casual user to the impulsive actions of a
(Robinson, 1988). Brain-derived neurotrophic factor (BDNF) is believed compulsive drug-taker (Johnson and Kenny, 2010). Impulsive actions
to modulate this behaviour through overexpression of the dopamine D3 such as over eating are modulated by the excitation of D1 dopaminergic
receptor (Bordet et al., 1997) and has been implicated in drug addic- neurons and inhibition of D2 dopaminergic neurons in the dorsal
tion, schizophrenia (Guillin et al., 2001; Staley and Mash, 1996; striatum and the NAc (Johnson and Kenny, 2010). Preclinical and
Gurevich et al., 1997) and novelty seeking behaviour in obese rats clinical studies have been conducted to investigate the role of dopamine
(Savage et al., 2014). in obesity with findings showing obese subjects have an inverse re-
Animals sensitized to one drug are often cross-sensitized to other lationship between the abundance of dopamine D2Rs and body mass
substances of abuse as well as non-drug substances (Avena et al., 2008). index, suggesting a dopaminergic role in compulsive eating (Wang
This has been demonstrated with rats exposed to amphetamines be- et al., 2001; Johnson and Kenny, 2010; Volkow and Wise, 2005; Pijl,
coming sensitized to cocaine (Pierce and Kalivas, 1995), cocaine cross- 2003). Male rats self-administering amphetamine showed a reduction
sensitizing to alcohol (Itzhak and Martin, 1999) and cocaine cross- in the ability of D2Rs to inhibit dopamine release in the NAc supporting
sensitizing with stress (Covington and Miczek, 2001). Similar to these, the theory that desensitization due to short term substance abuse
short term sucrose consumption increases binding affinity to opiates modulates addiction-related behaviours such as impulsivity (Calipari
(Avena et al., 2008), resulting in cross tolerance to other opioids such as et al., 2014; Bello et al., 2011; Cools et al., 2003). In a human study,
morphine, while chronic sucrose consumption reduces the analgesic twelve patients with Parkinson’s disease, a disorder of the mesolimbic
properties of morphine (Steensland et al., 2010; D’Anci et al., 1996; dopamine system, underwent several cognitive tests designed to pro-
Fidler et al., 1993; Kanarek et al., 1991). A number of studies have also vide a measure of impulsivity. Increased impulsive behaviour was

187
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

found to correlate with greater dopamine levels Cools et al., 2003). Two specific brain circuits are notable in anxiety disorders, the first
The neural mechanisms modulating motivation and drive are im- involves the dorsolateral PFC, anterior cingulate cortex, dorsal parietal
plicated in both the loss of self-control exhibited by over eating and cortex and precentral gyrus, all associated with cognitive control, ex-
addiction to drugs of abuse (Avena et al., 2008; Berridge, 1996). Neu- ecutive function, flexible cognition, working memory and attention
rofunctional imaging has documented neuroadaptations in the PFC (Coplan et al., 2018). The second circuit is known as the negative affect
(Volkow and Fowler, 2000), hippocampus (Wang et al., 2006), AMG circuit and as the name suggests is activated by negative stimuli
and NAc (Volkow and Wise, 2005). A functional MRI study was con- (Coplan et al., 2018). Anatomically the hippocampus, AMG, medial
ducted showing female adolescents an image of either highly palatable PFC, and the dorsal, subgenual and pregenual regions of the anterior
food (dessert) or a vegetable (Batterink et al., 2010). In comparison to cingulate cortex may undergo neural adaptation to skew negative bias
leaner adolescents, overweight adolescents had increased behavioural and threat by reducing attenuation through the cognitive control circuit
impulsivity and decreased neural activation in frontal inhibitory re- (Coplan et al., 2018).
gions of the brain (e.g. medial prefrontal cortex, and orbitofrontal Human studies show links between high caloric food consumption
cortex) (Batterink et al., 2010). In addition, activation of the reward and anxiety. Recent evidence from epidemiological studies found a
region was positively correlated with body mass index when shown the suggestive link between greater consumption of processed foods and
picture of dessert (Batterink et al., 2010). These findings suggest the widespread presence of anxiety disorders (Jacka et al., 2011; Quirk
reduced functioning of inhibitory controls combined with increased et al., 2013; Westover and Marangell, 2002). There are also numerous
responses in food reward regions are relevant to weight gain (Batterink studies linking adverse childhood experiences (early life stress) with an
et al., 2010). The Stroop test requires an individual to suppress their increased risk of body weight gain and obesity during adolescence and
automatic response to certain stimuli. This was used to assess the in- adulthood (Noll et al., 2007; Ramasubramanian et al., 2013; D’Argenio
hibitory control of overweight / obese 10 y old children in comparison et al., 2009; Gunstad et al., 2006; Lumeng et al., 2013). More specific
to normal weight children (Reyes et al., 2015). Results indicated behavioural studies observing acute and chronic withdrawal from sugar
changes to inhibitory control functions in overweight children sug- in rodents show anxiety is induced when withdrawal follows extensive
gesting less emotional self-regulation, which in conjunction with dys- periods of sugar consumption (Avena et al., 2008). Long exposures (1
functional impulsivity control may contribute to over eating behaviour month of 12 h daily access) to palatable foods did result in rats showing
in the overweight/ obese children (Reyes et al., 2015). increased anxiety-like behaviours on the elevated plus maze (EPM)
when tested 24 h after withdrawal (Avena et al., 2008). Rats exposed to
5. Common anatomical structures and neural substrates of stress 12 h intermittent access to 10% sucrose solution for 28 days also dis-
driven, emotional behaviour played anxious behaviour in the EPM after 36 h withdrawal (Avena
et al., 2008). These rats showed reduced conditioned suppression after
The insular cortex and cingulate cortex (part of the limbic system) 1 and 28 days of abstinence, measured by a failure to significantly re-
are responsible for the processing of emotions in conjunction with duce the number of lever-presses for sucrose during the presence of a
processes involving higher cognition (Rajmohan and Mohandas, 2007). tone stimulus paired with shock (Avena et al., 2008). Similar results
The hippocampus provides negative feedback for the HPA axis with have been reported using longer cycling periods whereby rats given
both neuroadaptations to its volume and capacity for neurogenesis intermittent access to a sucrose diet showed increased anxiety in the
implicated in emotional disorders (Jacobson and Sapolsky, 1991). The EPM and defensive withdrawal tests after 8 and 9 weeks on the diet
AMG processes, stores and retrieves fear memories, and initiates ap- cycle followed by 48 h withdrawal (Parylak et al., 2012). Alternate
propriate behavioural responses (Martin et al., 2009). It is responsible investigations have shown rats fed a carbohydrate rich diet to have
for the manner in which we express fear, aggression and defensive increased protein oxidation in the frontal cortex which correlated with
behaviour (Martin et al., 2009). The PFC regulates executive functions anxiogenic behaviour (Souza et al., 2007). These studies suggest a high
and reward processing while impulse control and mood are modulated probability of long term sugar consumption may contribute to symp-
by the orbitofrontal cortex (Martin et al., 2009). Emotional disorders toms of anxiety.
involve a complex interconnected number of neuroendocrine, neuro- A long term study has shown an opposite effect in terms of anxiety
peptide, neurotransmitter and neuroanatomical adaptations (Gulpinar and depressive like behaviours (Cao et al., 2007). Findings show that
and Yegen, 2004). These alterations may occur due to genetic predis- sugar consumption had no significant effects on motor activity in an
position or as a result of environmental influence (Martin et al., 2009). open field (test for locomotor and anxious like behaviour), on ex-
We assert that abundant evidence exists regarding the role over- ploration in a T-maze, or on anxiety in an EMP (Cao et al., 2007).
consumption of sugar plays in altering these brain regions and con- Another study looking at the anxiety effects of sugar after a 1 year in-
tributing to emotional disorders such as depression, anxiety and fear. tervention of sucrose and honey found that anxiety decreased after 3
months of consumption, as tested with the EPM and open field test
5.1. Anxiety (Chepulis et al., 2009). Importantly, this reduction was maintained
until the end of the experiment at 12 months (Chepulis et al., 2009).
Generalized anxiety disorder is defined by uncontrolled, ex- Despite this, honey-fed rats showed significantly less anxiety
aggerated concern over numerous endeavours Association, 2019. throughout the study as compared with those fed sucrose (Chepulis
Treatment often consists of anxiolytic drugs that act primarily on the et al., 2009). Together this data suggests that chronic (i.e., greater than
monoamines serotonin, noradrenalin and dopamine (Martin et al., 1 month) intermittent access to high energy foods may increase anxiety-
2009). In the central nervous system these neurotransmitters are re- like behaviour followed by a plateau observed after 3months (Chepulis
leased in conjunction with neuropeptides that have strong links to an- et al., 2009).
xiety, such as neuropeptide Y and cholecystokinin expressed in the
limbic cortex where they influence emotions and stress levels (Martin 5.2. Depression
et al., 2009). Other molecular substrates involved in regulating the
stress response include corticotropin-releasing factor and adrenocorti- Depression is characterized by feelings of sadness, hopelessness and
cotropic hormone in the HPA axis (Patriquin and Mathew, 2017). In reduced pleasure in daily activities Association, 2019. Functional,
cases of anxiety, those carrying the short-allele of the serotonin trans- structural and neurochemical factors associated with the pathophy-
porter gene (5-HTTLPR) demonstrate over activation of the AMGs in siology of depression include dysregulation of the HPA axis resulting in
response to viewing fearful and angry faces as compared to those car- reductions in hippocampal, PFC and striatal volumes (Koolschijn et al.,
rying the long-allele (Hariri et al., 2005). 2009). In areas of emotional processing such as the AMG and PFC

188
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

abnormal metabolism of glucose and changes to cerebral blood flow tracked dietary regimes and the corresponding medical health of 8000
have also been demonstrated (Drevets, 1998). Although the association participants over a 22 year period (Knüppel et al., 2017). Observing a
between obesity and depression is well documented, less is known five year duration they found a 23% increase in likelihood of men being
about the physiological and corresponding psychological influence diagnosed with depression if they consumed ≥ 67 g of sugar per day
long-term sugar consumption has on depression (Luppino et al., 2010). compared to ≤ 40 g (Knüppel et al., 2017). Cumulatively, these studies,
Recent research suggests a possible contributor to the incidence rate of while not able to confirm that sugar causes depression, appear to pro-
depression is sugar overconsumption (Westover and Marangell, 2002; duce enough evidence to show sugar overconsumption contributes to
Sánchez-Villegas et al., 2012). We suggest the neuroadaptations that an increased risk of developing depression.
occur to depression-related brain regions, namely the hippocampus,
PFC and AMG, following sugar intake (reviewed above) contributes to 5.3. Neurogenesis
the incidence of depression (Robinson et al., 2014; Moorman and
Aston-Jones, 2014; Hsu et al., 2015). Neurogenesis is the term given to new neurons generated from
The first study reporting a possible link between sugar and de- neural stem cells (Ming and Song, 2011). The molecular substrates of
pression was conducted in 2002 by Westover and Marangell (Westover stress, although not well understood are implicated in the regulation of
and Marangell (2002)). Data collected data from 6 countries showed a adult neurogenesis, through molecular pathways modulated by gluco-
correlation between the consumption of sugar (calories/capita/day) corticoids, inflammatory mediators and neurotrophic factors (Egeland
with the yearly rate of major depression disorder (Westover and et al., 2015). Studies on antipsychotic pharmacology support these
Marangell (2002)). Fast food further increases the potential of devel- findings by linking the blockade of dopamine D2 receptors with re-
oping depression, and commercial baked goods are also positively ductions in parkinsonian symptoms, anhedonia and increased neuro-
correlated with depressive disorders (Sánchez-Villegas et al., 2012). Not genesis (Aringhieri et al., 2018; Chikama et al., 2017). The effect of diet
only highly palatable food, but sweetened beverages (either artificially on hippocampal neurogenesis has been relatively unexplored.
or otherwise) also contribute to depression (Westover and Marangell Rats given 1 month access to fructose showed almost a 40% re-
(2002); Sánchez-Villegas et al., 2012). Regular consumption of swee- duction in the number of BrdU / NeuN-immunopositive cells (mature
tened beverages indeed increases the occurrence of depression and neurons) in the dentate gyrus of the hippocampus (Van der Borght
suicidal tendencies (Shi et al., 2010; Pan et al., 2011; Lien et al., 2006; et al., 2011). Alternatively, rats consuming glucose had a similar
Guo et al., 2014). number of BrdU / NeuN-immunopositive cells as the water controls
In rats given the same high sugar/chow diet cycle for 7 weeks, (Van der Borght et al., 2011). Density of immature neurons labelled
highly palatable foods induce depressive-like behaviour as evidenced with PSA-NCAM was decreased following sucrose, fructose and glucose
greater immobility in the Forced Swim Test (test for depression) and (Van der Borght et al., 2011). Sugar consumption increased the number
decreased preference for 0.8% sucrose (test for anhedonia) (Iemolo of proliferating cells positive for Ki67 (a marker of cell proliferation
et al., 2012). Correlations with symptoms of depression have been (Scholzen and Gerdes, 2000)) in all cases and rats offered sucrose or
observed following long-term exposure to sucrose, with long term fructose showed more cell death in the dentate gyrus of the hippo-
overstimulation of the dopaminergic system during adolescence, which campus (Van der Borght et al., 2011).
may occur through sucrose binging (Rada et al., 2005) resulting in Rats given a high fructose corn syrup solution throughout adoles-
deficits in later life that affect motivation, memory and happiness cence were tested for hippocampal-dependent contextual memory.
(Avena et al., 2008; Naneix et al., 2018; Kendig, 2014). When a model Impairments in memory function were found in later in life, suggesting
designed to generate a depressed-like state (known as social defeat-in- that sugar consumption early in life may have long-term negative ef-
duced persistent stress) was used, rats showed increased motivation to fects on memory function (Noble et al., 2017). High fructose con-
acquire a sucrose reward and reinstated sucrose-seeking induced by a sumption is also linked to insulin resistance (Stranahan et al., 2008)
cue (Riga et al., 2015). Importantly, these studies showed the long-term with reduced hippocampal neurogenesis (Van der Borght et al., 2011).
effects of stress exposure induced deficits in the ability to evaluate Hence it appears convincing that a high-sugar diet negatively affects
natural rewards (Riga et al., 2015). adult neurogenesis which may contribute to the anxiety and depressive
Congenitally helpless rats, a genetic model of predisposition to like behaviour demonstrated by animals after withdrawal. Decreased
major depression, were used to respond less to the reward of sucrose neurogenesis in the hippocampus has further been implicated in
solution (Vollmayr et al., 2004). This lack of motivation to partake in memory dysfunction and cognitive impairment disorders such as Alz-
sweet rewards is conversely shown after long term sucrose consumption heimer’s disease (Price et al., 2001) and the learning and memory
by adolescent rats, which results in depressive like behaviour in processes involved in appetitive control (Davidson et al., 2009; Lathe,
adulthood including symptoms of anhedonia (inability to feel pleasure) 2001). The hippocampus may hold further insight into the cognitive
and increased anxiety-like behaviour (Gueye et al., 2018). Adult rats processes by which food becomes entwined with motivation and new
consuming sucrose showed a similar depressive-like behaviour, but to a rewards.
lesser degree, suggesting the critical period of brain development that
occurs during adolescence can be moderated by sucrose consumption 5.4. Fear
and may increase the instance of disorders related to rewards, such as
depression (Gueye et al., 2018). The functional neuroanatomy of fear encompasses the AMG, which
As depression correlates with altered glucose metabolism, it is not stimulates the HPA axis and the hippocampus, which suppresses ac-
surprising that comorbidity of diabetes mellitus (DM) and depression tivity of the axis (Martin et al., 2009). Hyper activation of the AMG was
occurs quite frequently, greatly increasing the mortality risk (van recorded in response to viewing faces construed as fearful (Bryant et al.,
Dooren et al., 2013). A study highlighting the link between glucose and 2008a) and has been implicated in post-traumatic stress disorder
depression in patients whose histories included DM and cardiac disease (PTSD) (Cortese and Phan, 2005). The PFC is required for the extinction
found there was a 34% increased chance of depression in subjects with of fear memories (Giustino and Maren, 2015), and in line with these
elevated blood glucose levels (Azimova et al., 2015). A study including findings changes in volume of the anterior cingulate cortex have been
70,000 postmenopausal women found a higher risk of developing de- implicated in a reduced ability to extinguish fearful memories (Bryant
pression in women consuming a high-sugar diet than those consuming et al., 2008b). When fear memories become dysfunctional it is thought
high naturally occurring sugars (Gangwisch et al., 2015). However, that the intrusive, recurring thoughts result from an inability of the
research suggests sugar may contribute to depression more in the male cognitive control circuit to repress the negative effect circuit (Morey
population as was demonstrated by the Whitehall Study II which et al., 2008). In patients with PTSD their information processing may be

189
Table 3
Published reports on the effect of sucrose and sweetener consumption on cognition, emotion and stress.
Sucrose or sweetener consumption in rats Authors and year Subjects Tasks Brain region involved Findings
A. Jacques, et al.

2h/day 10% sucrose for 28 days Xu, T.J. and Reichelt, A.C., 2017 3 week-old male Sprague- EPM, open-field, NPR and short- and hippocampus, basolateral increased anxiety-like behaviours
(Xu and Reichelt, 2018) Dawley rats (n = 8) long-term NOR (Organization, W.H., amygdala
2017)
Rat chow supplemented with meat pies, cakes and Reichelt, A., et al., 2015 6 week-old male trace fear conditioning hippocampus strengthened visual fears and attenuated
biscuits for 10 weeks (Reichelt et al., 2015) Sprague–Dawley rats (n = 16) contextual fears
1
Powdered diet: 7.9% sucrose for 52 weeks Chepulis, L.M., et al., 2009 2-month old Sprague Dawley EPM, NOR Y maze, C NA reduced
(Chepulis et al., 2009) rats (n = 10-14) spatial memory and increased anxiety
35% sucrose solution for 9 weeks Lemos, C., et al., 2016 (Lemos 12-weeks old, male Wistar rats open field, object displacement, NOR, Hippocampus decreased memory performance and
et al., 2016) forced swimming test increased helpless behaviour
1
32% sucrose solution for eight weeks Jurdak, N. and Kanarek, R.B., 6 week old male Long-Evans NOR NA decreased cognitive performance on object
2008 (Jurdak and Kanarek, rats (n = 10) recognition
2009)
10% fructose in drinking water for 7 months Sangüesa, G., et al., 2017 female Sprague-Dawley rats Morris water maze, (Organization, frontal cortex and reduced performance in the NOR test
(Sangüesa et al., 2018) W.H., 2017) NOR hippocampus
30% sucrose solution, 30 m, twice daily, 2-4wk Ulrich-Lai, Y.M., et al., 2010 Adult male Long-Evans rats Restraint stress, social interaction hypothalamic - reduced ACTH by sucrose and saccharin
Or 0.1% saccharin (Ulrich-Lai et al., 2010) test, open field, EPM paraventricular nucleus, - reduced corticosterone only after sucrose
BLA sucrose
- reduced restraint-induced tachycardia and
behavioural anxiety
10-34% sucrose solution Wilmouth, C.E., and Spear, L.P., Adolescent male Taste reactivity and voluntary NA greater positive taste reactivity and reduced
2009. (Wilmouth and Spear, Sprague–Dawley rats consumption negative responding
2009)
Free access to 5% sucrose or water for 16d Vendruscolo, L.F., et al., Male Wistar rats fixed- and progressive-ratio self NA Sugar overconsumption during adolescence,
(Vendruscolo et al., 2010) administration of: reduced motivation for saccharin and
- saccharin maltodextrin

190
- maltodex-
trin
- cocaine
1% sugar solution (high fructose corn syrup) Noble, E.E., et al., 2019 (Noble Male rats novel object in context Hippocampus - impairments in hippocampal-dependent
throughout the adolescent phase of et al., 2017) memory function later in life
development (post-natal day 26-56). - NOIC performance impaired at PN 175

Sucrose and sweetener consumption in mice Authors and year Subjects Tasks Brain region involved Findings
High-carbohydrate diet (45% condensed milk, Santos, C.J., et al., 2018 (Santos 5 – 7 week old, male BALB/c restraint stress, 1EPM, contextual fear NA increased anxiety-like and depressive-like
10% sugar and 45% chow) for 8 weeks et al., 2018) mice conditioning, tail suspension test behaviour and aversive memory
10% sucrose, 3 I2BC for four weeks, withdrawal Kim, S., et al., 2017 (Kim et al., C57BL/6 mice tail suspension test, (Organization, nucleus accumbens withdrawal after sucrose overeating induces
animals received only water for one week after 2018) W.H., 2017) EPM, sucrose preference depression and anxiety-like behaviour.
the four weeks test
Acesulfame potassium and low carbohydrate diet Ibi, D., 2018 (Ibi et al., 2018) Male ddY strain mice (7–9 Y-maze and NOR, glucose levels frontal cortex decrease in short-term and object cognitive
for 4 weeks weeks old) memories
decreased glucose levels

Sucrose and sweetener consumption in humans Authors and year Subjects Tasks Brain region involved Findings

2
Sugar, sweetened beverages, and fruit intake was Cohen, J.F.W., et al., 2018 mother and child pairings, PPVT-III KBIT-II NA adverse impact on child memory and
assessed on a servings per day basis (Cohen et al., 2018) during pregnancy and KBIT-II learning
childhood mean ages 3.3 yrs, WRAVMA
7.7 yrs WRAML and HOME-SF
(n = 1,234)
High-sugar content milkshakes Shearrer, G.E., et al., 2018 133 adolescents food picture exposure during fMRI temporal gyrus, operculu, increased signal in the reward learning,
(Shearrer et al., 2018b) juxtapositional lobule, processing and motivation regions of the
thalamus, caudate brain
(continued on next page)
Neuroscience and Biobehavioral Reviews 103 (2019) 178–199
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

1 EPM: elevated plus maze; NPR: novel place recognition; NOR: novel object recognition. 2 PPVT-III: Peabody Picture Vocabulary Test, third edition for maternal testing; WRAVMA: Wide Range Assessment of Visual
Motor Abilities, KBIT-II: Kaufman Brief Intelligence Test, second edition, WRAML: Wide Range Assessment of Memory and Learning for childhood testing; HOME-SF: Environment short form test to evaluate home
overpowered by the hyper activation of the AMG when exposed to
threat related stimuli (Falconer et al., 2008).

nausea, and a trend toward lower cortisol


- higher activity in the left hippocampus
At the molecular level corticotrophin-releasing factor from the

- lower reactivity to naltrexone, lower


paraventricular nucleus of the hypothalamus initially activates the HPA
sucrose consumption resulted in:

- reduced stress-induced cortisol


axis in response to threatening stimuli (Martin et al., 2009). Low levels
of corticosterone, which acts through binding to mineralocorticoid and
glucocorticoid receptors, as well as enhanced negative feedback of the
HPA axis are associated with pathological fear (Han et al., 2014). Rats
that underwent a single prolonged stress paradigm (a reliable animal
model of PTSD) have shown a down-regulation of both these receptors
in the hippocampus, enhanced fear and altered neuronal morphology of
Findings

AMG neurons (Han et al., 2014). Glucocorticoid receptors and dopa-


mine receptors have been implicated in processes within the PFC that
drive extinction memory learning. Infusions of corticosterone or a
glucocorticoid receptor antagonist (RU38486) into the infralimbic
cortex and pretreatment with sulpiride (a dopamine D2 receptor an-
Brain region involved

tagonist) attenuated fear expression suggesting enhanced fear extinc-


tion (Dadkhah et al., 2018). As glucocorticoid receptors are also re-
hippocampus

quired for glucose homeostasis and play a role in the development of


hyperinsulinemia and obesity (Majer-Lobodzinska and Adamiec-
Mroczek, 2017) they are suspected as key players in neuroadaptations
to the AMG, hippocampus and PFC that occur after long-term sucrose
consumption.
Salivary cortisol, Montreal Imaging

Juxtaposing the combined psychological and physical effects high


sugar diets have on pathological fear has, until recently, been an un-
touched topic of research. Although no studies have been conducted on
sugar consumption alone with regards to pathological fear, some re-
environment for cognitive stimulation and emotional support. 3 I2BC: intermittent 2 bottle choice. 4 LTP: long-term potentiation.

search has begun to explore the combined effect of high-fat/sugar or


high sugar and high carbohydrate diets (Santos et al., 2018; Baker and
Stress Task

Reichelt, 2016; Williams-Spooner et al., 2017; Reichelt et al., 2015).


These studies are beginning to delineate the impact high caloric foods
Tasks

can have on the intensity and duration of pathological fear; however


research into the effect of sugar alone is required to delineate the me-
chanisms involved. More recent research has examined contextual fear
following a standard contextual fear conditioning protocol which was
contradictory to this finding (Santos et al., 2018). Specifically, high-
sugar/carbohydrate diets significantly enhance fear-related freezing to
Nineteen women

context (Santos et al., 2018). Whilst both examine contextual fear


(age 18–40 y)

memory formation, different fear conditioning protocols were used


Subjects

(trace fear conditioning versus contextual fear conditioning). Indeed


hippocampal function in contextual fear memory has been noted as
disparate, depending on the fear conditioning protocol (see recent re-
view by (Chaaya et al., 2018)). Nevertheless, these studies highlight
how diet can influence hippocampal-dependent memory, as well as
Tyron, M.S., et al., 2015

hippocampal function (Santos et al., 2018; Reichelt et al., 2015).


Fear extinction is a protocol whereby, following excessive exposure
(Tryon et al., 2015)
Authors and year

to the previously neutral fear conditioning CS (e.g. the context in


contextual fear conditioning, or a tone in auditory fear conditioning), a
reduction of fear-related freezing is seen (Maren, 2011). This reduction
in fear is similar to basic theories surrounding exposure therapy,
whereby excessive exposure to fear-inducing stimuli (via mental ima-
gery, for example) results in a reduction in fear-related symptoms
consumption three times per day for 2 weeks

(Maren, 2011; Milad and Quirk, 2012).


Sucrose and sweetener consumption in humans

Investigation into the effect of PTSD symptoms on the consumption


Sucrose- or aspartame-sweetened beverage

of highly palatable ‘fast’ food and sweetened beverages was conducted


to see if there was a correlation with emotional eating behaviours and
body mass index (Hirth et al., 2011). The answers to questions re-
garding frequency of consuming fast food were collected from 3154
females and analyzed using regression analyses (Hirth et al., 2011). To
determine unhealthy eating habits, participants were asked if they used
Table 3 (continued)

diet pills, laxative, diuretics, skipped meals or vomited after eating.


Findings suggested that PTSD symptoms increased the frequency of
consumption of high caloric food and sodas, as well as contributing to
unhealthy eating habits but did not increase overall body mass index
(Hirth et al., 2011). Cumulatively, data from these various studies show
a strong link between PTSD, fear memory and caloric foods suggesting a

191
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

possible correlation with high sugar consumption that requires further (Gillies et al., 2007). The dilemma with lifestyle interventions is that
investigation. regain of the weight lost is common after a period of time and often
results in weight gain greater than that originally lost (Barte et al.,
6. Sucrose consumption investigated 2010). Strategies that proved effective in maintaining weight loss in-
cluded a consistent regular meal pattern that included breakfast, a high
Negative states of emotion driven by substances of abuse are often expectation to succeed, a good support system and behavioural self-
shown by examining anxiety-like behaviours, decreased pain tolerance, monitoring (Barte et al., 2010). Mindfulness meditation is another in-
or an increase in the point at which reward becomes sufficient to pro- tervention studied as a way to regulate emotional eating. A self-re-
duce a stimulating effect and memory deficits (Koob, 2018). The ported survey was conducted to examine whether mindfulness practice
complexities of the systems involved, the interconnectedness of the could alter emotional eating (Levoy et al., 2017). Surveys conducted
systems and the myriad of protocols used to study sucrose dependence before and after the intervention revealed lower emotional eating
could potentially lead to a wide variety of outcomes and yet overall scores after the meditation, suggesting a possible role for mindfulness as
investigations into high-sugar consumption in rats, mice and humans a treatment for emotional eating (Levoy et al., 2017). Mindfulness
result in similar outcomes with regards to memory, stress and emotion studies do not show a direct effect on weight loss itself, but appear to be
(see Table 3.) quite successful in lessening the addictive-like behaviours pursuant to
relapse of weight gain (Moor et al., 2013; Goldbacher et al., 2016;
7. Therapeutics for obesity, derived from studies of sucrose Keesman et al., 2017).
consumption
7.3. Digital technology
7.1. Pharmacological approaches
Decreasing emotional eating has been proposed as a potential me-
Sucrose is shown to be addictive in rodents (Avena et al., 2008; chanism for the long-term maintenance of weight loss (Barte et al.,
Colantuoni et al., 2002; Rada et al., 2005) and the studies listed in 2010). Digital technology may be our best hope of achieving this goal.
Table 3 show correlations between sucrose consumption and emotional Interaction with online environments provide the social support often
disturbances. The NK1- (Neurokinin-1) receptor system is involved in lacking during maintenance of weight loss (Hwang et al., 2010). Apps
the reinforcement mechanism that motivates the desire to have stimuli for exercise regimes, food tracking, meditation and positive thinking all
that no longer create pleasure, a characteristic of addiction (Sandweiss provide a support system to the user and promote networking with
and Vanderah, 2015), and is implicated in both anxiety and depression others in similar situations. Support networks are useful tools as in-
(Mantyh, 2002). Using the intermittent access model with a 5% sucrose creases in body weight decrease an individual’s ability to make in-
solution, our laboratory showed the NK1-receptor antagonist ezlopitant formed decisions regarding highly palatable food, resist temptation and
(which possesses anxiolytic effects) was able to attenuate and inhibit regulate their emotions (Yeomans, 2017). These challenges were sup-
sucrose intake in Long Evans rats (Steensland et al., 2010). This finding ported through a study of 17 obese women and their physiological re-
suggests the NK1- receptor system to be a potential target for sugar- actions to verbal food cues which found that food and beverage pre-
related obesity therapeutics (Steensland et al., 2010). Other potential ferences affected physiologic responses as well as cognition and
options include antidiabetic drugs such as sodium-glucose co- attention (Fioravanti et al., 2004).
transporter 2 (SGLT2) inhibitors, which impede renal glucose re- Wearable technology such as Fitbits (activity trackers), that interact
absorption and are generally considered effective, though lose efficacy directly with the individual, provide information about the individual
when taken long term (Gillies et al., 2007). Indeed, a 4 week oral ad- and promote long term maintenance and support models of healthcare
ministration of ipragliflozin, (a SGLT2 inhibitor) with antidiabetic ef- where the patient takes an active role in their own wellness (Handel,
fects on type 2 diabetes, decreases the caloric balance and improves 2011; Sama et al., 2014). Wearable technologies appear to be extremely
symptoms of diabetes, including obesity in mice fed on 20% glucose or effective in assisting weight loss in patients with serious mental illness
sucrose solution (Tahara et al., 2018; Chao and Henry, 2010) (Aschbrenner et al., 2016). Thirteen obese individuals diagnosed with a
A recent review detailing the current management for type 2 dia- serious mental illness (e.g. schizophrenia and major depressive dis-
betes suggested a step wise approach; including diet and exercise the order) participated in 24 weeks of behavioural weight loss intervention
patient would take metformin (which lowers high blood glucose levels encompassing group sessions for weight management and exercise,
by decreasing the production of hepatic glucose, increasing insulin mobile health technology and social media for motivation, peer support
sensitivity and lowering intestinal glucose absorption (Rena et al., and a self-monitoring tool (Aschbrenner et al., 2016). At the end of 6
2017)), a glucagon-like peptide 1 receptor agonist (to increase insulin months of interventions, 45% of participants had reduced their weight
output from the pancreas (Drucker and Nauck, 2006)) or a SCLT2 in- (below their baseline) and increased fitness (as measured through
hibitor in addition to one of the approved weight-loss drugs (to control walking distance) (Aschbrenner et al., 2016).
appetite or the absorption of calories consumed (Padwal and
Majumdar, 2007)) (Burguera et al., 2017). Table 4 lists doses and me- 8. Discussion
chanisms of action of therapeutics used in trials to alter sucrose con-
sumption in an effort to discover alternate treatments for diabetes and The data summarized in this article suggests overconsumption of
obesity. sugar can lead to brain adaptations involving many different neural
systems, molecular substrates and subsequent changes in behaviour.
7.2. Lifestyle interventions The ease of availability and cost effectiveness of high caloric, sweetened
food and beverages appear to be a contributing factor in the world wide
Diet and exercise are highly recommended approaches to treatment increase in obesity (Sinha, 2018; He et al., 2018). Paradigms designed
for obesity (Bray and Bouchard, 2014). A 2007 systematic review and to investigate emotional eating show increases in weight gain due to
meta-analysis that evaluated lifestyle interventions for patients with higher caloric intake, nevertheless examination of the correlation be-
impaired glucose tolerance looked at 21 randomized controlled trials tween environmental and social inputs, individual thought processes
designed to delay or prevent the onset of type 2 diabetes (Gillies et al., and behaviour that maintains emotional eating is yet to be defined. It
2007). Data revealed evidence in support of interventions such as diet seems feasible to suggest the pleasurable sensations brought about
and exercise being at least as effective as oral diabetes drugs, a Chinese through the consumption of sugar may provide a self-medicating
herbal remedy (jiangtang bushen) and the anti-obesity drug orlistat method to deal with daily stresses (Fortuna, 2010; Brewerton, 2011).

192
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

Table 4
Therapeutics used in sugar consumption trials.
Author, year Drug / Dose Mechanism of action Subjects Findings

Richard, D., et al., 2000 (Richard et al., Topiramate (30 mg / kg) Blocks voltage gated sodium and calcium channels, ↓ Rats ↓ Weight
2000) glutamate, ↑ GABA (Anticonvulsant) ↓ sucrose intake
Beczkowska, I.W., 1992 (Beczkowska et al., Naltrexone Naloxone Kappa and Mu2 (opioid) receptor antagonists (Treat drug Rats ↓Sucrose intake
1992) and alcohol dependence
Steensland, P., et al., 2010 (Steensland et al., Ezlopitant (2,5,10 mg / kg) NK1 receptor antagonist (Anxiolytic and antiemetic) Rats ↓ obesity
2010)
Shariff, M., 2016 (Shariff et al., 2016) Varenicline (0.3,1,2 mg / kg) nAChR partial agonist (Smoking cessation) Rats ↓ sucrose intake
Tahara, A., et al., 2018 (Tahara et al., 2018) Ipragliflozin (0.1 – 3 mg / kg) SGLT2 inhibitor (Antidiabetic) Mice ↓ sucrose intake
Muscat, R. and Willner, P., 1989 (Muscat and Sulpiride (20,40 mg / kg) D2 and D3 receptor elective antagonist (Antipsychotic) Rats ↓ desire for sucrose
Willner, 1989)
Lof, E., et al., 2010 (Löf et al., 2010) Methyllycaconitine (0.3,1 mg/kg) α7 nAChRs selective antagonist Rats ↓ sucrose preference
Patkar, O., et al., 2017 (Patkar et al., 2017) Buspirone (1,2.5,5 mg / kg) 5-HT1A/1B partial agonist (Anxiolytic) Mice ↓ sucrose consumption
Lin, Z., et al., 2013 (Lin et al., 2013) Curcumin (40 mg / kg) Upregulation of PPAR-γ activation (Herbal supplement) Rats ↓ sucrose intake
Kurhe, Y., et al., 2014 (Kurhe et al., 2014) Ondansetron (1 mg / kg) Serotonin receptor (5-HT3) antagonist (Antiemetic) Mice ↑ Sucrose consumption
Badia‐Elder, N.E., et al., 2003 (Badia‐Elder NPY (5 μg / 5 μl) (10 μg / 10 μl) Inhibits GAD67 expression (Vasoconstrictor) Rats ↑ sucrose intake
et al., 2003)
Pandit, R., et al., 2015 (Pandit et al., 2015) AgRP (0.66 nmol) (1 nmol) MC 3/4 receptor inverse agonist (Decreases metabolism) Rats ↑ motivation for
sucrose

Common hedonistic mechanisms play a role in both obesity and ad- mechanism which induces the desire to overeat in the hope to restore
diction to drugs of abuse (Johnson and Kenny, 2010). While the root homeostatic dopamine levels and avoid mild depression. Opioids,
cause of obesity remains elusive, the estimated global annual healthcare which induce feeding through an abundance of brain regions, may be
cost of treating illnesses related to obesity may reach US $1.2 trillion responsible for cue-induced relapse into overeating behaviours, and
per year by 2025. binging on sugar postpones the release of acetylcholine required to
Every day new digital applications are being developed to assist our signal satiety. Each of these neuroadaptations implicates sucrose con-
pursuit of health and happiness. In the future it may be possible to sumptions ability to alter the way we perceive and process our emotions
collect personal information about the consumption of high-sugar foods and consequential behaviour. Perhaps a greater understanding of the
and beverages in conjunction with emotions felt on a daily basis. The neural mechanisms of impulsivity and overeating are required to assist
possibilities of personal wellness monitoring, implantable in vivo in the development of improved obesity treatments.
monitoring and drug delivery devices will also require further robust It has been estimated that by 2020, 1.5 million people will die each
study before they prove to be effective treatments for obesity and food year by suicide, with 15 to 30 million attempting it (Weissman et al.,
addiction. It would be interesting to investigate a possible correlation 1999). Children suffering anxiety disorders are twice as likely to at-
between sugar consumption, emotions and body mass index tempt suicide, while those suffering major depressive illnesses show a 3
(BMI > 30 denotes obesity). It would also be significant to know if fold chance at attempt (Weissman et al., 1999).In support of these
such applications combining game rewards might assist with the statistics, over 300 million people were reported as suffering depression
childhood obesity epidemic as current predictions state that globally, in 2016/17, with 264 million people reported to be suffering from
2.7 billion adults will be overweight and/or obese by 2025 (Haby et al., anxiety disorders (Organization, 2017). This review has examined how
2012). negative emotion can exacerbate sugar overconsumption, and vice
Functional brain imaging studies substantiate the higher preference versa. If negative emotions are so prevalent in our children, and sugar
and increased emotional activation that occurs in response to images intake so common, its consumption may be considered a threat to the
and verbal cues related to high sugar content foods and beverages, emotional stability of our race (see reviews on mental health in children
making it more difficult for overweight individuals to resist eating and adolescents (Wehry et al., 2015; Beesdo et al., 2009; Pine et al.,
unhealthy food (Batterink et al., 2010; Pursey et al., 2014). Optogenetic 2002)). More importantly, reduction of sugar overconsumption may be
and chemogenetic studies may assist in defining the combination of capable of significantly reducing the prevalence of negative emotion in
neural pathways and substrates involved. Other tools designed to un- a vast number of individuals around the world.
ravel the complexities between neuronal organisation and behaviour
include methods to quantify cellular populations that are functional for Funding
particular behaviours (Bergstrom et al., 2011). Using analytic methods
(including micro-binning and density mapping) to accurately compare This research was funded by NHMRC project grant (GNT1146417)
functional neural network activity it may be possible to produce mi- and Lady Cilento Children’s Hospital Research Foundation.
croanatomical topography of molecular activity resultant from the in-
fluence of long term sugar consumption on a variety of stress and an- Conflicts of interest
xiety related behaviours (Jacques et al., 2018). This would assist in
demonstrating pathophysiological neuroadaptive changes and perhaps The founding sponsors had no role in the design of the study; in the
lead to the development of enhanced pharmacotherapeutic and tech- collection, analyses, or interpretation of data; in the writing of the
nological strategies to assist the reduction of excess sugar consumption. manuscript, and in the decision to publish the results. S.E.B is currently
In conclusion, the ease of access to sugar rich diets today is an en- examining the role of emotions and high-fat/sugar containing foods
vironmental contributor to obesity, but it may be sugars ability to using digital technology platform.
generate a superior neurological reward signal which overrides self-
control mechanisms and leads to obesity (Lenoir et al., 2007; Yeomans, Acknowledgments
2017). Obesity and long term sugar consumption both result in low
basal levels of dopamine, particularly in the NAc, which may be the N/A.

193
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

References Beilharz, J.E., Maniam, J., Morris, M.J., 2014. Short exposure to a diet rich in both fat and
sugar or sugar alone impairs place, but not object recognition memory in rats. Brain
Behav. Immun. 37, 134–141.
Acevedo, B.P., Aron, A., Fisher, H.E., Brown, L.L., 2012. Neural correlates of long-term Belin, D., Dalley, J.W., 2012. Animal models in addiction research. Drug Abuse and
intense romantic love. Soc. Cogn. Affect. Neurosci. 7, 145–159. Addiction in Medical Illness. Springer, pp. 73–93.
Adcock, R.A., Thangavel, A., Whitfield-Gabrieli, S., Knutson, B., Gabrieli, J.D., 2006. Bello, N.T., Lucas, L.R., Hajnal, A., 2002. Repeated sucrose access influences dopamine
Reward-motivated learning: mesolimbic activation precedes memory formation. D2 receptor density in the striatum. Neuroreport 13 (1575).
Neuron 50, 507–517. Bello, E.P., et al., 2011. Cocaine supersensitivity and enhanced motivation for reward in
Adinoff, B., 2004. Neurobiologic processes in drug reward and addiction. Harv. Rev. mice lacking dopamine D 2 autoreceptors. Nat. Neurosci. 14 (1033).
Psychiatry 12, 305–320. Belzeaux, R., Lalanne, L., Kieffer, B.L., Lutz, P.-E., 2018. Focusing on the opioid system for
Ahima, R.S., Antwi, D.A., 2008. Brain regulation of appetite and satiety. Endocrinol. addiction biomarker discovery. Trends Mol. Med. 24, 206–220.
Metab. Clin. North Am. 37, 811–823. Benarroch, E.E., 2012. Endogenous opioid systems Current concepts and clinical corre-
Ahmed, S.H., Guillem, K., Vandaele, Y., 2013. Sugar addiction: pushing the drug-sugar lations. Neurology 79, 807–814.
analogy to the limit. Curr. Opin. Clin. Nutr. Metab. Care 16, 434–439. Benoit, S.C., Tracy, A.L., Davis, J.F., Choi, D., Clegg, D.J., 2008. Novel functions of or-
Alsiö, J., Olszewski, P.K., Levine, A.S., Schiöth, H.B., 2012. Feed-forward mechanisms: exigenic hypothalamic peptides: from genes to behavior. Nutrition 24, 843–847.
addiction-like behavioral and molecular adaptations in overeating. Front. Benowitz, N.L., 1988. Pharmacologic aspects of cigarette smoking and nicotine addiction.
Neuroendocrinol. 33, 127–139. N. Engl. J. Med. 319, 1318–1330.
Anderson, L.C., Petrovich, G.D., 2018. Distinct recruitment of the hippocampal, thalamic, Benton, D., 2010. The plausibility of sugar addiction and its role in obesity and eating
and amygdalar neurons projecting to the prelimbic cortex in male and female rats disorders. Clin. Nutr. 29, 288–303.
during context-mediated renewal of responding to food cues. Neurobiol. Learn. Mem. Bergstrom, H.C., McDonald, C.G., Johnson, L.R., 2011. Pavlovian fear conditioning ac-
150, 25–35. tivates a common pattern of neurons in the lateral amygdala of individual brains.
Anderson, E.M., et al., 2018. Overexpression of the histone dimethyltransferase G9a in PLoS One 6, e15698.
nucleus accumbens shell increases cocaine self-administration, stress-induced re- Berridge, K.C., 1996. Food reward: brain substrates of wanting and liking. Neurosci.
instatement, and anxiety. J. Neurosci. 38, 803–813. Biobehav. Rev. 20, 1–25.
Aosaki, T., Miura, M., Suzuki, T., Nishimura, K., Masuda, M., 2010. Berridge, K.C., Ho, C.-Y., Richard, J.M., DiFeliceantonio, A.G., 2010. The tempted brain
Acetylcholine–dopamine balance hypothesis in the striatum: an update. Geriatr. eats: pleasure and desire circuits in obesity and eating disorders. Brain Res. 1350,
Gerontol. Int. 10. 43–64.
Aringhieri, S., et al., 2018. Molecular targets of atypical antipsychotics: from mechanism Berthoud, H.-R., Münzberg, H., Morrison, C.D., 2017. Blaming the brain for obesity: in-
of action to clinical differences. Pharmacol. Ther. tegration of hedonic and homeostatic mechanisms. Gastroenterology 152,
Aschbrenner, K.A., Naslund, J.A., Shevenell, M., Mueser, K.T., Bartels, S.J., 2016. 1728–1738.
Feasibility of behavioral weight loss treatment enhanced with peer support and Bianchi, P.C., et al., 2018. Functional inactivation of the orbitofrontal cortex disrupts
mobile health technology for individuals with serious mental illness. Psychiatr. Q. 87, context-induced reinstatement of alcohol seeking in rats. Drug Alcohol Depend. 186,
401–415. 102–112.
Asnaghi, V., Gerhardinger, C., Hoehn, T., Adeboje, A., Lorenzi, M., 2003. A role for the Bienkowski, P., et al., 2004. Time-dependent changes in alcohol-seeking behaviour during
polyol pathway in the early neuroretinal apoptosis and glial changes induced by abstinence. Eur. Neuropsychopharmacol. 14, 355–360.
diabetes in the rat. Diabetes 52, 506–511. Bito‐Onon, J.J., Simms, J.A., Chatterjee, S., Holgate, J., Bartlett, S.E., 2011. Varenicline, a
Association, 2013. A.P. Diagnostic and Statistical Manual of Mental Disorders (DSM-5®). partial agonist at neuronal nicotinic acetylcholine receptors, reduces nicoti-
American Psychiatric Pub. ne‐induced increases in 20% ethanol operant self‐administration in Sprague‐Dawley
Atkins, C.M., Selcher, J.C., Petraitis, J.J., Trzaskos, J.M., Sweatt, J.D., 1998. The MAPK rats. Addict. Biol. 16, 440–449.
cascade is required for mammalian associative learning. Nat. Neurosci. 1, 602. BjÖrntorp, P., 2000. Hypertension and the metabolic syndrome: closely related central
Avena, N.M., Hoebel, B., 2003. A diet promoting sugar dependency causes behavioral origin? Blood Press. 9, 71–82.
cross-sensitization to a low dose of amphetamine. Neuroscience 122, 17–20. Bocarsly, M.E., 2018. Pharmacological interventions for obesity: current and future tar-
Avena, N.M., Long, K.A., Hoebel, B.G., 2005. Sugar-dependent rats show enhanced re- gets. Curr. Addict. Rep. 5, 202–211.
sponding for sugar after abstinence: evidence of a sugar deprivation effect. Physiol. Bordet, R., et al., 1997. Induction of dopamine D3 receptor expression as a mechanism of
Behav. 84, 359–362. behavioral sensitization to levodopa. Proc. Natl. Acad. Sci. 94, 3363–3367.
Avena, N.M., Rada, P., Moise, N., Hoebel, B., 2006. Sucrose sham feeding on a binge Bouchet, C.A., et al., 2018. Activation of nigrostriatal dopamine neurons during fear
schedule releases accumbens dopamine repeatedly and eliminates the acetylcholine extinction prevents the renewal of fear. Neuropsychopharmacology 43, 665.
satiety response. Neuroscience 139, 813–820. Bray, G.A., Bouchard, C., 2014. Handbook of Obesity–Volume 2: Clinical Applications.
Avena, N.M., Rada, P., Hoebel, B.G., 2008. Evidence for sugar addiction: behavioral and CRC Press.
neurochemical effects of intermittent, excessive sugar intake. Neurosci. Biobehav. Brewerton, T.D., 2011. Posttraumatic Stress Disorder and Disordered Eating: Food
Rev. 32, 20–39. Addiction As Self-medication. Mary Ann Liebert, Inc., 140 Huguenot Street, 3rd Floor
Avena, N.M., Rada, P., Hoebel, B.G., 2009. Sugar and fat bingeing have notable differ- New Rochelle, NY 10801 USA.
ences in addictive-like behavior. J. Nutr. 139, 623–628. Brown, R.J., Rother, K.I., 2012. Non-nutritive sweeteners and their role in the gastro-
Azimova, K., et al., 2015. Glucose levels and depression in Hispanic patients admitted to intestinal tract. J. Clin. Endocrinol. Metab. 97, 2597–2605.
the cardiovascular intensive care unit: a cross-sectional study. Angiology 66, 57–64. Bryant, R., et al., 2008a. Amygdala and ventral anterior cingulate activation predicts
Badia‐Elder, N.E., Stewart, R., Powrozek, T.A., Murphy, J., Li, T.K., 2003. Effects of treatment response to cognitive behaviour therapy for post-traumatic stress disorder.
neuropeptide Y on sucrose and ethanol intake and on anxiety‐like behavior in high Psychol. Med. 38, 555–561.
alcohol drinking (HAD) and low alcohol drinking (LAD) rats. Alcohol. Clin. Exp. Res. Bryant, R.A., et al., 2008b. Rostral anterior cingulate volume predicts treatment response
27, 894–899. to cognitive-behavioural therapy for posttraumatic stress disorder. J. Psychiatry
Bailey, A., Gianotti, R., Ho, A., Kreek, M.J., 2005. Persistent upregulation of μ‐opioid, but Neurosci.: JPN 33, 142.
not adenosine, receptors in brains of long‐term withdrawn escalating dose “binge” Burguera, B., Ali, K., Brito, J., 2017. Antiobesity drugs in the management of type 2
cocaine‐treated rats. Synapse 57, 160–166. diabetes: a shift in thinking? Cleve. Clin. J. Med. 84, S39–S46.
Baker, K.D., Reichelt, A.C., 2016. Impaired fear extinction retention and increased an- Burnett, C.J., et al., 2016. Hunger-driven motivational state competition. Neuron 92,
xiety-like behaviours induced by limited daily access to a high-fat/high-sugar diet in 187–201.
male rats: implications for diet-induced prefrontal cortex dysregulation. Neurobiol. Cabanac, M., Johnson, K., 1983. Analysis of a conflict between palatability and cold
Learn. Mem. 136, 127–138. exposure in rats. Physiol. Behav. 31, 249–253.
Bakke, A.J., et al., 2018. Mary Poppins was right: adding small amounts of sugar or salt Calipari, E.S., et al., 2014. Amphetamine self-administration attenuates dopamine D2
reduces the bitterness of vegetables. Appetite 126, 90–101. autoreceptor function. Neuropsychopharmacology 39 (1833).
Barnes, J.N., Joyner, M.J., 2012. Sugar highs and lows: the impact of diet on cognitive Cao, D., Lu, H., Lewis, T.L., Li, L., 2007. Intake of sucrose-sweetened water induces insulin
function. J. Physiol. 590 2831-2831. resistance and exacerbates memory deficits and amyloidosis in a transgenic mouse
Baron, D., Blum, K., Chen, A., Gold, M., Badgaiyan, R.D., 2018. Conceptualizing addiction model of Alzheimer disease. J. Biol. Chem. 282, 36275–36282.
from an osteopathic perspective: dopamine homeostasis. J. Am. Osteopath. Assoc. Capello, A.E., Markus, C.R., 2014. Differential influence of the 5-HTTLPR genotype,
118, 115–118. neuroticism and real-life acute stress exposure on appetite and energy intake.
Barrientos, C., et al., 2018. Cocaine induced structural plasticity in input regions to dis- Appetite 77, 83–93.
tinct cell types in nucleus accumbens. Biol. Psychiatry. Carr, D., Kalivas, P.W., 2006. Orexin: a gatekeeper of addiction. Nat. Med. 12, 274.
Barte, J., et al., 2010. Maintenance of weight loss after lifestyle interventions for over- Cha, S.H., Wolfgang, M., Tokutake, Y., Chohnan, S., Lane, M.D., 2008. Differential effects
weight and obesity, a systematic review. Obes. Rev. 11, 899–906. of central fructose and glucose on hypothalamic malonyl–CoA and food intake. Proc.
Batterink, L., Yokum, S., Stice, E., 2010. Body mass correlates inversely with inhibitory Natl. Acad. Sci.
control in response to food among adolescent girls: an fMRI study. Neuroimage 52, Chaaya, N., Battle, A.R., Johnson, L.R., 2018. An update on contextual fear memory
1696–1703. mechanisms: transition between amygdala and Hippocampus. Neurosci. Biobehav.
Beczkowska, I.W., Bowen, W.D., Bodnar, R.J., 1992. Central opioid receptor subtype Rev In Press.
antagonists differentially alter sucrose and deprivation-induced water intake in rats. Chao, E.C., Henry, R.R., 2010. SGLT2 inhibition—a novel strategy for diabetes treatment.
Brain Res. 589, 291–301. Nat. Rev. Drug Discov. 9, 551.
Beesdo, K., Knappe, S., Pine, D.S., 2009. Anxiety and anxiety disorders in children and Chepulis, L.M., Starkey, N.J., Waas, J.R., Molan, P.C., 2009. The effects of long-term
adolescents: developmental issues and implications for DSM-V. Psychiatric Clinics 32, honey, sucrose or sugar-free diets on memory and anxiety in rats. Physiol. Behav. 97,
483–524. 359–368.

194
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

Chikama, K., et al., 2017. Chronic atypical antipsychotics, but not haloperidol, increase Foley, K.A., Fudge, M.A., Kavaliers, M., Ossenkopp, K.-P., 2006. Quinpirole-induced be-
neurogenesis in the hippocampus of adult mouse. Brain Res. 1676, 77–82. havioral sensitization is enhanced by prior scheduled exposure to sucrose: a multi-
Clifton, P.G., 2017. Neural circuits of eating behaviour: opportunities for therapeutic variable examination of locomotor activity. Behav. Brain Res. 167, 49–56.
development. J. Psychopharmacol. 31, 1388–1402. Foo, H., Mason, P., 2005. Sensory suppression during feeding. Proc. Natl. Acad. Sci. 102,
Cocores, J.A., Gold, M.S., 2008. Varenicline, appetite, and weight reduction. J. 16865–16869.
Neuropsychiatry Clin. Neurosci. 20, 497–498. Fortuna, J.L., 2010. Sweet preference, sugar addiction and the familial history of alcohol
Cohen, J.F., Rifas-Shiman, S.L., Young, J., Oken, E., 2018. Associations of prenatal and dependence: shared neural pathways and genes. J. Psychoactive Drugs 42, 147–151.
child sugar intake with child cognition. Am. J. Prev. Med. 54, 727–735. Gangwisch, J.E., et al., 2015. High glycemic index diet as a risk factor for depression:
Colantuoni, C., et al., 2001. Excessive sugar intake alters binding to dopamine and mu- analyses from the Women’s Health Initiative. Am. J. Clin. Nutr. 102, 454–463.
opioid receptors in the brain. Neuroreport 12, 3549–3552. García-Cáceres, C., Quarta, C., Varela, L., Gao, Y., Gruber, T., Legutko, B., et al., 2016.
Colantuoni, C., et al., 2002. Evidence that intermittent, excessive sugar intake causes Astrocytic insulin signaling couples brain glucose uptake with nutrient availability.
endogenous opioid dependence. Obesity 10, 478–488. Cell 166, 867–880.
Comings, D., et al., 2001. The additive effect of neurotransmitter genes in pathological Gaysinskaya, V., Karatayev, O., Shuluk, J., Leibowitz, S., 2011. Hyperphagia induced by
gambling. Clin. Genet. 60, 107–116. sucrose: relation to circulating and CSF glucose and corticosterone and orexigenic
Cools, R., Barker, R.A., Sahakian, B.J., Robbins, T.W., 2003. L-Dopa medication re- peptides in the arcuate nucleus. Pharmacol. Biochem. Behav. 97, 521–530.
mediates cognitive inflexibility, but increases impulsivity in patients with Parkinson’s Gearhardt, A.N., Corbin, W.R., Brownell, K.D., 2009. Preliminary validation of the Yale
disease. Neuropsychologia 41, 1431–1441. food addiction scale. Appetite 52, 430–436.
Coplan, J.D., Webler, R., Gopinath, S., Abdallah, C.G., Mathew, S.J., 2018. Neurobiology Geiger, B.M., et al., 2008. Evidence for defective mesolimbic dopamine exocytosis in
of the dorsolateral prefrontal cortex in GAD: aberrant neurometabolic correlation to obesity-prone rats. FASEB J. 22, 2740–2746.
hippocampus and relationship to anxiety sensitivity and IQ. J. Affect. Disord. 229, Gerdeman, G.L., Partridge, J.G., Lupica, C.R., Lovinger, D.M., 2003. It could be habit
1–13. forming: drugs of abuse and striatal synaptic plasticity. Trends Neurosci. 26,
Corsica, J.A., Pelchat, M.L., 2010. Food addiction: true or false? Curr. Opin. 184–192.
Gastroenterol. 26, 165–169. Gillies, C.L., et al., 2007. Pharmacological and lifestyle interventions to prevent or delay
Corsini, R.J., 1999. The Dictionary of Psychology. Psychology Press. type 2 diabetes in people with impaired glucose tolerance: systematic review and
Cortese, B.M., Phan, K.L., 2005. The role of glutamate in anxiety and related disorders. meta-analysis. BMJ 334, 299.
CNS Spectr. 10, 820–830. Giustino, T.F., Maren, S., 2015. The role of the medial prefrontal cortex in the con-
Covington, H.E., Miczek, K.A., 2001. Repeated social-defeat stress, cocaine or morphine. ditioning and extinction of fear. Front. Behav. Neurosci. 9, 298.
Psychopharmacology 158, 388–398. Goldbacher, E., La Grotte, C., Komaroff, E., Vander Veur, S., Foster, G.D., 2016. An initial
Criscitelli, K., Avena, N.M., 2017. 5 Sugar and Fat Addiction. Processed Food Addiction: evaluation of a weight loss intervention for individuals who engage in emotional
Foundations, Assessment, and Recovery. eating. J. Behav. Med. 39, 139–150.
D’Anci, K.E., Kanarek, R.B., Marks-Kaufman, R., 1996. Duration of sucrose availability Gonzalez, J.T., Betts, J.A., 2018. Dietary fructose metabolism by splanchnic organs: size
differentially alters morphine-induced analgesia in rats. Pharmacol. Biochem. Behav. matters. Cell Metab. 27, 483–485.
54, 693–697. Gosnell, B., 1987. Central structures involved in opioid-induced feeding. Fed. Proc.
D’Argenio, A., et al., 2009. Early trauma and adult obesity: is psychological dysfunction 163–167.
the mediating mechanism? Physiol. Behav. 98, 543–546. Gosnell, B.A., 2005. Sucrose intake enhances behavioral sensitization produced by co-
Dadkhah, M., Abdullahi, P.R., Rashidy-Pour, A., Sameni, H.R., Vafaei, A.A., 2018. caine. Brain Res. 1031, 194–201.
Infralimbic dopamine D2 receptors mediate glucocorticoid-induced facilitation of Graham, M., Shutter, J.R., Sarmiento, U., Stark, K.L., 1997. Overexpression of Agrt leads
auditory fear memory extinction in rats. Brain Res. 1682, 84–92. to obesity in transgenic mice. Nat. Genet. 17, 273.
Dallman, M.F., et al., 2003. A spoonful of sugar: feedback signals of energy stores and Grant, A., Christie, M., Ashcroft, S., 1980. Insulin release from human pancreatic islets in
corticosterone regulate responses to chronic stress. Physiol. Behav. 79, 3–12. vitro. Diabetologia 19, 114–117.
Davidson, T.L., et al., 2009. Contributions of the hippocampus and medial prefrontal Gueye, A.B., et al., 2018. Unlimited sucrose consumption during adolescence generates a
cortex to energy and body weight regulation. Hippocampus 19, 235–252. depressive-like phenotype in adulthood. Neuropsychopharmacology 1.
de Jong, J.W., 2015. Eating Addiction? The Nerves and Fibers That Control Food Intake. Guillin, O., et al., 2001. BDNF controls dopamine D 3 receptor expression and triggers
Utrecht University. behavioural sensitization. Nature 411, 86.
De Jong, J.W., Vanderschuren, L.J., Adan, R.A., 2016. The mesolimbic system and eating Gulpinar, M., Yegen, B., 2004. The physiology of learning and memory: role of peptides
addiction: what sugar does and does not do. Curr. Opin. Behav. Sci. 9, 118–125. and stress. Curr. Protein Pept. Sci. 5, 457–473.
De Nijs, J., et al., 2018. O9. 7. Individualized LONG-TERM outcome prediction of psy- Gunstad, J., et al., 2006. Exposure to early life trauma is associated with adult obesity.
chosis in an observational study: a machine learning approach. Schizophr. Bull. 44, Psychiatry Res. 142, 31–37.
S101–S102. Guo, X., et al., 2014. Sweetened beverages, coffee, and tea and depression risk among
Denis, R.G., et al., 2015. Palatability can drive feeding independent of AgRP neurons. Cell older US adults. PLoS One 9, e94715.
Metab. 22, 646–657. Gurevich, E.V., et al., 1997. Mesolimbic dopamine D3 receptors and use of antipsychotics
Derman, R.C., Ferrario, C.R., 2017. Enhanced incentive motivation in obesity-prone rats in patients with schizophrenia: a postmortem study. Arch. Gen. Psychiatry 54,
is mediated by NAc core CP-AMPARs. Neuropharmacology. 225–232.
Di Chiara, G., Imperato, A., 1988. Drugs abused by humans preferentially increase sy- Haby, M.M., Markwick, A., Peeters, A., Shaw, J., Vos, T., 2012. Future predictions of body
naptic dopamine concentrations in the mesolimbic system of freely moving rats. Proc. mass index and overweight prevalence in Australia, 2005–2025. Health Promot. Int.
Natl. Acad. Sci. 85, 5274–5278. 27, 250–260.
Díaz-Aguila, Y., et al., 2016. Consumption of sucrose from infancy increases the visceral Hajnal, A., Smith, G.P., Norgren, R., 2004. Oral sucrose stimulation increases accumbens
fat accumulation, concentration of triglycerides, insulin and leptin, and generates dopamine in the rat. Am. J. Physiol. Regul. Integr. Comp. Physiol. 286, R31–R37.
abnormalities in the adrenal gland. Anat. Sci. Int. 91, 151–162. Halford, J.C., Boyland, E.J., Blundell, J.E., Kirkham, T.C., Harrold, J.A., 2010.
Domingos, A.I., et al., 2013. Hypothalamic melanin concentrating hormone neurons Pharmacological management of appetite expression in obesity. Nat. Rev. Endocrinol.
communicate the nutrient value of sugar. Elife 2, e01462. 6, 255.
Drevets, W.C., 1998. Functional neuroimaging studies of depression: the anatomy of Han, F., Ding, J., Shi, Y., 2014. Expression of amygdala mineralocorticoid receptor and
melancholia. Annu. Rev. Med. 49, 341–361. glucocorticoid receptor in the single-prolonged stress rats. BMC Neurosci. 15, 77.
Drucker, D.J., Nauck, M.A., 2006. The incretin system: glucagon-like peptide-1 receptor Handel, M.J., 2011. mHealth (mobile health)—using apps for health and wellness. Explor.
agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368, J. Sci. Heal. 7, 256–261.
1696–1705. Hariri, A.R., et al., 2005. A susceptibility gene for affective disorders and the response of
Egeland, M., Zunszain, P.A., Pariante, C.M., 2015. Molecular mechanisms in the regula- the human amygdala. Arch. Gen. Psychiatry 62, 146–152.
tion of adult neurogenesis during stress. Nat. Rev. Neurosci. 16, 189–200. Hayward, M.D., Schaich-Borg, A., Pintar, J.E., Low, M.J., 2006. Differential involvement
Bonin, R.P., et al., 2018. Systems, methods and apparatus for rodent behavioural mon- of endogenous opioids in sucrose consumption and food reinforcement. Pharmacol.
itoring. Google Patents,. Biochem. Behav. 85, 601–611.
Falconer, E., et al., 2008. The neural networks of inhibitory control in posttraumatic stress He, B., et al., 2018. Sugar-sweetened beverages consumption positively associated with
disorder. J. Psychiatry Neurosci. JPN 33, 413. the risks of obesity and hypertriglyceridemia among Children aged 7–18 years in
Feduccia, A., Simms, J., Mill, D., Yi, H., Bartlett, S., 2014. Varenicline decreases ethanol South China. J. Atheroscler. Thromb. 25, 81–89.
intake and increases dopamine release via neuronal nicotinic acetylcholine receptors Hebebrand, J., et al., 2014. “Eating addiction”, rather than “food addiction”, better
in the nucleus accumbens. Br. J. Pharmacol. 171, 3420–3431. captures addictive-like eating behavior. Neurosci. Biobehav. Rev. 47, 295–306.
Fidler, P., Kalman, B.A., Ziemer, H.E., Green, K.F., 1993. Early onset of reduced morphine Heim, C., Nemeroff, C.B., 2001. The role of childhood trauma in the neurobiology of
analgesia by ingestion of sweet solutions. Physiol. Behav. 53, 167–171. mood and anxiety disorders: preclinical and clinical studies. Biol. Psychiatry 49,
Fioravanti, M., et al., 2004. Emotional activation of obese and normal women due to 1023–1039.
imagery and food content of verbal stimuli in a memory task. Eat. Behav. 5, 47–54. Henry, C., Kabbaj, M., Simon, H., Le Moal, M., Maccari, S., 1994. Prenatal stress increases
Fiorentini, A., Müller, E., 1975. Sensitivity of central chemoreceptors controlling blood the hypothalamo‐pituitary‐adrenal axis response in young and adult rats. J.
glucose and body temperature during glucose deprivation. J. Physiol. 248, 247–271. Neuroendocrinol. 6, 341–345.
Flagel, S.B., et al., 2011. A selective role for dopamine in stimulus–reward learning. Herz, A., 1997. Endogenous opioid systems and alcohol addiction. Psychopharmacology
Nature 469, 53. 129, 99–111.
Floresco, S.B., Montes, D.R., Maric, M., van Holstein, M., 2018. Differential contributions Heyser, C.J., Schulteis, G., Koob, G.F., 1997. Increased ethanol self‐administration after a
of nucleus accumbens subregions to cue-guided risk/reward decision making and period of imposed ethanol deprivation in rats trained in a limited access paradigm.
implementation of conditional rules. J. Neurosci. 38, 1901–1914. Alcohol. Clin. Exp. Res. 21, 784–791.

195
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

Hirth, J.M., Rahman, M., Berenson, A.B., 2011. The association of posttraumatic stress alters the morphology of medium spiny neurons in the nucleus accumbens shell.
disorder with fast food and soda consumption and unhealthy weight loss behaviors Front. Behav. Neurosci. 10, 54.
among young women. J. Womens Health 20, 1141–1149. Knolle, F., et al., 2018. Altered cingulate and substantia nigra/ventral tegmental activa-
Hoebel, B.G., Avena, N.M., Rada, P., 2007. Accumbens dopamine-acetylcholine balance tion to novelty and emotional salience in antipsychotic naive first episode psychosis
in approach and avoidance. Curr. Opin. Pharmacol. 7, 617–627. patients. bioRxiv 263020.
Hoebel, B., Avena, N., Bocarsly, M., Rada, P., 2009. Natural addiction: a behavioral and Knüppel, A., Shipley, M.J., Llewellyn, C.H., Brunner, E.J., 2017. Sugar intake from sweet
circuit model based on sugar addiction in rats. J. Addict. Med. 3, 33–41. food and beverages, common mental disorder and depression: prospective findings
Holgate, J.Y., Shariff, M., Mu, E.W., Bartlett, S., 2017. A rat drinking in the dark model for from the Whitehall II study. Sci. Rep. 7, 6287.
studying ethanol and sucrose consumption. Front. Behav. Neurosci. 11, 29. Koob, G.F., 2009a. Brain stress systems in the amygdala and addiction. Brain Res. 1293,
Holstege, G., et al., 2003. Brain activation during human male ejaculation. J. Neurosci. 61–75.
23, 9185–9193. Koob, G.F., 2009b. Neurobiological substrates for the dark side of compulsivity in ad-
Hsu, T.M., et al., 2015. Effects of sucrose and high fructose corn syrup consumption on diction. Neuropharmacology 56, 18–31.
spatial memory function and hippocampal neuroinflammation in adolescent rats. Koob, G., 2018. 181. Drug addiction: the gain in the brain is in the pain. Biol. Psychiatry
Hippocampus 25, 227–239. 83, S72–S73.
Huang, S.-H., Wu, W.-R., Lee, L.-M., Huang, P.-R., Chen, J.-C., 2018. mTOR signaling in Koob, G.F., Le Moal, M., 2001. Drug addiction, dysregulation of reward, and allostasis.
the nucleus accumbens mediates behavioral sensitization to methamphetamine. Prog. Neuropsychopharmacology 24, 97.
Neuropsychopharmacol. Biol. Psychiatry. Koolschijn, P.C.M., van Haren, N.E., Lensvelt‐Mulders, G.J., Hulshoff Pol, H.E., Kahn,
Huber, R., et al., 2018. Drug-sensitive reward in crayfish: exploring the neural basis of R.S., 2009. Brain volume abnormalities in major depressive disorder: A meta‐analysis
addiction with automated learning paradigms. Behav. Processes. of magnetic resonance imaging studies. Hum. Brain Mapp. 30, 3719–3735.
Hwang, K.O., et al., 2010. Social support in an Internet weight loss community. Int. J. Kurhe, Y., Radhakrishnan, M., Gupta, D., 2014. Ondansetron attenuates depression co-
Med. Inform. 79, 5–13. morbid with obesity in obese mice subjected to chronic unpredictable mild stress; an
Hwang, J.J., et al., 2017. The human brain produces fructose from glucose. JCI Insight 2. approach using behavioral battery tests. Metab. Brain Dis. 29, 701–710.
Ibi, D., Suzuki, F., Hiramatsu, M., 2018. Effect of AceK (acesulfame potassium) on brain Kyriazis, G.A., Soundarapandian, M.M., Tyrberg, B., 2012. Sweet taste receptor signaling
function under dietary restriction in mice. Physiol. Behav. 188, 291–297. in beta cells mediates fructose-induced potentiation of glucose-stimulated insulin
Iemolo, A., et al., 2012. Withdrawal from chronic, intermittent access to a highly pala- secretion. Proc. Natl. Acad. Sci. 109, E524–E532.
table food induces depressive-like behavior in compulsive eating rats. Behav. Lam, K.C., Jadavji, N.M., 2018. Seeking Happiness: Understanding the Mechanisms of
Pharmacol. 23, 593. Mixing Music and Drugs.
Itzhak, Y., Martin, J.L., 1999. Effects of cocaine, nicotine, dizocipline and alcohol on mice Lathe, R., 2001. Hormones and the hippocampus. J. Endocrinol. 169, 205–231.
locomotor activity: cocaine–alcohol cross-sensitization involves upregulation of Lau, J., Herzog, H., 2014. CART in the regulation of appetite and energy homeostasis.
striatal dopamine transporter binding sites. Brain Res. 818, 204–211. Front. Neurosci. 8, 313.
Jacka, F.N., Mykletun, A., Berk, M., Bjelland, I., Tell, G.S., 2011. The association between Laughlin, M.R., 2014. Normal roles for dietary fructose in carbohydrate metabolism.
habitual diet quality and the common mental disorders in community-dwelling Nutrients 6, 3117–3129.
adults: the Hordaland Health study. Psychosom. Med. 73, 483–490. Leão, R.M., Cruz, F.C., Marin, M.T., da Silva Planeta, C., 2012. Stress induces behavioral
Jacobson, L., Sapolsky, R., 1991. The role of the hippocampus in feedback regulation of sensitization, increases nicotine-seeking behavior and leads to a decrease of CREB in
the hypothalamic-pituitary-adrenocortical axis. Endocr. Rev. 12, 118–134. the nucleus accumbens. Pharmacol. Biochem. Behav. 101, 434–442.
Jacques, A., et al., 2018. Techniques for mapping and measuring functional neuronal Lee, A.M., Messing, R.O., 2008. Protein kinases and addiction. Ann. N. Y. Acad. Sci. 1141,
topography for the decoding and prediction of behavior. Front. Neural Circuits 22–57.
12, 84. Legastelois, R., Botia, B., Coune, F., Jeanblanc, J., Naassila, M., 2014. Deciphering the
Jastreboff, A.M., et al., 2016. Altered brain response to drinking glucose and fructose in relationship between vulnerability to ethanol‐induced behavioral sensitization and
obese adolescents. Diabetes db151216. ethanol consumption in outbred mice. Addict. Biol. 19, 210–224.
Jellinek, E.M., 1952. Phases of alcohol addiction. Q. J. Stud. Alcohol 13, 673–684. Leigh, S.-J., Morris, M.J., 2016. The role of reward circuitry and food addiction in the
Jennings, J.H., Rizzi, G., Stamatakis, A.M., Ung, R.L., Stuber, G.D., 2013. The inhibitory obesity epidemic: an update. Biol. Psychol.
circuit architecture of the lateral hypothalamus orchestrates feeding. Science 341, Lemos, C., et al., 2016. High sucrose consumption induces memory impairment in rats
1517–1521. associated with electrophysiological modifications but not with metabolic changes in
Jennings, J.H., et al., 2015. Visualizing hypothalamic network dynamics for appetitive the hippocampus. Neuroscience 315, 196–205.
and consummatory behaviors. Cell 160, 516–527. Lenoir, M., Serre, F., Cantin, L., Ahmed, S.H., 2007. Intense sweetness surpasses cocaine
Jing, L., Liu, B., Zhang, M., Liang, J.-H., 2018. Involvement of dopamine D2 receptor in a reward. PLoS One 2, e698.
single methamphetamine-induced behavioral sensitization in C57BL/6J mice. Levine, A.S., Billington, C.J., 2004. Opioids as agents of reward-related feeding: a con-
Neurosci. Lett. sideration of the evidence. Physiol. Behav. 82, 57–61.
Johnson, P.M., Kenny, P.J., 2010. Dopamine D2 receptors in addiction-like reward dys- Levoy, E., Lazaridou, A., Brewer, J., Fulwiler, C., 2017. An exploratory study of
function and compulsive eating in obese rats. Nat. Neurosci. 13, 635. Mindfulness based Stress Reduction for emotional eating. Appetite 109, 124–130.
Joly-Amado, A., et al., 2014. The hypothalamic arcuate nucleus and the control of per- Li, Y., Acerbo, M.J., Robinson, T.E., 2004. The induction of behavioural sensitization is
ipheral substrates. Best Pract. Res. Clin. Endocrinol. Metab. 28, 725–737. associated with cocaine‐induced structural plasticity in the core (but not shell) of the
Jurdak, N., Kanarek, R.B., 2009. Sucrose-induced obesity impairs novel object recognition nucleus accumbens. Eur. J. Neurosci. 20, 1647–1654.
learning in young rats. Physiol. Behav. 96, 1–5. Lien, L., Lien, N., Heyerdahl, S., Thoresen, M., Bjertness, E., 2006. Consumption of soft
Kanarek, R.B., White, E.S., Biegen, M.T., Marks-Kaufman, R., 1991. Dietary influences on drinks and hyperactivity, mental distress, and conduct problems among adolescents
morphine-induced analgesia in rats. Pharmacol. Biochem. Behav. 38, 681–684. in Oslo, Norway. Am. J. Public Health 96, 1815–1820.
Kanoski, S.E., Davidson, T.L., 2011. Western diet consumption and cognitive impairment: Lin, Z., et al., 2013. Effects of curcumin on glucose metabolism in the brains of rats
links to hippocampal dysfunction and obesity. Physiol. Behav. 103, 59–68. subjected to chronic unpredictable stress: a 18 F-FDG micro-PET study. BMC
Karagiannides, I., et al., 2008. Substance P as a novel anti-obesity target. Complement. Altern. Med. 13, 202.
Gastroenterology 134, 747–755 e741. Lindgren, E., et al., 2018. Food addiction: a common neurobiological mechanism with
Kastin, A., 2013. Handbook of biologically active peptides. Academic press. drug abuse. Frontiers in bioscience (Landmark edition) 23, 811–836.
Kavaliers, M., Hirst, M., 1987. Slugs and snails and opiate tales: opioids and feeding Liu, S., 2018. What drives us? Chasing reward in a dopaminergic society. Columbia
behavior in invertebrates. Fed. Proc. 168–172. Undergrad. Sci. J. 11.
Keesman, M., Aarts, H., Häfner, M., Papies, E.K., 2017. Mindfulness reduces reactivity to Liu, C., Lee, S., Elmquist, J.K., 2014. Circuits controlling energy balance and mood: in-
food cues: underlying mechanisms and applications in daily life. Curr. Addict. Rep. 4, herently intertwined or just complicated intersections? Cell Metab. 19, 902–909.
151–157. Löf, E., Olausson, P., Stomberg, R., Taylor, J.R., Söderpalm, B., 2010. Nicotinic acet-
Kelley, A.E., Baldo, B.A., Pratt, W.E., 2005. A proposed hypothalamic–thalamic–striatal ylcholine receptors are required for the conditioned reinforcing properties of sucrose-
axis for the integration of energy balance, arousal, and food reward. J. Comp. Neurol. associated cues. Psychopharmacology 212, 321–328.
493, 72–85. Lowe, M.R., Butryn, M.L., 2007. Hedonic hunger: a new dimension of appetite? Physiol.
Kendig, M.D., 2014. Cognitive and behavioural effects of sugar consumption in rodents. A Behav. 91, 432–439.
review. Appetite 80, 41–54. Lu, L., Grimm, J.W., Hope, B.T., Shaham, Y., 2004. Incubation of cocaine craving after
Kenny, P.J., 2011. Reward mechanisms in obesity: new insights and future directions. withdrawal: a review of preclinical data. Neuropharmacology 47, 214–226.
Neuron 69, 664–679. Lumeng, J.C., et al., 2013. Overweight adolescents and life events in childhood. Pediatrics
Kikkawa, R., et al., 1987. Evidence for existence of polyol pathway in cultured rat me- 132, e1506–e1512.
sangial cells. Diabetes 36, 240–243. Luppino, F.S., et al., 2010. Overweight, obesity, and depression: a systematic review and
Kim, S., Shou, J., Abera, S., Ziff, E.B., 2018. Sucrose withdrawal induces depression and meta-analysis of longitudinal studies. Arch. Gen. Psychiatry 67, 220–229.
anxiety-like behavior by Kir2. 1 upregulation in the nucleus accumbens. Lustig, R.H., Schmidt, L.A., Brindis, C.D., 2012. Public health: the toxic truth about sugar.
Neuropharmacology 130, 10–17. Nature 482, 27.
Kinzig, K.P., Hargrave, S.L., Honors, M.A., 2008. Binge-type eating attenuates corticos- Majer-Lobodzinska, A., Adamiec-Mroczek, J., 2017. Glucocorticoid receptor poly-
terone and hypophagic responses to restraint stress. Physiol. Behav. 95, 108–113. morphism in obesity and glucose homeostasis. Adv. Clin. Exp. Med. 26, 143–148.
Kirshenbaum, A.P., Brown, S.J., Hughes, D.M., Doughty, A.H., 2008. Differential-re- Makarem, N., Bandera, E.V., Nicholson, J.M., Parekh, N., 2018. Consumption of sugars,
inforcement-of-low-rate-schedule performance and nicotine administration: a sys- sugary foods, and sugary beverages in relation to Cancer risk: a systematic review of
tematic investigation of dose, dose-regimen, and schedule requirement. Behav. longitudinal studies. Annu. Rev. Nutr.
Pharmacol. 19, 683. Maldonado, R., Valverde, O., Berrendero, F., 2006. Involvement of the endocannabinoid
Klenowski, P.M., et al., 2016. Prolonged consumption of sucrose in a binge-like manner, system in drug addiction. Trends Neurosci. 29, 225–232.

196
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

Malik, V.S., Pan, A., Willett, W.C., Hu, F.B., 2013. Sugar-sweetened beverages and weight Health Estimates.
gain in children and adults: a systematic review and meta-analysis. Am. J. Clin. Nutr. Organization, W.H, 2016. World Health Organization Obesity and Overweight Fact Sheet.
98, 1084–1102. Organization, W.H, 2017. Health Literacy. The Solid Facts. Health.
Mangabeira, V., Garcia-Mijares, M., Silva, M.T.A., 2015. Sugar withdrawal and differ- Oswald, K.D., Murdaugh, D.L., King, V.L., Boggiano, M.M., 2011. Motivation for palatable
ential reinforcement of low rate (DRL) performance in rats. Physiol. Behav. 139, food despite consequences in an animal model of binge eating. Int. J. Eat. Disord. 44,
468–473. 203–211.
Maniam, J., Antoniadis, C.P., Morris, M.J., 2015. The effect of early-life stress and chronic Otmakhova, N., Duzel, E., Deutch, A.Y., Lisman, J., 2013. The hippocampal-VTA loop: the
high-sucrose diet on metabolic outcomes in female rats. Stress 18, 524–537. role of novelty and motivation in controlling the entry of information into long-term
Mantyh, P.W., 2002. Neurobiology of substance P and the NK1 receptor. J. Clin. memory. Intrinsically Motivated Learning in Natural and Artificial Systems. Springer,
Psychiatry 63, 6–10. pp. 235–254.
Maren, S., 2011. Seeking a spotless mind: extinction, deconsolidation, and erasure of fear Padilla, S.L., et al., 2016. Agouti-related peptide neural circuits mediate adaptive beha-
memory. Neuron 70, 830–845. viors in the starved state. Nat. Neurosci. 19, 734.
Mark, G.P., et al., 2006. Injection of oxotremorine in nucleus accumbens shell reduces Padwal, R.S., Majumdar, S.R., 2007. Drug treatments for obesity: orlistat, sibutramine,
cocaine but not food self-administration in rats. Brain Res. 1123, 51–59. and rimonabant. Lancet 369, 71–77.
Martin, E.I., Ressler, K.J., Binder, E., Nemeroff, C.B., 2009. The neurobiology of anxiety Page, K.A., et al., 2013. Effects of fructose vs glucose on regional cerebral blood flow in
disorders: brain imaging, genetics, and psychoneuroendocrinology. Psychiatric brain regions involved with appetite and reward pathways. Jama 309, 63–70.
Clinics 32, 549–575. Palmiter, R.D., 2007. Is dopamine a physiologically relevant mediator of feeding beha-
Mathes, C.M., Gregson, J.R., Spector, A.C., 2013. The selective serotonin reuptake in- vior? Trends Neurosci. 30, 375–381.
hibitor paroxetine decreases breakpoint of rats engaging in a progressive ratio licking Pan, X., Zhang, C., Shi, Z., 2011. Soft drink and sweet food consumption and suicidal
task for sucrose and quinine solutions. Chem. Senses 38, 211–220. behaviours among Chinese adolescents. Acta Paediatr. 100, e215–e222.
Meye, F.J., Adan, R.A., 2014. Feelings about food: the ventral tegmental area in food Pandit, R., et al., 2015. Central melanocortins regulate the motivation for sucrose reward.
reward and emotional eating. Trends Pharmacol. Sci. 35, 31–40. PLoS One 10, e0121768.
Milad, M.R., Quirk, G.J., 2012. Fear extinction as a model for translational neuroscience: Parylak, S.L., Cottone, P., Sabino, V., Rice, K.C., Zorrilla, E.P., 2012. Effects of CB1 and
ten years of progress. Annu. Rev. Psychol. 63, 129–151. CRF1 receptor antagonists on binge-like eating in rats with limited access to a sweet
Miller, C.C., Martin, R.J., Whitney, M.L., Edwards, G.L., 2002. Intracerebroventricular fat diet: lack of withdrawal-like responses. Physiol. Behav. 107, 231–242.
injection of fructose stimulates feeding in rats. Nutr. Neurosci. 5, 359–362. Patkar, O.L., et al., 2017. The antihypertensive drug pindolol attenuates long‐term but not
Ming, G.-l., Song, H., 2011. Adult neurogenesis in the mammalian brain: significant an- short‐term binge‐like ethanol consumption in mice. Addict. Biol. 22, 679–691.
swers and significant questions. Neuron 70, 687–702. Patriquin, M.A., Mathew, S.J., 2017. The neurobiological mechanisms of generalized
Misra, M., et al., 2008. Lower growth hormone and higher cortisol are associated with anxiety disorder and chronic stress. Chronic Stress 1 2470547017703993.
greater visceral adiposity, intramyocellular lipids, and insulin resistance in over- Pecina, S., Berridge, K., 1995. Central enhancement of taste pleasure by intraventricular
weight girls. Am. J. Physiol. Endocrinol. Metab. 295, E385–E392. morphine. Neurobiology (Budapest, Hungary) 3, 269–280.
Mitra, A., Gosnell, B.A., Schiöth, H.B., Grace, M.K., Klockars, A., Olszewski, P.K., Levine, Peet, M., 2004. International variations in the outcome of schizophrenia and the pre-
A.S., 2010. Chronic sugar intake dampens feeding-related activity of neurons syn- valence of depression in relation to national dietary practices: an ecological analysis.
thesizing a satiety mediator, oxytocin. Peptides 31, 1346–1352. Br. J. Psychiatry 184, 404–408.
Moghaddam, B., 2018. 27.3 discrete and coordinated encoding of rewarded actions by Peng, X.X., Ziff, E.B., Carr, K.D., 2011. Effects of food restriction and sucrose intake on
prefrontal cortex and dopamine neurons. Schizophr. Bull. 44, S45. synaptic delivery of AMPA receptors in nucleus accumbens. Synapse 65, 1024–1031.
Molnar, S.M., et al., 2018. Behavioral and brain activity indices of cognitive control Petrovich, G.D., Gallagher, M., 2003. Amygdala subsystems and control of feeding be-
deficits in binge drinkers. Brain Sci. 8, 9. havior by learned cues. Ann. N. Y. Acad. Sci. 985, 251–262.
Molteni, R., Barnard, R., Ying, Z., Roberts, C., Gomez-Pinilla, F., 2002. A high-fat, refined Petrovich, G.D., Hobin, M., Reppucci, C., 2012. Selective Fos induction in hypothalamic
sugar diet reduces hippocampal brain-derived neurotrophic factor, neuronal plasti- orexin/hypocretin, but not melanin-concentrating hormone neurons, by a learned
city, and learning. Neuroscience 112, 803–814. food-cue that stimulates feeding in sated rats. Neuroscience 224, 70–80.
Moor, K.R., Scott, A.J., McIntosh, W.D., 2013. Mindful eating and its relationship to body Pierce, R.C., Kalivas, P.W., 1995. Amphetamine produces sensitized increases in loco-
mass index and physical activity among university students. Mindfulness 4, 269–274. motion and extracellular dopamine preferentially in the nucleus accumbens shell of
Moore, C.F., Sabino, V., Cottone, P., 2018. Trace amine associated receptor 1 (TAAR1) rats administered repeated cocaine. J. Pharmacol. Exp. Ther. 275, 1019–1029.
modulation of food reward. Front. Pharmacol. 9, 129. Pijl, H., 2003. Reduced dopaminergic tone in hypothalamic neural circuits: expression of
Moorman, D.E., Aston-Jones, G., 2014. Orbitofrontal cortical neurons encode expecta- a “thrifty” genotype underlying the metabolic syndrome? Eur. J. Pharmacol. 480,
tion-driven initiation of reward-seeking. J. Neurosci. 34, 10234–10246. 125–131.
Morey, R.A., Petty, C.M., Cooper, D.A., LaBar, K.S., McCarthy, G., 2008. Neural systems Pine, D.S., Cohen, P., Johnson, J.G., Brook, J.S., 2002. Adolescent life events as predictors
for executive and emotional processing are modulated by symptoms of posttraumatic of adult depression. J. Affect. Disord. 68, 49–57.
stress disorder in Iraq War veterans. Psychiatry Res. Neuroimaging 162, 59–72. Posadas, I., López-Hernández, B., Ceña, V., 2013. Nicotinic receptors in neurodegenera-
Moscarello, J.M., Maren, S., 2018. Flexibility in the face of fear: hippocampal–prefrontal tion. Curr. Neuropharmacol. 11, 298–314.
regulation of fear and avoidance. Curr. Opin. Behav. Sci. 19, 44–49. Powers, S.K., Howley, E.T., 2007. Exercise Physiology: Theory and Application to Fitness
Moss, M., 2013. Salt, Sugar, Fat: How the Food Giants Hooked Us. Random House. and Performance.
Murray, S., Tulloch, A., Criscitelli, K., Avena, N.M., 2016. Recent studies of the effects of Pratt, W.E., Kelley, A.E., 2004. Nucleus accumbens acetylcholine regulates appetitive
sugars on brain systems involved in energy balance and reward: relevance to low learning and motivation for food via activation of muscarinic receptors. Behav.
calorie sweeteners. Physiol. Behav. 164, 504–508. Neurosci. 118, 730.
Muscat, R., Willner, P., 1989. Effects of dopamine receptor antagonists on sucrose con- Price, J.L., et al., 2001. Neuron number in the entorhinal cortex and CA1 in preclinical
sumption and preference. Psychopharmacology 99, 98–102. Alzheimer disease. Arch. Neurol. 58, 1395–1402.
Nakagawa, T., et al., 2006. A causal role for uric acid in fructose-induced metabolic Pursey, K.M., et al., 2014. Neural responses to visual food cues according to weight status:
syndrome. Am. J. Physiol. Renal Physiol. 290, F625–F631. a systematic review of functional magnetic resonance imaging studies. Front. Nutr. 1.
Nakamura, K., Ono, T., Tamura, R., 1987. Central sites involved in lateral hypothalamus Qualls-Creekmore, E., et al., 2017. Galanin-expressing GABA neurons in the lateral hy-
conditioned neural responses to acoustic cues in the rat. J. Neurophysiol. 58, pothalamus modulate food reward and noncompulsive locomotion. J. Neurosci. 37,
1123–1148. 6053–6065.
Naneix, F., Darlot, F., Coutureau, E., Cador, M., 2016. Long‐lasting deficits in hedonic and Quiñones, M., et al., 2018. p53 in AgRP neurons is required for protection against diet-
nucleus accumbens reactivity to sweet rewards by sugar overconsumption during induced obesity via JNK1. Nat. Commun. 9, 3432.
adolescence. Eur. J. Neurosci. 43, 671–680. Quirk, S.E., et al., 2013. The association between diet quality, dietary patterns and de-
Naneix, F., et al., 2018. Protracted motivational dopamine-related deficits following pression in adults: a systematic review. BMC Psychiatry 13, 175.
adolescence sugar overconsumption. Neuropharmacology 129, 16–25. Rada, P., Hoebel, B.G., 2005. Acetylcholine in the accumbens is decreased by diazepam
Nestler, E.J., Aghajanian, G.K., 1997. Molecular and cellular basis of addiction. Science and increased by benzodiazepine withdrawal: a possible mechanism for dependency.
278, 58–63. Eur. J. Pharmacol. 508, 131–138.
Nieh, E.H., et al., 2015. Decoding neural circuits that control compulsive sucrose seeking. Rada, P., Jensen, K., Hoebel, B.G., 2001. Effects of nicotine and mecamylamine-induced
Cell 160, 528–541. withdrawal on extracellular dopamine and acetylcholine in the rat nucleus ac-
Noble, E.E., Hsu, T.M., Liang, J., Kanoski, S.E., 2017. Early-life sugar consumption has cumbens. Psychopharmacology 157, 105–110.
long-term negative effects on memory function in male rats. Nutr. Neurosci. 1–11. Rada, P., Avena, N., Hoebel, B., 2005. Daily bingeing on sugar repeatedly releases do-
Noll, J.G., Zeller, M.H., Trickett, P.K., Putnam, F.W., 2007. Obesity risk for female victims pamine in the accumbens shell. Neuroscience 134, 737–744.
of childhood sexual abuse: a prospective study. Pediatrics 120, e61–e67. Rahman, S., Engleman, E.A., Bell, R.L., 2015. Nicotinic receptor modulation to treat al-
Nona, C.N., Nobrega, J.N., 2018. A role for nucleus accumbens glutamate in the ex- cohol and drug dependence. Front. Neurosci. 8, 426.
pression but not the induction of behavioural sensitization to ethanol. Behav. Brain Rajmohan, V., Mohandas, E., 2007. The limbic system. Indian J. Psychiatry 49, 132.
Res. 336, 269–281. Ramasubramanian, L., Lane, S., Rahman, A., 2013. The association between maternal
Novelle, M.G., Diéguez, C., 2018. Food addiction and binge eating: lessons learned from serious psychological distress and child obesity at 3 years: a cross‐sectional analysis
animal models. Nutrients 10, 71. of the UK Millennium Cohort Data. Child Care Health Dev. 39, 134–140.
Nummenmaa, L., Tuominen, L., 2017. Opioid system and human emotions. Br. J. Reichelt, A.C., Maniam, J., Westbrook, R.F., Morris, M.J., 2015. Dietary-induced obesity
Pharmacol. disrupts trace fear conditioning and decreases hippocampal reelin expression. Brain
Olds, M.E., Olds, J., 1963. Approach‐avoidance analysis of rat diencephalon. J. Comp. Behav. Immun. 43, 68–75.
Neurol. 120, 259–295. Reker, A.N., et al., 2018. Neurogranin in the nucleus accumbens regulates NMDA receptor
Organization, W.H., 2017. Depression and Other Common Mental Disorders: Global tolerance and motivation for ethanol seeking. Neuropharmacology 131, 58–67.

197
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

Rena, G., Hardie, D.G., Pearson, E.R., 2017. The mechanisms of action of metformin. related central pathways in control or high-fat fed mice. Physiol. Behav. 44–53.
Diabetologia 60, 1577–1585. Southgate, D., 1995. Digestion and metabolism of sugars. Am. J. Clin. Nutr. 62,
Reyes, S., Peirano, P., Peigneux, P., Lozoff, B., Algarin, C., 2015. Inhibitory control in 203S–210S.
otherwise healthy overweight 10-year-old children. Int. J. Obes. 39 (1230). Souza, C., et al., 2007. Highly palatable diet consumption increases protein oxidation in
Richard, D., Ferland, J., Lalonde, J., Samson, P., Deshaies, Y., 2000. Influence of topir- rat frontal cortex and anxiety-like behavior. Life Sci. 81, 198–203.
amate in the regulation of energy balance. Nutrition 16, 961–966. Spangler, R., et al., 2004. Opiate-like effects of sugar on gene expression in reward areas
Riga, D., Theijs, J.T., De Vries, T.J., Smit, A.B., Spijker, S., 2015. Social defeat-induced of the rat brain. Mol. Brain Res. 124, 134–142.
anhedonia: effects on operant sucrose-seeking behavior. Front. Behav. Neurosci. 9, Staley, J.K., Mash, D.C., 1996. Adaptive increase in D3 dopamine receptors in the brain
195. reward circuits of human cocaine fatalities. J. Neurosci. 16, 6100–6106.
Ritter, R.C., Slusser, P.G., Stone, S., 1981. Glucoreceptors controlling feeding and blood Stanley, B.G., Lanthier, D., Leibowitz, S.F., 1988. Multiple brain sites sensitive to feeding
glucose: location in the hindbrain. Science 213, 451–452. stimulation by opioid agonists: a cannula-mapping study. Pharmacol. Biochem.
Robinson, T.E., 1988. Stimulant drugs and stress: factors influencing individual differ- Behav. 31, 825–832.
ences in the susceptibility to sensitization. Sensitization of the Nervous System. pp. Steensland, P., et al., 2010. The neurokinin 1 receptor antagonist, ezlopitant, reduces
145–173. appetitive responding for sucrose and ethanol. PLoS One 5, e12527.
Robinson, T.E., Berridge, K.C., 1993. The neural basis of drug craving: an incentive- Stranahan, A.M., et al., 2008. Diet‐induced insulin resistance impairs hippocampal sy-
sensitization theory of addiction. Brain Res. Rev. 18, 247–291. naptic plasticity and cognition in middle‐aged rats. Hippocampus 18, 1085–1088.
Robinson, T.E., Berridge, K.C., 2001. Incentive‐sensitization and addiction. Addiction 96, Stroebe, W., Papies, E.K., Aarts, H., 2008. From homeostatic to hedonic theories of eating:
103–114. Self‐regulatory failure in food‐rich environments. Appl. Psychol. 57, 172–193.
Robinson, T.E., Berridge, K.C., 2008. The incentive sensitization theory of addiction: some Stunkard, A.J., Faith, M.S., Allison, K.C., 2003. Depression and obesity. Biol. Psychiatry
current issues. Philos. Trans. R. Soc. B: Biol. Sci. 363, 3137–3146. 54, 330–337.
Robinson, M.J., Warlow, S.M., Berridge, K.C., 2014. Optogenetic excitation of central Sweeney, P., Yang, Y., 2017. Neural circuit mechanisms underlying emotional regulation
amygdala amplifies and narrows incentive motivation to pursue one reward above of homeostatic feeding. Trends Endocrinol. Metab. 28, 437–448.
another. J. Neurosci. 34, 16567–16580. Tabuchi, E., et al., 2002. Spatio–temporal dynamics of brain activated regions during
Rosas, M., et al., 2018. Effects of morphine on place conditioning and ERK 1/2 phos- drinking behavior in rats. Brain Res. 951, 270–279.
phorylation in the nucleus accumbens of psychogenetically selected Roman low-and Tahara, A., Takasu, T., Yokono, M., Imamura, M., Kurosaki, E., 2018. Antidiabetic and
high-avoidance rats. Psychopharmacology 235, 59–69. antiobesity effects of SGLT2 inhibitor ipragliflozin in type 2 diabetic mice fed sugar
Ruegg, H., Yu, W.-Z., Bodnar, R.J., 1997. Opioid-receptor subtype agonist-induced en- solution. Eur. J. Pharmacol. 818, 545–553.
hancements of sucrose intake are dependent upon sucrose concentration. Physiol. Teff, K.L., et al., 2004. Dietary fructose reduces circulating insulin and leptin, attenuates
Behav. 62, 121–128. postprandial suppression of ghrelin, and increases triglycerides in women. J. Clin.
Salamone, J.D., Correa, M., 2012. The mysterious motivational functions of mesolimbic Endocrinol. Metab. 89, 2963–2972.
dopamine. Neuron 76, 470–485. Tellez, L.A., et al., 2016. Separate circuitries encode the hedonic and nutritional values of
Sama, P.R., Eapen, Z.J., Weinfurt, K.P., Shah, B.R., Schulman, K.A., 2014. An evaluation sugar. Nat. Neurosci. 19, 465.
of mobile health application tools. JMIR mhealth uhealth 2. Tryon, M.S., et al., 2015. Excessive sugar consumption may be a difficult habit to break: a
Sánchez-Villegas, A., et al., 2012. Fast-food and commercial baked goods consumption view from the brain and body. J. Clin. Endocrinol. Metab. 100, 2239–2247.
and the risk of depression. Public Health Nutr. 15, 424–432. Tye, K.M., Stuber, G.D., De Ridder, B., Bonci, A., Janak, P.H., 2008. Rapid strengthening
Sandweiss, A.J., Vanderah, T.W., 2015. The pharmacology of neurokinin receptors in of thalamo-amygdala synapses mediates cue–reward learning. Nature 453 (1253).
addiction: prospects for therapy. Subst. Abuse Rehabil. 6, 93. Ulrich, M., Stauß, P., Grön, G., 2018. Glucose modulates human ventral tegmental ac-
Sangüesa, G., et al., 2018. Impairment of novel object recognition memory and brain tivity in response to sexual stimuli. J. Sex. Med. 15, 20–28.
insulin signaling in fructose-but not glucose-drinking female rats. Mol. Neurobiol. Ulrich-Lai, Y.M., et al., 2010. Pleasurable behaviors reduce stress via brain reward
1–16. pathways. Proc. Natl. Acad. Sci. 107, 20529–20534.
Santos, C.J., et al., 2018. Carbohydrate-enriched diet predispose to anxiety and depres- Ulrich-Lai, Y.M., Ostrander, M.M., Herman, J.P., 2011. HPA axis dampening by limited
sion-like behavior after stress in mice. Nutr. Neurosci. 21, 33–39. sucrose intake: reward frequency vs. Caloric consumption. Physiol. Behav. 103,
Savage, S.W., et al., 2014. Regulation of novelty seeking by midbrain dopamine D2/D3 104–110.
signaling and ghrelin is altered in obesity. Obesity 22, 1452–1457. Ursano, R.J., et al., 2009. PTSD and traumatic stress: from gene to community and bench
Schneider, L.H., 1989. Orosensory Self‐Stimulation by Sucrose Involves Brain to bedside. Brain Res. 1293, 2–12.
Dopaminergic Mechanisms a. Ann. N. Y. Acad. Sci. 575, 307–320. Valencia-Torres, L., et al., 2017. Activation of ventral tegmental area 5-HT 2C receptors
Scholzen, T., Gerdes, J., 2000. The Ki‐67 protein: from the known and the unknown. J. reduces incentive motivation. Neuropsychopharmacology 42 (1511).
Cell. Physiol. 182, 311–322. Van der Borght, K., et al., 2011. Reduced neurogenesis in the rat hippocampus following
Schwartzbaum, J., 1988. Electrophysiology of taste, feeding and reward in lateral hy- high fructose consumption. Regul. Pept. 167, 26–30.
pothalamus of rabbit. Physiol. Behav. 44, 507–526. van Dooren, F.E., et al., 2013. Depression and risk of mortality in people with diabetes
See, R.E., Fuchs, R.A., Ledford, C.C., McLAUGHLIN, J., 2003. Drug addiction, relapse, and mellitus: a systematic review and meta-analysis. PLoS One 8, e57058.
the amygdala. Ann. N. Y. Acad. Sci. 985, 294–307. Van Strien, T., Frijters, J.E., Bergers, G.P., Defares, P.B., 1986. The Dutch Eating Behavior
Shariff, M., et al., 2016. Neuronal nicotinic acetylcholine receptor modulators reduce Questionnaire (DEBQ) for assessment of restrained, emotional, and external eating
sugar intake. PLoS One 11, e0150270. behavior. Int. J. Eat. Disord. 5, 295–315.
Shariff, M., et al., 2017. Binge-like sucrose consumption reduces the dendritic length and Vendruscolo, L.F., Gueye, A.B., Darnaudéry, M., Ahmed, S.H., Cador, M., 2010. Sugar
complexity of principal neurons in the adolescent rat basolateral amygdala. PLoS One overconsumption during adolescence selectively alters motivation and reward func-
12, e0183063. tion in adult rats. PLoS One 5, e9296.
Sharpe, M., Clemens, K., Morris, M., Westbrook, R., 2016. Daily exposure to sucrose Verty, A.N., McGregor, I.S., Mallet, P.E., 2004. The dopamine receptor antagonist SCH
impairs subsequent learning about food cues: a role for alterations in ghrelin sig- 23390 attenuates feeding induced by Δ9-tetrahydrocannabinol. Brain Res. 1020,
naling and dopamine D2 receptors. Neuropsychopharmacology 41 (1357). 188–195.
Shearrer, G., et al., 2016. Associations among sugar sweetened beverage intake, visceral Vigano, D., et al., 2003. Μ opioid receptor signaling in morphine sensitization.
fat, and cortisol awakening response in minority youth. Physiol. Behav. 167, Neuroscience 117, 921–929.
188–193. Volkow, N.D., Fowler, J.S., 2000. Addiction, a disease of compulsion and drive: in-
Shearrer, G.E., Stice, E., Burger, K.S., 2018a. Adolescents at high risk of obesity show volvement of the orbitofrontal cortex. Cereb. Cortex 10, 318–325.
greater striatal response to increased sugar content in milkshakes. Am. J. Clin. Nutr Volkow, N.D., Wise, R.A., 2005. How can drug addiction help us understand obesity? Nat.
nqy050-nqy050. Neurosci. 8, 555.
Shearrer, G.E., Stice, E., Burger, K.S., 2018b. Adolescents at high risk of obesity show Volkow, N.D., Fowler, J., Wang, G.-J., 2002. Role of dopamine in drug reinforcement and
greater striatal response to increased sugar content in milkshakes. Am. J. Clin. Nutr. addiction in humans: results from imaging studies. Behav. Pharmacol. 13, 355–366.
107, 859–866. Volkow, N.D., Fowler, J.S., Wang, G.-J., Swanson, J.M., 2004. Dopamine in drug abuse
Shi, Z., Taylor, A.W., Wittert, G., Goldney, R., Gill, T.K., 2010. Soft drink consumption and addiction: results from imaging studies and treatment implications. Mol.
and mental health problems among adults in Australia. Public Health Nutr. 13, Psychiatry 9, 557.
1073–1079. Vollmayr, B., et al., 2004. Rats with congenital learned helplessness respond less to su-
Simon, G.E., et al., 2006. Association between obesity and psychiatric disorders in the US crose but show no deficits in activity or learning. Behav. Brain Res. 150, 217–221.
adult population. Arch. Gen. Psychiatry 63, 824–830. Wade, T.R., de Wit, H., Richards, J.B., 2000. Effects of dopaminergic drugs on delayed
Sinha, R., 2008. Chronic stress, drug use, and vulnerability to addiction. Ann. N. Y. Acad. reward as a measure of impulsive behavior in rats. Psychopharmacology 150,
Sci. 1141, 105–130. 90–101.
Sinha, R., 2018. Role of addiction and stress neurobiology on food intake and obesity. Wallace, D.L., et al., 2008. The influence of ΔFosB in the nucleus accumbens on natural
Biol. Psychol. 131, 5–13. reward-related behavior. J. Neurosci. 28, 10272–10277.
Smith, I., 1960. Carbohydrates. Chromatographic and Electrophoretic Techniques 1. pp. Wang, G.-J., et al., 2001. Brain dopamine and obesity. Lancet 357, 354–357.
246–260. Wang, G.-J., et al., 2006. Gastric stimulation in obese subjects activates the hippocampus
Smith, S.M., Vale, W.W., 2006. The role of the hypothalamic-pituitary-adrenal axis in and other regions involved in brain reward circuitry. Proc. Natl. Acad. Sci. 103,
neuroendocrine responses to stress. Dialogues Clin. Neurosci. 8 (383). 15641–15645.
Sohn, J.-W., Elmquist, J.K., Williams, K.W., 2013. Neuronal circuits that regulate feeding Weeks, J.R., 1962. Experimental morphine addiction: method for automatic intravenous
behavior and metabolism. Trends Neurosci. 36, 504–512. injections in unrestrained rats. Science 138, 143–144.
Soto, M., Chaumontet, C., Even, P.C., Nadkarni, N., Piedcoq, J., Darcel, N., et al., 2015. Wehry, A.M., Beesdo-Baum, K., Hennelly, M.M., Connolly, S.D., Strawn, J.R., 2015.
Intermittent access to liquid sucrose differentially modulates energy intake and Assessment and treatment of anxiety disorders in children and adolescents. Curr.

198
A. Jacques, et al. Neuroscience and Biobehavioral Reviews 103 (2019) 178–199

Psychiatry Rep. 17, 52. Woods, J.S., Leibowitz, S.F., 1985. Hypothalamic sites sensitive to morphine and na-
Weihrauch, M., Diehl, V., 2004. Artificial sweeteners—do they bear a carcinogenic risk? loxone: effects on feeding behavior. Pharmacol. Biochem. Behav. 23, 431–438.
Ann. Oncol. 15, 1460–1465. Wu, J., 2009. Understanding of nicotinic acetylcholine receptors. Acta Pharmacol. Sin.
Weissman, M.M., et al., 1999. Children with prepubertal-onset major depressive disorder 30, 653.
and anxiety grown up. Arch. Gen. Psychiatry 56, 794–801. Wyvell, C.L., Berridge, K.C., 2001. Incentive sensitization by previous amphetamine ex-
Welberg, L.A., Seckl, J.R., 2001. Prenatal stress, glucocorticoids and the programming of posure: increased cue-triggered “wanting” for sucrose reward. J. Neurosci. 21,
the brain. J. Neuroendocrinol. 13, 113–128. 7831–7840.
Westover, A.N., Marangell, L.B., 2002. A cross‐national relationship between sugar con- Xiong, L., et al., 2018. CART peptide in the nucleus accumbens shell inhibits cocai-
sumption and major depression? Depress. Anxiety 16, 118–120. ne‐induced locomotor sensitization to transient overexpression of α‐Ca2+/
Westwater, M.L., Fletcher, P.C., Ziauddeen, H., 2016. Sugar addiction: the state of the Calmodulin‐dependent Protein Kinase II. J. Neurochem.
science. Eur. J. Nutr. 55, 55–69. Xu, T.J., Reichelt, A.C., 2018. Sucrose or sucrose and caffeine differentially impact
Williams-Spooner, M., Richardson, R., Baker, K., 2017. 835. A high-fat high-sugar diet- memory and anxiety-like behaviours, and alter hippocampal parvalbumin and dou-
induced impairment of fear inhibition and place-recognition memory in adolescent blecortin. Neuropharmacology 137, 24–32.
rats. Biol. Psychiatry 81, S338–S339. Yamanaka, A., et al., 2000. Orexin-induced food intake involves neuropeptide Y pathway.
Wilmouth, C.E., Spear, L.P., 2009. Hedonic sensitivity in adolescent and adult rats: taste Brain Res. 859, 404–409.
reactivity and voluntary sucrose consumption. Pharmacol. Biochem. Behav. 92, Yang, H., et al., 2018. Nucleus accumbens subnuclei regulate motivated behavior via
566–573. direct inhibition and disinhibition of VTA dopamine subpopulations. Neuron.
Wilson, G.T., 2010. Eating disorders, obesity and addiction. Eur. Eat. Disord. Rev. 18, Yeomans, M.R., 2017. Adverse effects of consuming high fat–sugar diets on cognition:
341–351. implications for understanding obesity. Proc. Nutr. Soc. 76, 455–465.
Wise, R.A., 1996. Addictive drugs and brain stimulation reward. Annu. Rev. Neurosci. 19, Yu, J.H., Kim, M.-S., 2012. Molecular mechanisms of appetite regulation. Diabetes Metab.
319–340. J. 36, 391–398.
Wise, R.A., 2004. Dopamine, learning and motivation. Nat. Rev. Neurosci. 5, 483. Zehra, A., et al., 2018. Cannabis addiction and the brain: a review. J. Neuroimmune
Wise, R.A., 2006. Role of brain dopamine in food reward and reinforcement. Philos. Pharmacol. 1-15.
Trans. R. Soc. Lond., B, Biol. Sci. 361, 1149–1158. Zhang, C., et al., 2018a. Cholinergic tone in ventral tegmental area: functional organi-
Wise, R.A., et al., 1995. Fluctuations in nucleus accumbens dopamine concentration zation and behavioral implications. Neurochem. Int.
during intravenous cocaine self-administration in rats. Psychopharmacology 120, Zhang, S., et al., 2018b. Sex differences in the neuroadaptations of reward-related circuits
10–20. in response to subchronic variable stress. Neuroscience 376, 108–116.
Wolf, A., Bray, G., Popkin, B., 2008. A short history of beverages and how our body treats Ziauddeen, H., Farooqi, I.S., Fletcher, P.C., 2012. Obesity and the brain: how convincing
them. Obes. Rev. 9, 151–164. is the addiction model? Nat. Rev. Neurosci. 13, 279.

199

You might also like