You are on page 1of 10

G Model

NBR-2089; No. of Pages 10 ARTICLE IN PRESS


Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Neuroscience and Biobehavioral Reviews


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

Review

Why is obesity such a problem in the 21st century? The intersection


of palatable food, cues and reward pathways, stress, and cognition
Margaret J. Morris a,∗ , Jessica E. Beilharz a , Jayanthi Maniam a ,
Amy C. Reichelt a,b , R. Frederick Westbrook b
a
School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia
b
School of Psychology, UNSW Australia, Sydney, NSW 2052, Australia

a r t i c l e i n f o a b s t r a c t

Article history: Changes in food composition and availability have contributed to the dramatic increase in obesity over
Received 1 July 2014 the past 30–40 years in developed and, increasingly, in developing countries. The brain plays a critical
Received in revised form role in regulating energy balance. Some human studies have demonstrated increased preference for high
15 November 2014
fat and high sugar foods in people reporting greater stress exposure. We have examined neurochemical
Accepted 2 December 2014
changes in the brain in rodent models during the development of obesity, including the impact of obe-
Available online xxx
sity on cognition, reward neurocircuitry and stress responsiveness. Using supermarket foods high in fat
and sugar, we showed that such a diet leads to changes in neurotransmitters involved in the hedonic
Keywords:
Obesity
appraisal of foods, indicative of an addiction-like capacity of foods high in fat and/or sugar. Importantly,
Stress withdrawal of the palatable diet led to a stress-like response. Furthermore, access to this palatable diet
Overeating attenuated the physiological effects of acute stress (restraint), indicating that it could act as a comfort
Hippocampus food. In more chronic studies, the diet also attenuated anxiety-like behavior in rats exposed to stress
Reward (maternal separation) early in life, but these rats may suffer greater metabolic harm than rats exposed
Dopamine to the early life stressor but not provided with the palatable diet.
Memory Impairments in cognitive function have been associated with obesity in both people and rodents.
However, as little as 1 week of exposure to a high fat, high sugar diet selectively impaired place but
not object recognition memory in the rat. Excess sugar alone had similar effects, and both diets were
linked to increased inflammatory markers in the hippocampus, a critical region involved in memory.
Obesity-related inflammatory changes have been found in the human brain. Ongoing work examines
interventions to prevent or reverse diet-induced cognitive impairments. These data have implications
for minimizing harm caused by unhealthy eating.
© 2014 Elsevier Ltd. All rights reserved.

Contents

1. Introduction – what are the drivers of obesity? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00


2. Effect of obesogenic diets on reward processing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.1. Intermittent access models of binge-like behavior . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.2. Incentive learning processes that may underpin overconsumption . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
2.3. Cue driven mechanisms of food intake . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3. Stress and eating behavior . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.1. Early life stress and other influences on eating behavior/obesity risk . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
3.2. Rodent model of early life stress–maternal separation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4. Effects of high energy diets on cognition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.1. Evidence for effects of high energy diets on human cognition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
4.2. Effects of high energy diets on learning and memory in rodents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

∗ Corresponding author. Tel.: +61 2 9385 1560; fax: +61 2 9385 0023.
E-mail address: m.morris@unsw.edu.au (M.J. Morris).

http://dx.doi.org/10.1016/j.neubiorev.2014.12.002
0149-7634/© 2014 Elsevier Ltd. All rights reserved.

Please cite this article in press as: Morris, M.J., et al., Why is obesity such a problem in the 21st century? The intersection of palatable food,
cues and reward pathways, stress, and cognition. Neurosci. Biobehav. Rev. (2014), http://dx.doi.org/10.1016/j.neubiorev.2014.12.002
G Model
NBR-2089; No. of Pages 10 ARTICLE IN PRESS
2 M.J. Morris et al. / Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx

5. Summary and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00


Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

1. Introduction – what are the drivers of obesity? than nutrient requirements, and driven by the availability of such
foods (Lutter and Nestler, 2009; Meye and Adan, 2014; Saper et al.,
Obesity is a major health issue in the developed and, increas- 2002). The mesocortical dopamine system plays an important role
ingly, in the developing world; indeed obesity is considered the in reward-related processes. This system extends from the ventral
most serious health issue facing the developed world, formally rec- tegmental area (VTA) via the nucleus accumbens (NAc) to corti-
ognized as such by the World Health Organization (WHO, 2000). cal regions (Di Chiara, 1998), and contains circuits that function to
Obesity is defined based on measurements of body mass index motivate and reinforce food seeking and eating behaviors (Ikemoto
(BMI, overweight ≥25; obese ≥30) (Mei et al., 2002). Clinically, it and Panksepp, 1999). This system also underlies seeking and tak-
is characterized as a condition where excess body fat has accu- ing psychoactive drugs (Ahmed and Koob, 2005; Koob and Le Moal,
mulated to an extent that is detrimental to overall health (James 2005). The fact that such drugs can result in addiction raises the
et al., 2000; Ogden et al., 2007). Current worldwide estimates indi- possibility that highly rewarding, sugar and fat rich foods might
cate that over one billion adults are overweight and that at least also lead to a form of “food addiction”, as binging on such foods,
400 million of these are obese (Finucane et al., 2011). Weight gain like taking psychoactive drugs, evoke a phasic increase in dopamine
is influenced by a range of factors including inherited biological in the NAc shell. This dopamine increase is also observed utilizing
traits, early life experiences, and behavioral, environmental and NAc shell microdialysis in rats binging on sugar solution and high
social factors that influence individual behavior. Work from the fat foods, mimicking the pharmacological effects of many drugs of
Quebec family study has shown that high dietary restraint, high abuse (Avena et al., 2006, 2008; Bocarsly et al., 2014; Rada et al.,
disinhibition and susceptibility to hunger behaviors, in addition 2005).
to short sleep duration, are all associated with excess adiposity
and/or obesity (Chaput et al., 2014). The dramatic increase in obe- 2.1. Intermittent access models of binge-like behavior
sity rates over the past few decades suggest that these changes
are due to environmental, rather than genetic factors (Swinburn Rats provided with intermittent access to sugar, or sugar and
et al., 2004, 2009). Energy intake has increased through the ready fat rich foods for a few hours each day over several weeks exhibit
availability and low cost of energy dense, nutrient-poor foods and behaviors characteristic of addiction. These behaviors include
drinks; especially cereal-based foods (including cakes, biscuits, pies binging, withdrawal, increased motivation or “craving”, and hyper-
and pizza), confectionery and sugar-sweetened drinks (Cook et al., activity similar to that produced by stimulant drugs (Avena et al.,
2001)). These foods contributed almost 36% of adults’ and 41% of 2006; Rada et al., 2005). Consistent with the suggestion that
children’s total energy intake and to 41% and 47% respectively of such foods have addictive-like properties, rats fed palatable high
their total fat intake (Rangan et al., 2008, 2009). While there is fat/high sugar diets have reduced sensitivity to psychostimulant
debate regarding the degree to which sweetened beverage intake rewards, for example, such rats are impaired in acquiring a cocaine-
has contributed to this rise in obesity, the US Department of Agri- reinforced instrumental response (Carroll and Lac, 1998; Wellman
culture reported that per capita soft-drink consumption increased et al., 2007). Moreover, chronic exposure to such foods is associ-
by almost 500% in the past 50 years (Putnam and Allshouse, 1999). ated with long-term alterations in brain reward regions including
Another factor contributing to the rise in obesity is the decrease the striatum (Furlong et al., 2014), again similar to the changes pro-
in energy expenditure through changes in urban design and an duced by chronic exposure to stimulant drugs, such as cocaine and
increased reliance on vehicles and labor-saving devices (Badland amphetamine (LeBlanc et al., 2013; Nelson and Killcross, 2006).
and Schofield, 2006). Obesity is such a health issue because it is an
important determinant of a range of health disorders. Being over- 2.2. Incentive learning processes that may underpin
weight or obese increases the risk of metabolic syndrome, type 2 overconsumption
diabetes, cardiovascular disease, some cancers, respiratory condi-
tions, fatty liver disease, reproductive disorders, depression and Food seeking behaviors are directed to enable the acquisi-
other mental health conditions (Bruce-Keller et al., 2009; Flegal tion of these intrinsically rewarding outcomes. This can lead to
et al., 2007; Haslam and James, 2005). modification of neural processes that underpin appetitive (reward-
This review is based on an invited lecture entitled ‘Why is obe- driven) learning processes – attributing significance to the cues
sity such a problem in the 21st century?’ delivered by Morris to the and responses that predict a significant outcome, termed incen-
International Behavioral Neuroscience Meeting, Las Vegas 2014. tive salience (Robinson and Berridge, 1993). As such, both food and
Covering a range of aspects relevant to our research, it addresses drug associated cues have been shown to increase activity in brain
a number of factors that are associated with obesity and the con- regions involved in learning, memory and motivation including the
sequences of the ready availability of palatable food, including the amygdala, orbitofrontal cortex, and the striatum (Tang et al., 2012).
effects of palatable food on the brain, the role of food cues and the The ‘incentive salience theory’ distinguishes between the hedo-
rewarding aspects of food, in addition to the relationship between nic/pleasure component (‘liking’) of eating and the incentive to eat
stress and food intake. (‘wanting’ or ‘reward-seeking’) when animals are allowed access
to palatable foods. Dysfunction of brain reward systems may lead
2. Effect of obesogenic diets on reward processing to abnormal responsiveness to palatable food and food-associated
cues. Environmental cues or stimuli that occur in conjunction with
Many people report that they feel compelled to eat sweet foods, highly rewarding, palatable foods become imbued with “incentive
even in the absence of metabolic requirements, and animal mod- value”. Over eating as a response to food-associated cues may there-
els have been developed that capture addiction-like aspects of fore reflect a distinction between “wanting” and “liking” (Robinson
palatable high fat/high sugar food consumption that can lead to and Berridge, 1993). “Wanting” is identified with the attribution of
obesity. This consumption is based largely on palatability rather incentive salience to cues associated with palatable food rewards

Please cite this article in press as: Morris, M.J., et al., Why is obesity such a problem in the 21st century? The intersection of palatable food,
cues and reward pathways, stress, and cognition. Neurosci. Biobehav. Rev. (2014), http://dx.doi.org/10.1016/j.neubiorev.2014.12.002
G Model
NBR-2089; No. of Pages 10 ARTICLE IN PRESS
M.J. Morris et al. / Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx 3

and “liking” is the pleasurable effects of the outcome. This model input to reward systems and neurotransmitters, including CB1R,
views obesity as the product of incentive learning processes that orexin and ghrelin (Meye and Adan, 2014).
potentiate the effectiveness of food-related cues, promoting eating Studies indicate that differences in food reward processing
behaviors. exist between overweight and lean individuals and in the way
Furthermore, changes in opioid receptors that regulate the they respond to food associated cues (Burger and Stice, 2011;
hedonic value of palatable foods may disrupt feedback signaling Castellanos et al., 2009; Kenny, 2011; Volkow et al., 2012). These
resulting in enhanced non-homeostatic consumption of palatable cues can promote food consumption even in the absence of hunger
food (Kenny, 2011). This hypothesis is supported by studies in – so-called cue-induced feeding (Petrovich et al., 2005, 2007, 2012;
rodents, including obesity-prone rats, which reveal addiction-like Reppucci and Petrovich, 2012; Walker et al., 2012; Weingarten,
neuroadaptive responses of the reward circuits following chronic 1983). In this paradigm, hungry rats or mice are trained to asso-
consumption of obesogenic foods. Johnson and Kenny (2010) ciate an auditory cue (CS+) or distinct context with deliveries of
reported that extended exposure to palatable foods led to feeding a palatable food. Following training, the animals are permitted to
behaviors in rats that were resistant to disruption when an aversive freely feed on chow in their home cages, so they become sated.
CS predicting electric footshock was presented, and proposed that Rats are returned to the training chambers in a sated state and are
such exposure had led to compulsive-like feeding behaviors. The presented with the palatable food; greater consumption occurred
compulsive nature of this feeding could mean that dietary obese when the cue associated with the food (CS+) was presented as
rats fail to modulate feeding based on negative consequences, or opposed to a cue that was not paired with food (CS−), indicating
that the incentive value of the palatable foods was increased to a that the food-associated cue was capable of evoking a specific moti-
point where feeding overpowers aversive events. vational state, akin to craving (Petrovich et al., 2005, 2007, 2012;
In terms of behavioral changes in rodents following palat- Reppucci and Petrovich, 2012). Furthermore, following training on
able food exposure, differences have been observed in producing a pavlovian discrimination task where CS1 (e.g., click) predicted
goal-directed responses for rewarding foods, altered craving-like delivery of outcome 1 (cherry sucrose) and CS2 (e.g., tone) predicted
behaviors such as enhanced responding for sugar following ces- delivery of outcome 2 (grape maltodextrin), we demonstrated that
sation of the diet (Avena et al., 2005), and changed thresholds rats fed a high fat/high sugar cafeteria diet no longer exhibited CS
in the self-administration of electrical stimulation of the lateral specific food-seeking responses (magazine entries) in a motivation-
hypothalamus (Johnson and Kenny, 2010; la Fleur et al., 2007; ally appropriate manner following devaluation of one outcome by
Pickering et al., 2009). Furthermore, studies of withdrawal of palat- sensory specific satiety (Reichelt et al., 2014). This indicates that
able food access (Martire et al., 2014; South et al., 2012) indicate access to high fat high/sugar diets impaired control of pavlovian
that rats restrict their intake of standard laboratory chow following responding (Reichelt et al., 2014).
cessation of palatable high fat/high sugar foods, suggestive of anhe- It has been proposed that the mesolimbic reward system
donic or withdrawal-like responses. This suggestion is supported increases responsiveness to visual food cues and that the magni-
by decreases in mu-opioid and cannabinoid CB1 receptor mRNA, tude of this neural responsiveness is increased in obesity (Volkow
indicating changes in hedonic processing following cessation of et al., 2012). However, deficits in reward processing may also
access to the palatable diet (Martire et al., 2014). Furthermore, be present in obese individuals in response to overconsumption
these behavioral changes may also be associated with reduced of palatable food during the development of obesity. A marked
dopamine levels and dopamine receptor expression following pro- decline in striatal activity in response to palatable food over this
longed palatable food intake (Alsio et al., 2010; Di Chiara and Tanda, time period was observed in women who gained weight over a
1997; Geiger et al., 2009; Johnson and Kenny, 2010; Kenny et al., 6-month period (mean BMI increase of 4.1%, range 2.5–8.2%) com-
2013; Thanos et al., 2008). pared with women who did not gain weight and maintained a
stable BMI, and the increased BMI negatively correlated with stri-
atal activity (Stice et al., 2010). This reduced striatal response to
2.3. Cue driven mechanisms of food intake the consumption of palatable foods may imply a reduction in sen-
sitivity in reward neurocircuitry, leading to overconsumption to
Unrestrained consumption of palatable food has been shown reach a rewarding “threshold”. Recent work links the fat mass
to increase the reinforcing and rewarding value of foods, trigger- and obesity associated gene (Fto) to the control of dopamine-
ing learned associations between cues that predict the availability 2 (D2R) and dopamine-3 receptor (D3R) dependent signaling,
of food rewards. These external, environmental cues, such as the suggesting that humans carrying FTO gene variants may have
sight and smell of particular foods, or locations where certain foods alterations in D2R-and D3R-dependent behaviors (Hess et al.,
are procured, evoke expectations that a reward will be imminently 2013).
received. These cue-specific reactions are proposed to be respon- A large body of literature indicates that obese individuals can be
sible for continuing drug use in drug-addiction, and in a standard hyper-responsive to food cues, demonstrated by increased BOLD
population, can evoke cravings for particular foods (Jastreboff et al., response in reward processing brain regions (Davids et al., 2010;
2013; Meule et al., 2012, 2014). Demos et al., 2012; Gearhardt et al., 2011; Rothemund et al.,
Cue-evoked food seeking behaviors are consistent with the pro- 2007), indicating that obesity changes food-cue reactivity, partic-
posal that consumption of palatable foods enhances sensitivity to ularly in individuals with increased genetic risk of obesity (Karra
cues associated with food reward and increases responsiveness of et al., 2013; Velders et al., 2012). Furthermore, mesolimbic activ-
reward circuitry to food consumption, potentiating feeding behav- ity following the presentation of food cues is positively correlated
iors and leading to overconsumption (Volkow et al., 2012). Food with BMI (Grosshans et al., 2012; Loeber et al., 2012). This hyper-
and food-related cues are capable of activating reward associated responsiveness to food cues in obese people may be underpinned by
brain circuits, and regions including the OFC, insula, amygdala, the assignment of an increased incentive value to highly palatable,
hypothalamus, striatum, and midbrain regions including the VTA energy dense foods, driving overconsumption through augmented
and substantia nigra (SN) (Bragulat et al., 2010; Pelchat et al., 2004; activity within the mesocorticolimbic circuitry (Burger and Stice,
Schur et al., 2009; Simmons et al., 2005). The VTA plays a pivotal 2011; Castellanos et al., 2009; Volkow et al., 2012). Furthermore,
role in cue-induced food seeking, and several neuromodulators negative affect, such as when under stress, can enhance respon-
including neuropeptides and hormones involved in hedonic and siveness to food cues, evoking an attentional bias (Hepworth et al.,
homeostatic control of feeding behavior appear to have critical 2010). Thus, food-associated cues may promote overconsumption

Please cite this article in press as: Morris, M.J., et al., Why is obesity such a problem in the 21st century? The intersection of palatable food,
cues and reward pathways, stress, and cognition. Neurosci. Biobehav. Rev. (2014), http://dx.doi.org/10.1016/j.neubiorev.2014.12.002
G Model
NBR-2089; No. of Pages 10 ARTICLE IN PRESS
4 M.J. Morris et al. / Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx

in both normal weight and overweight subjects particularly when diet (cafeteria style food) relative to rats provided with chow
experiencing a negative mood. (Maniam and Morris, 2010b). Eating hedonically pleasant food
during stress may dampen HPA axis activity through activation of
3. Stress and eating behavior dopamine activity in the limbic area. Dopamine release is proposed
to act as negative feedback to the hypothalamus to inhibit further
The neural circuits that regulate energy intake converge on release of CRH, a process involved in stress recovery in the HPA
the paraventricular nucleus, which contains the cell bodies of axis that results in reduced corticosterone/cortisol levels.
corticotrophin releasing hormone (CRH) neurons. The paraventric- Several studies have also demonstrated that increases in gluco-
ular nucleus regulates hypothalamo–pituitary–adrenal (HPA) axis corticoid levels promote the intake of palatable foods (Bell et al.,
activity and responds to circulating glucocorticoids and insulin 2000; Bhatnagar et al., 2000). In a recent prospective study exam-
(Dallman et al., 1995; Nieuwenhuizen and Rutters, 2008), thereby ining almost 7000 adolescents, greater stress-related eating was
providing overlap between the stress and feeding systems. Glu- observed in girls, and the so-called ‘stress-driven’ eaters had a
cocorticoids, secreted by the adrenal gland, and insulin from higher prevalence of overweight, obesity and abdominal adipos-
the pancreas, have opposite effects on feeding responses; the ity compared to those who did not eat in response to stress
former increasing and the latter reducing food intake (Strack (Jaaskelainen et al., 2014). A systematic review examining the
et al., 1995). Administration of synthetic glucocorticoid was shown relationship between stress and obesity reported that positive asso-
to promote hyperphagia, body weight gain and hyperinsuline- ciations between stress and body weight were more often identified
mia (Veyrat-Durebex et al., 2012). Glucocorticoids also stimulate in females (Moore and Cunningham, 2012). The possibility of gen-
insulin secretion but affect food intake via the orexigenic neuro- der specific effects warrants further investigation. Another recent
peptide, neuropeptide Y (NPY) (Hanson and Dallman, 1995; Strack study of more than 65,000 older (>50) adults reported that higher
et al., 1995). Glucocorticoids can affect the expression of other levels of perceived stress were associated with greater intake of
hypothalamic feeding neuropeptides; adrenalectomy decreased high fat snacks and fast foods (Barrington et al., 2014). In summary,
proopiomelanocortin (POMC) and agouti-related peptide (AgRP) it appears that stress induced positive energy intake is associated
while replacement of glucocorticoids reversed these effects in with increased risk of developing obesity and its associated diseases
rats (Savontaus et al., 2002). A more detailed discussion of the such as diabetes and cardiovascular disease, which have become a
interactions between glucocorticoids, hypothalamic appetite regu- primary public health concern.
latory neuropeptides, and feeding hormones is provided elsewhere
(Maniam and Morris, 2012). Exposure to chronic stress is also asso- 3.1. Early life stress and other influences on eating
ciated with the development of abdominal obesity in humans. behavior/obesity risk
Bjorntorp (2001) proposed that chronic activation of the HPA
axis by stressors increases glucocorticoid binding to glucocorticoid In addition to the impact of stressful events in adulthood, grow-
receptors (GR). These receptors are highly expressed in abdomi- ing evidence points to the possibility of long term metabolic effects
nal fat, and in the presence of insulin, stimulation of GR activates of an adverse early environment. Severe emotional or physical
lipoprotein lipase which leads to inhibition of lipid mobilization, stress early in life may have long lasting effects, increasing the risk
accumulation of triglyceride and retention of abdominal fat. In line of depression and a range of psychiatric disorders. Early disruption
with the link between stress and obesity, those people reporting of the mother–child relationship, child abuse, neglect, or exposure
a greater number of stressful events had higher BMI (Sinha and to war, modifies stress pathways and elevates plasma corticoste-
Jastreboff, 2013). rone levels in later life (Cohen et al., 2006; Heim and Nemeroff,
Stress can increase or decrease food intake, in both rodents and 1999). Adverse childhood experiences have also been associated
people (Foster et al., 2006; Groesz et al., 2012; Harris et al., 1998; with increased risk of body weight gain across the lifespan. Cross
Marti et al., 1994; McIntosh et al., 1999; Pecoraro et al., 2004; Ryu sectional analysis of a large UK sample showed that maternal stress
et al., 2008; Schulz and Laessle, 2012; Tomiyama et al., 2011). In was associated with early childhood obesity (odds ratio 1.62 at age 3
rodents, most studies demonstrate that stress reduces food intake, years), which was taken to be mediated by an influence on the early
unless palatable food is present during the stress period; this is life environment of the child (Ramasubramanian et al., 2013). Chil-
in line with human studies reporting ‘comfort’ eating in response dren who experienced many negative life events were more likely
to stressful situations (Adam and Epel, 2007; Dallman et al., 2006; to be obese at 15 years of age (Lumeng et al., 2013). A prospec-
Pecoraro et al., 2004; Tomiyama et al., 2011). Such eating is thought tive cohort study of female victims of sexual abuse between the
to involve subcortical areas controlling stress arousal and energy ages of 6 and 27 years reported increased incidence of obesity dur-
storage (HPA axis and limbic areas) as strong motivational drive ing young adulthood (Noll et al., 2007). Adversity during early life
and impulsivity (NAc) (Adam and Epel, 2007). The dopaminergic and adolescence in the form of parental conflict or parental sepa-
system may have a direct influence on the HPA axis. CRH release ration increased the risk of later life obesity (D’Argenio et al., 2009;
following stress has been shown to increase dopamine activity in Gunstad et al., 2006) and markers of metabolic risk (Danese et al.,
the VTA. Wanat et al. (2008) reported that VTA dopamine neuron 2009). Similarly, experience of a range of early life stressors was
firing was dose-dependently increased by CRH, and this firing was positively correlated with increased adult BMI in men, independent
inhibited with CRH-1 receptor antagonism. This effect of CRH on of mental health condition (Gunstad et al., 2006).
dopamine suggests that stress evokes a response to avoid the stres- Alterations in diet during fetal development or the postna-
sor, promoting seeking and eating hedonically pleasant food (Bello tal period can constitute a form of stress. Changes in overall
and Hajnal, 2010). The intake of palatable food (lard or sucrose) food availability (malnutrition or overnutrition), or limitations in
was increased during restraint stress exposure and the physi- selected nutrients (macronutrient and mineral deficiency) dur-
ological stress response was reduced, as evidenced by reduced ing fetal or early postnatal development alters cell structure and
levels of plasma corticosterone, in rats consuming palatable food metabolism, which reprograms stress responsiveness (HPA axis
relative to those only consuming chow (Bell et al., 2002; la Fleur activity) (Barker, 1998; Poore et al., 2010; Schmidt et al., 2008).
et al., 2005). Similar effects were also reported in human studies Early life stress induces hypersecretion of glucocorticoids via
(Macht and Mueller, 2007; Tomiyama et al., 2011). Moreover, the hyperactivation of HPA axis (Lehmann and Feldon, 2000; Lippmann
increased anxiety and hyperactivity of HPA axis induced by early et al., 2007; Macri et al., 2008; Maniam and Morris, 2010a,b). This
life stress were reduced in rats provided with a highly palatable physiological response exposes peripheral tissues to high levels

Please cite this article in press as: Morris, M.J., et al., Why is obesity such a problem in the 21st century? The intersection of palatable food,
cues and reward pathways, stress, and cognition. Neurosci. Biobehav. Rev. (2014), http://dx.doi.org/10.1016/j.neubiorev.2014.12.002
G Model
NBR-2089; No. of Pages 10 ARTICLE IN PRESS
M.J. Morris et al. / Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx 5

of glucocorticoids. At pharmacological doses, glucocorticoids have determine how exposure to adverse early life events may impose
anti-inflammatory actions. However, chronic exposure to high lev- metabolic deficits on offspring.
els of glucocorticoids may elicit metabolic derangements including In summary, stress in adulthood may lead to changes in feeding
accumulation of abdominal fat, dyslipidemia, and insulin/glucose behavior that increase the risk of metabolic disease, and emerg-
intolerance later in life (Maniam et al., 2014; Maniam and Morris, ing evidence points to impacts of early life experience, induced by
2010a,b; Paternain et al., 2013). For example, chronic consump- altered nutrition, or stress exposure in utero or early in life, on off-
tion of high fat diet post-weaning in rats previously exposed to spring food consumption or metabolism, that may increase the risk
early life stress led to a doubling of plasma insulin levels versus of developing obesity in later years.
rats exposed to early life stress but consuming only chow (Maniam
and Morris, 2010a,b). Rats subjected to early life stress and consum-
4. Effects of high energy diets on cognition
ing a diet deficient in omega-3 had elevated plasma insulin levels
and impaired insulin sensitivity compared to rats exposed to early
Cognitive impairments are one of the many adverse health
life stress but fed a control diet (Bernardi et al., 2013). Despite the
effects of the chronic consumption of high energy diets (Alencar
known effects of early life stress on HPA axis activity in later life,
et al., 2010; Lu et al., 2012). However, an important new challenge
little is known regarding how early life stress exposure alters hypo-
has emerged; these cognitive deficits can occur rapidly and in some
thalamic feeding neuropeptides. Feeding is regulated by orexigenic
cases, independently of the metabolic and cardiovascular disorders
and anorexigenic hypothalamic neuropeptides, and is also influ-
associated with obesity.
enced by HPA axis activity and circulating glucocorticoids (Krysiak
A number of cross-sectional, prospective and longitudinal stud-
et al., 1999; Maniam and Morris, 2012). Stress has stimulatory
ies have found associations between diets rich in saturated fat
effects on hypothalamic NPY expression (Dean and White, 1990;
and/or sugar with cognitive deficits after adjusting for demographic
Goto et al., 2006; Sato et al., 2005). We recently proposed that early
and vascular factors. Higher saturated fat intake in mid and later life
life stress exposure may alter hypothalamic feeding neuropeptide
has been associated with worse global cognitive function, impair-
expression which may partly mediate increased food intake and
ments in prospective memory, memory retrieval and flexibility,
adiposity in later life (Maniam et al., 2014).
as well as an increased vulnerability to both normal age related
It is likely that other factors such as 11␤-hydroxysteroid dehy-
cognitive decline and neurological diseases including dementia
drogenase type I (11␤-HSD1) also play a role in mediating the
and Alzheimer’s disease (Eskelinen et al., 2008; Grant et al., 2002;
metabolic effects of early life stress (Maniam et al., 2014). In addi-
Solfrizzi et al., 2010). Likewise, people whose diet contains exces-
tion to circulating cortisol, tissue-specific glucocorticoids play an
sive amounts of refined sugar are more likely to have lower overall
important role in regulating metabolic function (Morgan et al.,
cognition in middle and older age (Ye et al., 2011). Roberts et al.
2014). Intracellular levels of glucocorticoids are tightly regulated
(2012) found that the risk of mild cognitive impairment or demen-
by 11␤-HSD1, which converts inactive glucocorticoid to active glu-
tia was increased in elderly subjects who derived a high percentage
cocorticoid in insulin sensitive tissues such as liver, adipose tissue
of their energy from carbohydrates, but was reduced in those who
and skeletal muscle (Tomlinson et al., 2004). Chronically increased
derived a high percentage of energy from fat and protein. The effects
glucocorticoid action as a result of elevated 11␤-HSD1 activity is
of diet on cognition are not confined to adults. Abargouei et al.
associated with obesity, insulin resistance, type 2 diabetes melli-
(2012) reported that higher sugar intake in school-aged children
tus (Morton et al., 2004; Rask et al., 2001; Tomlinson et al., 2008).
was inversely related to non-verbal intelligence including spatial
Increased 11␤-HSD1 mRNA expression was observed in visceral
memory as measured by Raven’s Progressive Matrices
and subcutaneous fat of obese versus lean women and men (Alberti
et al., 2007; Paulsen et al., 2007). Taken together, these studies sug-
gest a role of increased glucocorticoid availability, particularly in 4.1. Evidence for effects of high energy diets on human cognition
adipose tissue, in promoting obesity.
Only a handful of studies have experimentally controlled their
3.2. Rodent model of early life stress–maternal separation participants’ diet and measured the effects on cognition. As accu-
rate food consumption data is notoriously difficult to obtain outside
Rodent models have shown that early life stress induced by an experimental context, such studies are important as they allow
maternal separation may also affect feeding behavior and influ- causal inferences to be drawn. In healthy, young adult males, a 5 day
ence metabolic disease risk. In our hands rats appear to consume high fat diet (75% of energy) was sufficient to impair attention and
similar amounts of energy, independent of diet, following early speed of retrieval, and to depress mood, as measured by the Cog-
life stress induced by maternal separation, but those consuming nitive Drug Research assessment battery (Holloway et al., 2011).
a high fat/high sugar diet had increased circulating insulin relative Similarly, in sedentary male subjects (25–45 years, <2 h/week phys-
to non-stressed controls, suggesting long term metabolic conse- ical exertion), 7 days of a high fat diet (74% of energy) decreased
quences of early stress (Maniam et al., 2014; Maniam and Morris, reaction times and attention (Edwards et al., 2011). Moreover, eat-
2010a,b). Others have reported changes in food intake following ing a single high glycemic meal before testing impaired memory
maternal separation that were only apparent in the presence of a performance, but increased vigilance, in normal weight undergrad-
stressor in later life, such as isolation stress or restricted feeding. uate women (Nabb and Benton, 2006), school aged children (Micha
For example, post-weaning isolation following maternal separation et al., 2011) and in adults with well-controlled type 2 diabetes
resulted in weight gain and increased food intake (Ryu et al., 2008). (Papanikolaou et al., 2006). A longer term study in children, how-
In an earlier study, maternal separation was associated with greater ever, found no behavioral or cognitive effects of 3 weeks of sucrose,
rebound hyperphagia following restricted feeding (2 h daily for a saccharin or aspartame sweetened diets, regardless of whether the
period of 6 days) in female but not male rats (White et al., 1990). children were reported to respond adversely or normally to sugar
Stress during pregnancy was also shown to affect metabolic func- by their parents (Wolraich et al., 1994).
tion of offspring. Exposure to chronic mild stress during the 3rd A different experimental approach was taken in a study by
week of gestation increased fat mass in those offspring consuming Francis and Stevenson (2011). In Experiment 1, participants com-
high fat/high sucrose diet, suggesting that maternal stress during pleted neuropsychological tests and questionnaires that assessed
development enhances the predisposition to obesity induced by an their saturated fat and refined sugar intake. Food, not BMI or
unhealthy diet (Paternain et al., 2013). Further work is required to general cognitive function, was found to be the best predictor

Please cite this article in press as: Morris, M.J., et al., Why is obesity such a problem in the 21st century? The intersection of palatable food,
cues and reward pathways, stress, and cognition. Neurosci. Biobehav. Rev. (2014), http://dx.doi.org/10.1016/j.neubiorev.2014.12.002
G Model
NBR-2089; No. of Pages 10 ARTICLE IN PRESS
6 M.J. Morris et al. / Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx

We recently showed that rats exposed to a cafeteria diet containing


many of the foods eaten by people with or without a 10% sucrose
solution, or a regular chow diet with a 10% sucrose solution were
impaired on a hippocampal-dependent place recognition task after
5, 11 and 21 days (Beilharz et al., 2014), however they displayed
comparable performance to control rats on a perirhinal dependent
object recognition task (Aggleton and Brown, 2005), suggesting
selective hippocampal deficits. Body weight differences had not
emerged after only 5 days of diet exposure when the impair-
ments were first evident and therefore, obesity was not a causal
factor. Hippocampal inflammation (TNF-␣ and IL-1␤ mRNA) and
oxidative stress (NRF-1 mRNA) markers were significantly elevated
after 1 month of exposure to either cafeteria diet with or without
sucrose or chow supplemented with sucrose, and these markers
were negatively correlated with performance on the place recog-
nition task (see Fig. 2). Hippocampal neurotrophic factors such as
brain-derived neurotrophic factor (BDNF) were not affected by the
Fig. 1. High fat diet increases oxidative stress (OS) in the hippocampus, contributing month long exposure to these diets (Beilharz et al., 2014). Davidson
to decreased levels of neurotrophins such as BDNF, and downstream effects, includ-
ing reduced phosphorylation of synapsin 1, growth-associated protein (GAP)-43 and
et al. (2013) demonstrated that rodents appear to recover from the
(cAMP response element-binding protein) CREB. acute effects of western diet on cognition, and that high ketone
Taken from Wu et al. (2004) with permission. bodies maybe protective (Davidson et al., 2013). Interestingly, a
western diet was also shown to impact on hypothalamic inflam-
mation, eliciting early increases in inflammatory markers in the
of subsequent performance in the hippocampal-dependent ver- mediobasal hypothalamus, which subsided before returning after
bal paired associate task and the logical memory subtests of the one month’s exposure (Thaler et al., 2012). While several stud-
Wechsler memory scale revised. Experiment 2 then compared a ies now indicate that cognitive deficits may precede the onset of
subset of these participants, matched on factors such as BMI and obesity, identification of the relative impact and interrelationships
age, but who reported the highest and lowest intakes of saturated between adiposity, body weight and glucose regulation requires
fats and refined sugars. Individuals with higher saturated fat and more investigation. Taken together these results show that short-
refined sugar consumption performed worse on the hippocampal- term exposure to diets high in fat and sugar, or just high in
dependent memory test (visual reproduction, logical memory, and sugar, impairs aspects of cognition, suggesting that the modern
verbal paired associates) and were less accurate at recalling what diet may rapidly produce impairments in people, including children
they had previously eaten in an experimental snack meal 60 min (Davidson et al., 2005).
prior than those who consumed low amounts of fat and sugar. These results are important because the hippocampus is not
Importantly, these results were obtained in a young population just involved in learning and memory, but also in the control of
with a mainly BMI healthy range, no comorbid medical conditions, eating. The hippocampus contains receptors for hormones and adi-
such as diabetes or cardiovascular disease, and who did not dif- posity signals, such as ghrelin and leptin, and hippocampal neurons
fer on measures of general cognitive function such as attention receive input from multiple brain regions critical for food intake.
and concentration (digit span subtest of the Wechsler adult intel- Previous studies have shown that damage to the hippocampus
ligence scale – fourth edition) and general intelligence (National can increase food intake and result in body weight gain (Davidson
Adult Reading Test). These experimental results in humans there- et al., 2009). Moreover, reversible inactivation of the hippocam-
fore suggest that the effects of fat and sugar on memory may occur pus results in decreased latency in meal onset, increased meal size
independently of any effects on body weight or general health. and a disrupted relationship between meal size and the interval
to the next meal (Henderson et al., 2013). Given that overeat-
4.2. Effects of high energy diets on learning and memory in ing can impair hippocampal function, excessive food intake may
rodents contribute to body weight gain by interfering with hippocampal-
dependent higher-order learning and memory processes. These
Studies with rodents have confirmed that the long-term intake processes include episodic memory, for example, remembering
of high fat/high sugar diets impair cognition. These impairments where, when and what we have previously eaten. This may create a
are especially evident in tasks which require the hippocampus and vicious cycle with high energy diets causing hippocampal dysfunc-
surrounding cortices, such as the Morris water maze where rodents tion which leads to a further overeating of the foods that caused
must use spatial relations among cues to navigate to a hidden plat- the dysfunction in the first place (Davidson et al., 2005, 2007).
form (Heyward et al., 2012; McNay et al., 2010). Impairments have
also been reported on a number of other maze tasks, including
the T-maze (Farr et al., 2008; Pistell et al., 2010), the radial arm 5. Summary and future directions
maze (Murray et al., 2009) and the four arm maze (Valladolid-
Acebes et al., 2011), as well as hippocampal independent operant Unless the current obesity trend is halted, the burden of chronic
lever pressing tasks (Farr et al., 2008) and conditioned discrimi- disease in future generations will lead to a crisis in health and
nation (Greenwood and Winocur, 1990, 1996, 2005; Winocur and economic systems around the world. It is of vital importance to
Greenwood, 1999, 2005). Some of these studies have also identified understand how the modern diet, the foods that we manufac-
changes in the hippocampus which may mediate the link between ture, plus the prevailing stressful environment influence individual
obesity and cognitive deficits including oxidative stress, neuroin- food choices, potentially driving overconsumption of energy. The
flammation, as well as decreased levels of neurotrophic factors omnipresence of cues promoting intake of palatable food, coupled
(Fig. 1). with the rewarding aspects of these appealing, heavily processed
There is also emerging evidence that high energy diets can foods may increase the motivation to eat to excess. Stress also
impair memory in advance of substantial increases in body weight. affects food choice. People may increase the amount of high fat/high

Please cite this article in press as: Morris, M.J., et al., Why is obesity such a problem in the 21st century? The intersection of palatable food,
cues and reward pathways, stress, and cognition. Neurosci. Biobehav. Rev. (2014), http://dx.doi.org/10.1016/j.neubiorev.2014.12.002
G Model
NBR-2089; No. of Pages 10 ARTICLE IN PRESS
M.J. Morris et al. / Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx 7

Fig. 2. Relationship between hippocampal TNF-␣ mRNA expression and exploration ratios on the object (left) and place (right) recognition tasks after 20–21 days on the
regular (open circle), cafeteria with sugar (closed circle), regular with sugar (open triangle) and cafeteria alone (solid square) diets. n = 5–8 rats per group. TNF-␣ mRNA was
negatively correlated with exploration ratios for the place task.
Reproduced from Beilharz et al. (2014) with permission.

sugar food consumed in response to a stressor, making stress Ahmed, S.H., Koob, G.F., 2005. Transition to drug addiction: a negative reinforcement
a risk factor for the development of obesity in vulnerable indi- model based on an allostatic decrease in reward function. Psychopharmacology
(Berl.) 180, 473–490.
viduals. Emerging evidence linking severe early life stress and Alberti, L., Girola, A., Gilardini, L., Conti, A., Cattaldo, S., Micheletto, G., Invitti, C.,
later metabolic disorders needs additional work to identify the 2007. Type 2 diabetes and metabolic syndrome are associated with increased
mechanisms underlying this association. Animal and human data expression of 11beta-hydroxysteroid dehydrogenase 1 in obese subjects. Int. J.
Obes. (Lond.) 31, 1826–1831.
indicate that high fat/high sugar foods can also impair some forms Alencar, R.C., Cobas, R.A., Gomes, M.B., 2010. Assessment of cognitive status in
of hippocampal-dependent memories, which may further impair patients with type 2 diabetes through the mini-mental status examination: a
appetite control by undermining the processes involved in seeking cross-sectional study. Diabetol. Metab. Syndr. 2, 10.
Alsio, J., Olszewski, P.K., Norback, A.H., Gunnarsson, Z.E., Levine, A.S., Pickering,
and consuming food. Further research is also necessary to deter-
C., Schioth, H.B., 2010. Dopamine D1 receptor gene expression decreases in
mine the mechanisms through which obesity and its comorbidities the nucleus accumbens upon long-term exposure to palatable food and dif-
may exacerbate cognitive impairments, particularly with aging, fers depending on diet-induced obesity phenotype in rats. Neuroscience 171,
779–787.
where optimal nutrition is critical to preserve cognitive function.
Avena, N.M., Bocarsly, M.E., Rada, P., Kim, A., Hoebel, B.G., 2008. After daily binge-
Understanding the mechanisms underpinning the impact of other ing on a sucrose solution, food deprivation induces anxiety and accumbens
preventive/supportive strategies such as increasing physical activ- dopamine/acetylcholine imbalance. Physiol. Behav. 94, 309–315.
ity, should also be a priority. Such information is vital to provide Avena, N.M., Long, K.A., Hoebel, B.G., 2005. Sugar-dependent rats show enhanced
responding for sugar after abstinence: evidence of a sugar deprivation effect.
important insights into the drivers of obesity and to enable novel Physiol. Behav. 84, 359–362.
interventions to ameliorate the epidemic. Avena, N.M., Rada, P., Hoebel, B.G., 2006. Sugar bingeing in rats. Curr. Protoc. Neu-
In the 21st century, obesity should be viewed not only as a rosci. 36, 23, C:29.23C.21–29.23C.26.
Badland, H., Schofield, G., 2006. Understanding the relationship between town size
metabolic disorder, but as a multifactorial disease. Thus, strate- and physical activity levels: a population study. Health Place 12, 538–546.
gies that focus on modulation of the reward system, reduction of Barker, D.J., 1998. In utero programming of chronic disease. Clin. Sci. (Lond.) 95,
stress reactivity and reverse compromised hippocampal function 115–128.
Barrington, W.E., Beresford, S.A., McGregor, B.A., White, E., 2014. Perceived stress
may be more effective than the currently available pharmacolog- and eating behaviors by sex, obesity status, and stress vulnerability: find-
ical approaches typically targeting appetite suppression. Overall, ings from the vitamins and lifestyle (VITAL) study. J. Acad. Nutr. Diet 14,
understanding the factors that increase the vulnerability of indi- 1791–1799.
Beilharz, J.E., Maniam, J., Morris, M.J., 2014. Short exposure to a diet rich in both fat
viduals to develop obesity is essential in developing more effective
and sugar or sugar alone impairs place, but not object recognition memory in
treatments. Of course, providing education and support to indi- rats. Brain Behav. Immun. 37, 134–141.
viduals and populations in strategies for promoting healthy food Bell, M.E., Bhargava, A., Soriano, L., Laugero, K., Akana, S.F., Dallman, M.F., 2002.
Sucrose intake and corticosterone interact with cold to modulate ingestive
intake, and appropriate physical activity, are also of critical impor-
behaviour, energy balance, autonomic outflow and neuroendocrine responses
tance in the prevention of obesity. during chronic stress. J. Neuroendocrinol. 14, 330–342.
Bell, M.E., Bhatnagar, S., Akana, S.F., Choi, S., Dallman, M.F., 2000. Disruption of
arcuate/paraventricular nucleus connections changes body energy balance and
Acknowledgement response to acute stress. J. Neurosci. 20, 6707–6713.
Bello, N.T., Hajnal, A., 2010. Dopamine and binge eating behaviors. Pharmacol.
Biochem. Behav. 97, 25–33.
This work was supported by NHMRC Project grant 1023073
Bernardi, J.R., Ferreira, C.F., Senter, G., Krolow, R., de Aguiar, B.W., Portella, A.K.,
awarded to MJM and RFW. Kauer-Sant’anna, M., Kapczinski, F., Dalmaz, C., Goldani, M.Z., Silveira, P.P., 2013.
Early life stress interacts with the diet deficiency of omega-3 fatty acids during
the life course increasing the metabolic vulnerability in adult rats. PLOS ONE 8,
References e62031.
Bhatnagar, S., Bell, M.E., Liang, J., Soriano, L., Nagy, T.R., Dallman, M.F., 2000.
Abargouei, A.S., Kalantari, N., Omidvar, N., Rashidkhani, B., Rad, A.H., Ebrahimi, A.A., Corticosterone facilitates saccharin intake in adrenalectomized rats: does corti-
Khosravi-Boroujeni, H., Esmaillzadeh, A., 2012. Refined carbohydrate intake in costerone increase stimulus salience? J. Neuroendocrinol. 12, 453–460.
relation to non-verbal intelligence among Tehrani schoolchildren. Public Health Bjorntorp, P., 2001. Do stress reactions cause abdominal obesity and comorbidities?
Nutr. 15, 1925–1931. Obes. Rev. 2, 73–86.
Adam, T.C., Epel, E.S., 2007. Stress, eating and the reward system. Physiol. Behav. 91, Bocarsly, M.E., Hoebel, B.G., Paredes, D., von Loga, I., Murray, S.M., Wang, M., Arolfo,
449–458. M.P., Yao, L., Diamond, I., Avena, N.M., 2014. GS 455534 selectively suppresses
Aggleton, J.P., Brown, M.W., 2005. Contrasting hippocampal and perirhinal cortex binge eating of palatable food and attenuates dopamine release in the accum-
function using immediate early gene imaging. Q. J. Exp. Psychol. B 58, 218–233. bens of sugar-bingeing rats. Behav. Pharmacol. 25, 147–157.

Please cite this article in press as: Morris, M.J., et al., Why is obesity such a problem in the 21st century? The intersection of palatable food,
cues and reward pathways, stress, and cognition. Neurosci. Biobehav. Rev. (2014), http://dx.doi.org/10.1016/j.neubiorev.2014.12.002
G Model
NBR-2089; No. of Pages 10 ARTICLE IN PRESS
8 M.J. Morris et al. / Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx

Bragulat, V., Dzemidzic, M., Bruno, C., Cox, C.A., Talavage, T., Considine, R.V., Kareken, Furlong, T.M., Jayaweera, H.K., Balleine, B.W., Corbit, L.H., 2014. Binge-like consump-
D.A., 2010. Food-related odor probes of brain reward circuits during hunger: a tion of a palatable food accelerates habitual control of behavior and is dependent
pilot FMRI study. Obesity (Silver Spring) 18, 1566–1571. on activation of the dorsolateral striatum. J. Neurosci. 34, 5012–5022.
Bruce-Keller, A.J., Keller, J.N., Morrison, C.D., 2009. Obesity and vulnerability of the Gearhardt, A.N., Yokum, S., Orr, P.T., Stice, E., Corbin, W.R., Brownell, K.D., 2011.
CNS. Biochim. Biophys. Acta 1792, 395–400. Neural correlates of food addiction. Arch. Gen. Psychiatry 68, 808–816.
Burger, K.S., Stice, E., 2011. Variability in reward responsivity and obesity: evidence Geiger, B.M., Haburcak, M., Avena, N.M., Moyer, M.C., Hoebel, B.G., Pothos, E.N.,
from brain imaging studies. Curr. Drug Abuse Rev. 4, 182–189. 2009. Deficits of mesolimbic dopamine neurotransmission in rat dietary obesity.
Carroll, M.E., Lac, S.T., 1998. Dietary additives and the acquisition of cocaine self- Neuroscience 159, 1193–1199.
administration in rats. Psychopharmacology (Berl.) 137, 81–89. Goto, M., Arima, H., Watanabe, M., Hayashi, M., Banno, R., Sato, I., Nagasaki, H.,
Castellanos, E.H., Charboneau, E., Dietrich, M.S., Park, S., Bradley, B.P., Mogg, Oiso, Y., 2006. Ghrelin increases neuropeptide Y and agouti-related peptide
K., Cowan, R.L., 2009. Obese adults have visual attention bias for food cue gene expression in the arcuate nucleus in rat hypothalamic organotypic cultures.
images: evidence for altered reward system function. Int. J. Obes. (Lond.) 33, Endocrinology 147, 5102–5109.
1063–1073. Grant, W.B., Campbell, A., Itzhaki, R.F., Savory, J., 2002. The significance of envi-
Chaput, J.P., Perusse, L., Despres, J.P., Tremblay, A., Bouchard, C., 2014. Findings from ronmental factors in the etiology of Alzheimer’s disease. J. Alzheimers Dis. 4,
the Quebec family study on the etiology of obesity: genetics and environmental 179–189.
highlights. Curr. Obes. Rep. 3, 54–66. Greenwood, C.E., Winocur, G., 1990. Learning and memory impairment in rats fed a
Cohen, R.A., Grieve, S., Hoth, K.F., Paul, R.H., Sweet, L., Tate, D., Gunstad, J., Stroud, L., high saturated fat diet. Behav. Neural Biol. 53, 74–87.
McCaffery, J., Hitsman, B., Niaura, R., Clark, C.R., McFarlane, A., Bryant, R., Gordon, Greenwood, C.E., Winocur, G., 1996. Cognitive impairment in rats fed high-fat diets:
E., Williams, L.M., 2006. Early life stress and morphometry of the adult anterior a specific effect of saturated fatty-acid intake. Behav. Neurosci. 110, 451–459.
cingulate cortex and caudate nuclei. Biol. Psychiatry 59, 975–982. Greenwood, C.E., Winocur, G., 2005. High-fat diets, insulin resistance and declining
Cook, T., Rutishauser, I., Seelig, M., 2001. Comparable Measurements from the 1983, cognitive function. Neurobiol. Aging 26 (Suppl. 1), 42–45.
1985 and 1995 National Nutrition Surveys. Australian Food and Nutrition Mon- Groesz, L.M., McCoy, S., Carl, J., Saslow, L., Stewart, J., Adler, N., Laraia, B., Epel, E.,
itoring Unit, Canberra. 2012. What is eating you? Stress and the drive to eat. Appetite 58, 717–721.
D’Argenio, A., Mazzi, C., Pecchioli, L., Di Lorenzo, G., Siracusano, A., Troisi, A., 2009. Grosshans, M., Vollmert, C., Vollstadt-Klein, S., Tost, H., Leber, S., Bach, P., Buhler, M.,
Early trauma and adult obesity: is psychological dysfunction the mediating von der Goltz, C., Mutschler, J., Loeber, S., Hermann, D., Wiedemann, K., Meyer-
mechanism? Physiol. Behav. 98, 543–546. Lindenberg, A., Kiefer, F., 2012. Association of leptin with food cue-induced
Dallman, M.F., Akana, S.F., Strack, A.M., Hanson, E.S., Sebastian, R.J., 1995. The neu- activation in human reward pathways. Arch. Gen. Psychiatry 69, 529–537.
ral network that regulates energy balance is responsive to glucocorticoids and Gunstad, J., Paul, R.H., Spitznagel, M.B., Cohen, R.A., Williams, L.M., Kohn, M., Gordon,
insulin and also regulates HPA axis responsivity at a site proximal to CRF neu- E., 2006. Exposure to early life trauma is associated with adult obesity. Psychiatry
rons. Ann. N. Y. Acad. Sci. 771, 730–742. Res. 142, 31–37.
Dallman, M.F., Pecoraro, N.C., La Fleur, S.E., Warne, J.P., Ginsberg, A.B., Akana, S.F., Hanson, E.S., Dallman, M.F., 1995. Neuropeptide Y (NPY) may integrate responses of
Laugero, K.C., Houshyar, H., Strack, A.M., Bhatnagar, S., Bell, M.E., 2006. Gluco- hypothalamic feeding systems and the hypothalamo–pituitary–adrenal axis. J.
corticoids, chronic stress, and obesity. Prog. Brain Res. 153, 75–105. Neuroendocrinol. 7, 273–279.
Danese, A., Moffitt, T.E., Harrington, H., Milne, B.J., Polanczyk, G., Pariante, C.M., Poul- Harris, R.B., Zhou, J., Youngblood, B.D., Rybkin, I.I., Smagin, G.N., Ryan, D.H., 1998.
ton, R., Caspi, A., 2009. Adverse childhood experiences and adult risk factors for Effect of repeated stress on body weight and body composition of rats fed low-
age-related disease: depression, inflammation, and clustering of metabolic risk and high-fat diets. Am. J. Physiol. 275, R1928–R1938.
markers. Arch. Pediatr. Adolesc. Med. 163, 1135–1143. Haslam, D.W., James, W.P., 2005. Obesity. Lancet 366, 1197–1209.
Davids, S., Lauffer, H., Thoms, K., Jagdhuhn, M., Hirschfeld, H., Domin, M., Hamm, Heim, C., Nemeroff, C.B., 1999. The impact of early adverse experiences on brain
A., Lotze, M., 2010. Increased dorsolateral prefrontal cortex activation in obese systems involved in the pathophysiology of anxiety and affective disorders. Biol.
children during observation of food stimuli. Int. J. Obes. (Lond.) 34, 94–104. Psychiatry 46, 1509–1522.
Davidson, T.L., Chan, K., Jarrard, L.E., Kanoski, S.E., Clegg, D.J., Benoit, S.C., 2009. Con- Henderson, Y.O., Smith, G.P., Parent, M.B., 2013. Hippocampal neurons inhibit meal
tributions of the hippocampus and medial prefrontal cortex to energy and body onset. Hippocampus 23, 100–107.
weight regulation. Hippocampus 19, 235–252. Hepworth, R., Mogg, K., Brignell, C., Bradley, B.P., 2010. Negative mood increases
Davidson, T.L., Hargrave, S.L., Swithers, S.E., Sample, C.H., Fu, X., Kinzig, K.P., Zheng, selective attention to food cues and subjective appetite. Appetite 54, 134–142.
W., 2013. Inter-relationships among diet, obesity and hippocampal-dependent Hess, M.E., Hess, S., Meyer, K.D., Verhagen, L.A., Koch, L., Bronneke, H.S., Dietrich,
cognitive function. Neuroscience 253, 110–122. M.O., Jordan, S.D., Saletore, Y., Elemento, O., Belgardt, B.F., Franz, T., Horvath, T.L.,
Davidson, T.L., Kanoski, S.E., Schier, L.A., Clegg, D.J., Benoit, S.C., 2007. A potential role Ruther, U., Jaffrey, S.R., Kloppenburg, P., Bruning, J.C., 2013. The fat mass and
for the hippocampus in energy intake and body weight regulation. Curr. Opin. obesity associated gene (Fto) regulates activity of the dopaminergic midbrain
Pharmacol. 7, 613–616. circuitry. Nat. Neurosci. 16, 1042–1048.
Davidson, T.L., Kanoski, S.E., Walls, E.K., Jarrard, L.E., 2005. Memory inhibition and Heyward, F.D., Walton, R.G., Carle, M.S., Coleman, M.A., Garvey, W.T., Sweatt, J.D.,
energy regulation. Physiol. Behav. 86, 731–746. 2012. Adult mice maintained on a high-fat diet exhibit object location mem-
Dean, R.G., White, B.D., 1990. Neuropeptide Y expression in rat brain: effects of ory deficits and reduced hippocampal SIRT1 gene expression. Neurobiol. Learn.
adrenalectomy. Neurosci. Lett. 114, 339–344. Mem. 98, 25–32.
Demos, K.E., Heatherton, T.F., Kelley, W.M., 2012. Individual differences in nucleus Holloway, C.J., Cochlin, L.E., Emmanuel, Y., Murray, A., Codreanu, I., Edwards, L.M.,
accumbens activity to food and sexual images predict weight gain and sexual Szmigielski, C., Tyler, D.J., Knight, N.S., Saxby, B.K., Lambert, B., Thompson, C.,
behavior. J. Neurosci. 32, 5549–5552. Neubauer, S., Clarke, K., 2011. A high-fat diet impairs cardiac high-energy phos-
Di Chiara, G., 1998. A motivational learning hypothesis of the role of mesolimbic phate metabolism and cognitive function in healthy human subjects. Am. J. Clin.
dopamine in compulsive drug use. J. Psychopharmacol. 12, 54–67. Nutr. 93, 748–755.
Di Chiara, G., Tanda, G., 1997. Blunting of reactivity of dopamine transmission to Ikemoto, S., Panksepp, J., 1999. The role of nucleus accumbens dopamine in
palatable food: a biochemical marker of anhedonia in the CMS model? Psy- motivated behavior: a unifying interpretation with special reference to reward-
chopharmacology (Berl.) 134, 351–353. seeking. Brain Res. Brain Res. Rev. 31, 6–41.
Edwards, L.M., Murray, A.J., Holloway, C.J., Carter, E.E., Kemp, G.J., Codreanu, I., Jaaskelainen, A., Nevanpera, N., Remes, J., Rahkonen, F., Jarvelin, M.R., Laitinen, J.,
Brooker, H., Tyler, D.J., Robbins, P.A., Clarke, K., 2011. Short-term consump- 2014. Stress-related eating, obesity and associated behavioural traits in ado-
tion of a high-fat diet impairs whole-body efficiency and cognitive function in lescents: a prospective population-based cohort study. BMC Public Health 14,
sedentary men. FASEB J. 25, 1088–1096. 321.
Eskelinen, M.H., Ngandu, T., Helkala, E.L., Tuomilehto, J., Nissinen, A., Soininen, H., James, W.P., Astrup, A., Finer, N., Hilsted, J., Kopelman, P., Rossner, S., Saris, W.H., Van
Kivipelto, M., 2008. Fat intake at midlife and cognitive impairment later in life: Gaal, L.F., 2000. Effect of sibutramine on weight maintenance after weight loss:
a population-based CAIDE study. Int. J. Geriatr. Psychiatry 23, 741–747. a randomised trial. STORM study group. Sibutramine trial of obesity reduction
Farr, S.A., Yamada, K.A., Butterfield, D.A., Abdul, H.M., Xu, L., Miller, N.E., Banks, and maintenance. Lancet 356, 2119–2125.
W.A., Morley, J.E., 2008. Obesity and hypertriglyceridemia produce cognitive Jastreboff, A.M., Sinha, R., Lacadie, C., Small, D.M., Sherwin, R.S., Potenza, M.N., 2013.
impairment. Endocrinology 149, 2628–2636. Neural correlates of stress- and food cue-induced food craving in obesity: asso-
Finucane, M.M., Stevens, G.A., Cowan, M.J., Danaei, G., Lin, J.K., Paciorek, C.J., Singh, ciation with insulin levels. Diabetes Care 36, 394–402.
G.M., Gutierrez, H.R., Lu, Y., Bahalim, A.N., Farzadfar, F., Riley, L.M., Ezzati, M., Johnson, P.M., Kenny, P.J., 2010. Dopamine D2 receptors in addiction-like reward
Global Burden of Metabolic Risk Factors of Chronic Diseases Collaborating, G., dysfunction and compulsive eating in obese rats. Nat. Neurosci. 13, 635–641.
2011. National, regional, and global trends in body-mass index since 1980: sys- Karra, E., O’Daly, O.G., Choudhury, A.I., Yousseif, A., Millership, S., Neary, M.T., Scott,
tematic analysis of health examination surveys and epidemiological studies with W.R., Chandarana, K., Manning, S., Hess, M.E., Iwakura, H., Akamizu, T., Millet, Q.,
960 country-years and 9.1 million participants. Lancet 377, 557–567. Gelegen, C., Drew, M.E., Rahman, S., Emmanuel, J.J., Williams, S.C., Ruther, U.U.,
Flegal, K.M., Graubard, B.I., Williamson, D.F., Gail, M.H., 2007. Cause-specific excess Bruning, J.C., Withers, D.J., Zelaya, F.O., Batterham, R.L., 2013. A link between
deaths associated with underweight, overweight, and obesity. JAMA 298, FTO, ghrelin, and impaired brain food-cue responsivity. J. Clin. Invest. 123,
2028–2037. 3539–3551.
Foster, M.T., Solomon, M.B., Huhman, K.L., Bartness, T.J., 2006. Social defeat increases Kenny, P.J., 2011. Reward mechanisms in obesity: new insights and future directions.
food intake, body mass, and adiposity in Syrian hamsters. Am. J. Physiol. Regul. Neuron 69, 664–679.
Integr. Comp. Physiol. 290, R1284–R1293. Kenny, P.J., Voren, G., Johnson, P.M., 2013. Dopamine D2 receptors and striatopallidal
Francis, H.M., Stevenson, R.J., 2011. Higher reported saturated fat and refined sugar transmission in addiction and obesity. Curr. Opin. Neurobiol. 23, 535–538.
intake is associated with reduced hippocampal-dependent memory and sensi- Koob, G.F., Le Moal, M., 2005. Plasticity of reward neurocircuitry and the ‘dark side’
tivity to interoceptive signals. Behav. Neurosci. 125, 943–955. of drug addiction. Nat. Neurosci. 8, 1442–1444.

Please cite this article in press as: Morris, M.J., et al., Why is obesity such a problem in the 21st century? The intersection of palatable food,
cues and reward pathways, stress, and cognition. Neurosci. Biobehav. Rev. (2014), http://dx.doi.org/10.1016/j.neubiorev.2014.12.002
G Model
NBR-2089; No. of Pages 10 ARTICLE IN PRESS
M.J. Morris et al. / Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx 9

Krysiak, R., Obuchowicz, E., Herman, Z.S., 1999. Interactions between the neu- Nabb, S., Benton, D., 2006. The influence on cognition of the interaction between the
ropeptide Y system and the hypothalamic–pituitary–adrenal axis. Eur. J. macro-nutrient content of breakfast and glucose tolerance. Physiol. Behav. 87,
Endocrinol. 140, 130–136. 16–23.
la Fleur, S.E., Houshyar, H., Roy, M., Dallman, M.F., 2005. Choice of lard, but not total Nelson, A., Killcross, S., 2006. Amphetamine exposure enhances habit formation. J.
lard calories, damps adrenocorticotropin responses to restraint. Endocrinology Neurosci. 26, 3805–3812.
146, 2193–2199. Nieuwenhuizen, A.G., Rutters, F., 2008. The hypothalamic–pituitary–adrenal-axis in
la Fleur, S.E., Vanderschuren, L.J., Luijendijk, M.C., Kloeze, B.M., Tiesjema, B., Adan, the regulation of energy balance. Physiol. Behav. 94, 169–177.
R.A., 2007. A reciprocal interaction between food-motivated behavior and diet- Noll, J.G., Zeller, M.H., Trickett, P.K., Putnam, F.W., 2007. Obesity risk for female
induced obesity. Int. J. Obes. (Lond.) 31, 1286–1294. victims of childhood sexual abuse: a prospective study. Pediatrics 120, e61–e67.
LeBlanc, K.H., Maidment, N.T., Ostlund, S.B., 2013. Repeated cocaine exposure facili- Ogden, C.L., Yanovski, S.Z., Carroll, M.D., Flegal, K.M., 2007. The epidemiology of
tates the expression of incentive motivation and induces habitual control in rats. obesity. Gastroenterology 132, 2087–2102.
PLOS ONE 8, e61355. Papanikolaou, Y., Palmer, H., Binns, M.A., Jenkins, D.J., Greenwood, C.E., 2006. Better
Lehmann, J., Feldon, J., 2000. Long-term biobehavioral effects of maternal separation cognitive performance following a low-glycaemic-index compared with a high-
in the rat: consistent or confusing? Rev. Neurosci. 11, 383–408. glycaemic-index carbohydrate meal in adults with type 2 diabetes. Diabetologia
Lippmann, M., Bress, A., Nemeroff, C.B., Plotsky, P.M., Monteggia, L.M., 2007. Long- 49, 855–862.
term behavioural and molecular alterations associated with maternal separation Paternain, L., de la Garza, A.L., Batlle, M.A., Milagro, F.I., Martinez, J.A., Campion, J.,
in rats. Eur. J. Neurosci. 25, 3091–3098. 2013. Prenatal stress increases the obesogenic effects of a high-fat-sucrose diet
Loeber, S., Grosshans, M., Korucuoglu, O., Vollmert, C., Vollstadt-Klein, S., Schneider, in adult rats in a sex-specific manner. Stress 16, 220–232.
S., Wiers, R.W., Mann, K., Kiefer, F., 2012. Impairment of inhibitory control in Paulsen, S.K., Pedersen, S.B., Fisker, S., Richelsen, B., 2007. 11Beta-HSD type 1 expres-
response to food-associated cues and attentional bias of obese participants and sion in human adipose tissue: impact of gender, obesity, and fat localization.
normal-weight controls. Int. J. Obes. (Lond.) 36, 1334–1339. Obesity (Silver Spring) 15, 1954–1960.
Lu, Y., Lu, J., Wang, S., Li, C., Liu, L., Zheng, R., Tian, H., Wang, X., Yang, L., Zhang, Y., Pecoraro, N., Reyes, F., Gomez, F., Bhargava, A., Dallman, M.F., 2004. Chronic stress
Pan, C., 2012. Cognitive function with glucose tolerance status and obesity in promotes palatable feeding, which reduces signs of stress: feedforward and
Chinese middle-aged and aged adults. Aging Ment. Health 16, 911–914. feedback effects of chronic stress. Endocrinology 145, 3754–3762.
Lumeng, J.C., Wendorf, K., Pesch, M.H., Appugliese, D.P., Kaciroti, N., Corwyn, R.F., Pelchat, M.L., Johnson, A., Chan, R., Valdez, J., Ragland, J.D., 2004. Images of desire:
Bradley, R.H., 2013. Overweight adolescents and life events in childhood. Pedi- food-craving activation during fMRI. Neuroimage 23, 1486–1493.
atrics 132, e1506–e1512. Petrovich, G.D., Hobin, M.P., Reppucci, C.J., 2012. Selective Fos induction in hypo-
Lutter, M., Nestler, E.J., 2009. Homeostatic and hedonic signals interact in the regu- thalamic orexin/hypocretin, but not melanin-concentrating hormone neurons,
lation of food intake. J. Nutr. 139, 629–632. by a learned food-cue that stimulates feeding in sated rats. Neuroscience 224,
Macht, M., Mueller, J., 2007. Immediate effects of chocolate on experimentally 70–80.
induced mood states. Appetite 49, 667–674. Petrovich, G.D., Holland, P.C., Gallagher, M., 2005. Amygdalar and prefrontal path-
Macri, S., Chiarotti, F., Wurbel, H., 2008. Maternal separation and maternal care act ways to the lateral hypothalamus are activated by a learned cue that stimulates
independently on the development of HPA responses in male rats. Behav. Brain eating. J. Neurosci. 25, 8295–8302.
Res. 191, 227–234. Petrovich, G.D., Ross, C.A., Gallagher, M., Holland, P.C., 2007. Learned contextual cue
Maniam, J., Antoniadis, C., Morris, M.J., 2014. Early-life stress, HPA axis adapta- potentiates eating in rats. Physiol. Behav. 90, 362–367.
tion, and mechanisms contributing to later health outcomes. Front. Endocrinol. Pickering, C., Alsio, J., Hulting, A.L., Schioth, H.B., 2009. Withdrawal from free-choice
(Lausanne) 5, 73. high-fat high-sugar diet induces craving only in obesity-prone animals. Psy-
Maniam, J., Morris, M.J., 2010a. Long-term postpartum anxiety and depression-like chopharmacology (Berl.) 204, 431–443.
behavior in mother rats subjected to maternal separation are ameliorated by Pistell, P.J., Morrison, C.D., Gupta, S., Knight, A.G., Keller, J.N., Ingram, D.K., Bruce-
palatable high fat diet. Behav. Brain Res. 208, 72–79. Keller, A.J., 2010. Cognitive impairment following high fat diet consumption is
Maniam, J., Morris, M.J., 2010b. Palatable cafeteria diet ameliorates anxiety and associated with brain inflammation. J. Neuroimmunol. 219, 25–32.
depression-like symptoms following an adverse early environment. Psychoneu- Poore, K.R., Boullin, J.P., Cleal, J.K., Newman, J.P., Noakes, D.E., Hanson, M.A., Green,
roendocrinology 35, 717–728. L.R., 2010. Sex- and age-specific effects of nutrition in early gestation and
Maniam, J., Morris, M.J., 2012. The link between stress and feeding behaviour. Neu- early postnatal life on hypothalamo-pituitary–adrenal axis and sympathoad-
ropharmacology 63, 97–110. renal function in adult sheep. J. Physiol. 588, 2219–2237.
Marti, O., Marti, J., Armario, A., 1994. Effects of chronic stress on food intake in rats: Putnam, J.J., Allshouse, J.E., 1999. Food Consumption, Prices, and Expenditures,
influence of stressor intensity and duration of daily exposure. Physiol. Behav. 1970–97. E.R.S. Food and Consumers Economics Division, U.S. Department of
55, 747–753. Agriculture, Washington, DC.
Martire, S.I., Maniam, J., South, T., Holmes, N., Westbrook, R.F., Morris, M.J., 2014. Rada, P., Avena, N.M., Hoebel, B.G., 2005. Daily bingeing on sugar repeatedly releases
Extended exposure to a palatable cafeteria diet alters gene expression in dopamine in the accumbens shell. Neuroscience 134, 737–744.
brain regions implicated in reward, and withdrawal from this diet alters gene Ramasubramanian, L., Lane, S., Rahman, A., 2013. The association between mater-
expression in brain regions associated with stress. Behav. Brain Res. 265, nal serious psychological distress and child obesity at 3 years: a cross-sectional
132–141. analysis of the UK Millennium Cohort Data. Child Care Health Dev. 39, 134–140.
McIntosh, J., Anisman, H., Merali, Z., 1999. Short- and long-periods of neona- Rangan, A.M., Randall, D., Hector, D.J., Gill, T.P., Webb, K.L., 2008. Consumption of
tal maternal separation differentially affect anxiety and feeding in adult rats: ‘extra’ foods by Australian children: types, quantities and contribution to energy
gender-dependent effects. Brain Res. Dev. Brain Res. 113, 97–106. and nutrient intakes. Eur. J. Clin. Nutr. 62, 356–364.
McNay, E.C., Ong, C.T., McCrimmon, R.J., Cresswell, J., Bogan, J.S., Sherwin, R.S., 2010. Rangan, A.M., Schindeler, S., Hector, D.J., Gill, T.P., Webb, K.L., 2009. Consumption of
Hippocampal memory processes are modulated by insulin and high-fat-induced ‘extra’ foods by Australian adults: types, quantities and contribution to energy
insulin resistance. Neurobiol. Learn. Mem. 93, 546–553. and nutrient intakes. Eur. J. Clin. Nutr. 63, 865–871.
Mei, Z., Grummer-Strawn, L.M., Pietrobelli, A., Goulding, A., Goran, M.I., Dietz, W.H., Rask, E., Olsson, T., Soderberg, S., Andrew, R., Livingstone, D.E., Johnson, O., Walker,
2002. Validity of body mass index compared with other body-composition B.R., 2001. Tissue-specific dysregulation of cortisol metabolism in human obe-
screening indexes for the assessment of body fatness in children and adoles- sity. J. Clin. Endocrinol. Metab. 86, 1418–1421.
cents. Am. J. Clin. Nutr. 75, 978–985. Reichelt, A.C., Morris, M.J., Westbrook, R.F., 2014. Cafeteria diet impairs expression of
Meule, A., Lutz, A., Vogele, C., Kubler, A., 2012. Food cravings discriminate differen- sensory-specific satiety and stimulus-outcome learning. Front. Psychol. 5, 852.
tially between successful and unsuccessful dieters and non-dieters. Validation Reppucci, C.J., Petrovich, G.D., 2012. Learned food-cue stimulates persistent feeding
of the Food Cravings Questionnaires in German. Appetite 58, 88–97. in sated rats. Appetite 59, 437–447.
Meule, A., Lutz, A.P., Vogele, C., Kubler, A., 2014. Impulsive reactions to food-cues Roberts, R.O., Roberts, L.A., Geda, Y.E., Cha, R.H., Pankratz, V.S., O’Connor, H.M., Knop-
predict subsequent food craving. Eat. Behav. 15, 99–105. man, D.S., Petersen, R.C., 2012. Relative intake of macronutrients impacts risk of
Meye, F.J., Adan, R.A., 2014. Feelings about food: the ventral tegmental area in food mild cognitive impairment or dementia. J. Alzheimers Dis. 32, 329–339.
reward and emotional eating. Trends Pharmacol. Sci. 35, 31–40. Robinson, T.E., Berridge, K.C., 1993. The neural basis of drug craving: an incentive-
Micha, R., Rogers, P.J., Nelson, M., 2011. Glycaemic index and glycaemic load of sensitization theory of addiction. Brain Res. Brain Res. Rev. 18, 247–291.
breakfast predict cognitive function and mood in school children: a randomised Rothemund, Y., Preuschhof, C., Bohner, G., Bauknecht, H.C., Klingebiel, R., Flor, H.,
controlled trial. Br. J. Nutr. 106, 1552–1561. Klapp, B.F., 2007. Differential activation of the dorsal striatum by high-calorie
Moore, C.J., Cunningham, S.A., 2012. Social position, psychological stress, and obe- visual food stimuli in obese individuals. Neuroimage 37, 410–421.
sity: a systematic review. J. Acad. Nutr. Diet. 112, 518–526. Ryu, V., Lee, J.H., Yoo, S.B., Gu, X.F., Moon, Y.W., Jahng, J.W., 2008. Sustained hyper-
Morgan, S.A., McCabe, E.L., Gathercole, L.L., Hassan-Smith, Z.K., Larner, D.P., Bujalska, phagia in adolescent rats that experienced neonatal maternal separation. Int. J.
I.J., Stewart, P.M., Tomlinson, J.W., Lavery, G.G., 2014. 11␤-HSD1 is the major Obes. (Lond.) 32, 1355–1362.
regulator of the tissue-specific effects of circulating glucocorticoid excess. Proc. Saper, C.B., Chou, T.C., Elmquist, J.K., 2002. The need to feed: homeostatic and hedonic
Natl. Acad. Sci. 111, E2482–E2491. control of eating. Neuron 36, 199–211.
Morton, N.M., Paterson, J.M., Masuzaki, H., Holmes, M.C., Staels, B., Fievet, C., Walker, Sato, I., Arima, H., Ozaki, N., Watanabe, M., Goto, M., Hayashi, M., Banno, R., Nagasaki,
B.R., Flier, J.S., Mullins, J.J., Seckl, J.R., 2004. Novel adipose tissue-mediated resis- H., Oiso, Y., 2005. Insulin inhibits neuropeptide Y gene expression in the arcuate
tance to diet-induced visceral obesity in 11 beta-hydroxysteroid dehydrogenase nucleus through GABAergic systems. J. Neurosci. 25, 8657–8664.
type 1-deficient mice. Diabetes 53, 931–938. Savontaus, E., Conwell, I.M., Wardlaw, S.L., 2002. Effects of adrenalectomy on AGRP,
Murray, A.J., Knight, N.S., Cochlin, L.E., McAleese, S., Deacon, R.M., Rawlins, J.N., POMC, NPY and CART gene expression in the basal hypothalamus of fed and
Clarke, K., 2009. Deterioration of physical performance and cognitive function fasted rats. Brain Res. 958, 130–138.
in rats with short-term high-fat feeding. FASEB J. 23, 4353–4360.

Please cite this article in press as: Morris, M.J., et al., Why is obesity such a problem in the 21st century? The intersection of palatable food,
cues and reward pathways, stress, and cognition. Neurosci. Biobehav. Rev. (2014), http://dx.doi.org/10.1016/j.neubiorev.2014.12.002
G Model
NBR-2089; No. of Pages 10 ARTICLE IN PRESS
10 M.J. Morris et al. / Neuroscience and Biobehavioral Reviews xxx (2014) xxx–xxx

Schmidt, M.V., Liebl, C., Sterlemann, V., Ganea, K., Hartmann, J., Harbich, Tomlinson, J.W., Walker, E.A., Bujalska, I.J., Draper, N., Lavery, G.G., Cooper, M.S.,
D., Alam, S., Muller, M.B., 2008. Neuropeptide Y mediates the initial Hewison, M., Stewart, P.M., 2004. 11beta-hydroxysteroid dehydrogenase type
hypothalamic–pituitary–adrenal response to maternal separation in the neona- 1: a tissue-specific regulator of glucocorticoid response. Endocr. Rev. 25,
tal mouse. J. Endocrinol. 197, 421–427. 831–866.
Schulz, S., Laessle, R.G., 2012. Stress-induced laboratory eating behavior in obese Valladolid-Acebes, I., Stucchi, P., Cano, V., Fernandez-Alfonso, M.S., Merino, B., Gil-
women with binge eating disorder. Appetite 58, 457–461. Ortega, M., Fole, A., Morales, L., Ruiz-Gayo, M., Del Olmo, N., 2011. High-fat diets
Schur, E.A., Kleinhans, N.M., Goldberg, J., Buchwald, D., Schwartz, M.W., Maravilla, impair spatial learning in the radial-arm maze in mice. Neurobiol. Learn. Mem.
K., 2009. Activation in brain energy regulation and reward centers by food cues 95, 80–85.
varies with choice of visual stimulus. Int. J. Obes. (Lond.) 33, 653–661. Velders, F.P., De Wit, J.E., Jansen, P.W., Jaddoe, V.W., Hofman, A., Verhulst, F.C.,
Simmons, W.K., Martin, A., Barsalou, L.W., 2005. Pictures of appetizing foods activate Tiemeier, H., 2012. FTO at rs9939609, food responsiveness, emotional control
gustatory cortices for taste and reward. Cereb. Cortex 15, 1602–1608. and symptoms of ADHD in preschool children. PLoS ONE 7, e49131.
Sinha, R., Jastreboff, A.M., 2013. Stress as a common risk factor for obesity and Veyrat-Durebex, C., Deblon, N., Caillon, A., Andrew, R., Altirriba, J., Odermatt,
addiction. Biol. Psychiatry 73, 827–835. A., Rohner-Jeanrenaud, F., 2012. Central glucocorticoid administration pro-
Solfrizzi, V., Frisardi, V., Capurso, C., D’Introno, A., Colacicco, A.M., Vendemiale, G., motes weight gain and increased 11beta-hydroxysteroid dehydrogenase type
Capurso, A., Panza, F., 2010. Dietary fatty acids in dementia and predementia syn- 1 expression in white adipose tissue. PLoS ONE 7, e34002.
dromes: epidemiological evidence and possible underlying mechanisms. Ageing Volkow, N.D., Wang, G.J., Fowler, J.S., Tomasi, D., Baler, R., 2012. Food and drug
Res. Rev. 9, 184–199. reward: overlapping circuits in human obesity and addiction. Curr. Top. Behav.
South, T., Westbrook, F., Morris, M.J., 2012. Neurological and stress related effects of Neurosci. 11, 1–24.
shifting obese rats from a palatable diet to chow and lean rats from chow to a Walker, A.K., Ibia, I.E., Zigman, J.M., 2012. Disruption of cue-potentiated feeding in
palatable diet. Physiol. Behav. 105, 1052–1057. mice with blocked ghrelin signaling. Physiol. Behav. 108, 34–43.
Stice, E., Yokum, S., Blum, K., Bohon, C., 2010. Weight gain is associated with reduced Wanat, M.J., Hopf, F.W., Stuber, G.D., Phillips, P.E., Bonci, A., 2008. Corticotropin-
striatal response to palatable food. J. Neurosci. 30, 13105–13109. releasing factor increases mouse ventral tegmental area dopamine neuron
Strack, A.M., Sebastian, R.J., Schwartz, M.W., Dallman, M.F., 1995. Glucocorti- firing through a protein kinase C-dependent enhancement of Ih. J. Physiol. 586,
coids and insulin: reciprocal signals for energy balance. Am. J. Physiol. 268, 2157–2170.
R142–R149. Weingarten, H.P., 1983. Conditioned cues elicit feeding in sated rats: a role for
Swinburn, B., Sacks, G., Ravussin, E., 2009. Increased food energy supply is more learning in meal initiation. Science 220, 431–433.
than sufficient to explain the US epidemic of obesity. Am. J. Clin. Nutr. 90, Wellman, P.J., Nation, J.R., Davis, K.W., 2007. Impairment of acquisition of cocaine
1453–1456. self-administration in rats maintained on a high-fat diet. Pharmacol. Biochem.
Swinburn, B.A., Caterson, I., Seidell, J.C., James, W.P., 2004. Diet, nutrition and the Behav. 88, 89–93.
prevention of excess weight gain and obesity. Public Health Nutr. 7, 123–146. White, B.D., Dean, R.G., Martin, R.J., 1990. Adrenalectomy decreases neuropeptide Y
Tang, D.W., Fellows, L.K., Small, D.M., Dagher, A., 2012. Food and drug cues activate mRNA levels in the arcuate nucleus. Brain Res. Bull. 25, 711–715.
similar brain regions: a meta-analysis of functional MRI studies. Physiol. Behav. World Health Organization. (2000). Obesity: preventing and managing the global
106, 317–324. epidemic (No. 894). World Health Organization.
Thaler, J.P., Yi, C.X., Schur, E.A., Guyenet, S.J., Hwang, B.H., Dietrich, M.O., Zhao, X., Winocur, G., Greenwood, C.E., 1999. The effects of high fat diets and environ-
Sarruf, D.A., Izgur, V., Maravilla, K.R., Nguyen, H.T., Fischer, J.D., Matsen, M.E., mental influences on cognitive performance in rats. Behav. Brain Res. 101,
Wisse, B.E., Morton, G.J., Horvath, T.L., Baskin, D.G., Tschop, M.H., Schwartz, M.W., 153–161.
2012. Obesity is associated with hypothalamic injury in rodents and humans. J. Winocur, G., Greenwood, C.E., 2005. Studies of the effects of high fat diets on cogni-
Clin. Invest. 122, 153–162. tive function in a rat model. Neurobiol Aging 26 (Suppl. 1), 46–49.
Thanos, P.K., Michaelides, M., Piyis, Y.K., Wang, G.J., Volkow, N.D., 2008. Food restric- Wolraich, M.L., Lindgren, S.D., Stumbo, P.J., Stegink, L.D., Appelbaum, M.I., Kiritsy,
tion markedly increases dopamine D2 receptor (D2R) in a rat model of obesity M.C., 1994. Effects of diets high in sucrose or aspartame on the behavior and
as assessed with in-vivo muPET imaging ([11C] raclopride) and in-vitro ([3H] cognitive performance of children. N. Engl. J. Med. 330, 301–307.
spiperone) autoradiography. Synapse 62, 50–61. Wu, A., Ying, Z., Gomez-Pinilla, F., 2004. The interplay between oxidative stress and
Tomiyama, A.J., Dallman, M.F., Epel, E.S., 2011. Comfort food is comforting to those brain-derived neurotrophic factor modulates the outcome of a saturated fat diet
most stressed: evidence of the chronic stress response network in high stress on synaptic plasticity and cognition. Eur. J. Neurosci. 19, 1699–1707.
women. Psychoneuroendocrinology 36, 1513–1519. Ye, X., Gao, X., Scott, T., Tucker, K.L., 2011. Habitual sugar intake and cognitive func-
Tomlinson, J.W., Finney, J., Gay, C., Hughes, B.A., Hughes, S.V., Stewart, P.M., 2008. tion among middle-aged and older Puerto Ricans without diabetes. Br. J. Nutr.
Impaired glucose tolerance and insulin resistance are associated with increased 106, 1423–1432.
adipose 11beta-hydroxysteroid dehydrogenase type 1 expression and elevated
hepatic 5alpha-reductase activity. Diabetes 57, 2652–2660.

Please cite this article in press as: Morris, M.J., et al., Why is obesity such a problem in the 21st century? The intersection of palatable food,
cues and reward pathways, stress, and cognition. Neurosci. Biobehav. Rev. (2014), http://dx.doi.org/10.1016/j.neubiorev.2014.12.002

You might also like