You are on page 1of 10

Neuroscience and Biobehavioral Reviews 58 (2015) 3645

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
b

School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia


School of Psychology, UNSW Australia, Sydney, NSW 2052, Australia

a r t i c l e

i n f o

Article history:
Received 1 July 2014
Received in revised form
15 November 2014
Accepted 2 December 2014
Available online 10 December 2014
Keywords:
Obesity
Stress
Overeating
Hippocampus
Reward
Dopamine
Memory

a b s t r a c t
Changes in food composition and availability have contributed to the dramatic increase in obesity over
the past 3040 years in developed and, increasingly, in developing countries. The brain plays a critical
role in regulating energy balance. Some human studies have demonstrated increased preference for high
fat and high sugar foods in people reporting greater stress exposure. We have examined neurochemical
changes in the brain in rodent models during the development of obesity, including the impact of obesity 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
appraisal of foods, indicative of an addiction-like capacity of foods high in fat and/or sugar. Importantly,
withdrawal of the palatable diet led to a stress-like response. Furthermore, access to this palatable diet
attenuated the physiological effects of acute stress (restraint), indicating that it could act as a comfort
food. In more chronic studies, the diet also attenuated anxiety-like behavior in rats exposed to stress
(maternal separation) early in life, but these rats may suffer greater metabolic harm than rats exposed
to the early life stressor but not provided with the palatable diet.
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 inammatory markers in the hippocampus, a critical region involved in memory.
Obesity-related inammatory 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.
2.

3.

4.

Introduction what are the drivers of obesity? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .


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

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.

37
37
37
37
38
39
39
40
40
40
41

M.J. Morris et al. / Neuroscience and Biobehavioral Reviews 58 (2015) 3645

5.

Summary and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .


Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

1. Introduction what are the drivers of obesity?


Obesity is a major health issue in the developed and, increasingly, in the developing world; indeed obesity is considered the
most serious health issue facing the developed world, formally recognized as such by the World Health Organization (WHO, 2000).
Obesity is dened based on measurements of body mass index
(BMI, overweight 25; obese 30) (Mei et al., 2002). Clinically, it
is characterized as a condition where excess body fat has accumulated to an extent that is detrimental to overall health (James
et al., 2000; Ogden et al., 2007). Current worldwide estimates indicate that over one billion adults are overweight and that at least
400 million of these are obese (Finucane et al., 2011). Weight gain
is inuenced by a range of factors including inherited biological
traits, early life experiences, and behavioral, environmental and
social factors that inuence individual behavior. Work from the
Quebec family study has shown that high dietary restraint, high
disinhibition and susceptibility to hunger behaviors, in addition
to short sleep duration, are all associated with excess adiposity
and/or obesity (Chaput et al., 2014). The dramatic increase in obesity rates over the past few decades suggest that these changes
are due to environmental, rather than genetic factors (Swinburn
et al., 2004, 2009). Energy intake has increased through the ready
availability and low cost of energy dense, nutrient-poor foods and
drinks; especially cereal-based foods (including cakes, biscuits, pies
and pizza), confectionery and sugar-sweetened drinks (Cook et al.,
2001)). These foods contributed almost 36% of adults and 41% of
childrens total energy intake and to 41% and 47% respectively of
their total fat intake (Rangan et al., 2008, 2009). While there is
debate regarding the degree to which sweetened beverage intake
has contributed to this rise in obesity, the US Department of Agriculture reported that per capita soft-drink consumption increased
by almost 500% in the past 50 years (Putnam and Allshouse, 1999).
Another factor contributing to the rise in obesity is the decrease
in energy expenditure through changes in urban design and an
increased reliance on vehicles and labor-saving devices (Badland
and Schoeld, 2006). Obesity is such a health issue because it is an
important determinant of a range of health disorders. Being overweight or obese increases the risk of metabolic syndrome, type 2
diabetes, cardiovascular disease, some cancers, respiratory conditions, fatty liver disease, reproductive disorders, depression and
other mental health conditions (Bruce-Keller et al., 2009; Flegal
et al., 2007; Haslam and James, 2005).
This review is based on an invited lecture entitled Why is obesity such a problem in the 21st century? delivered by Morris to the
International Behavioral Neuroscience Meeting, Las Vegas 2014.
Covering a range of aspects relevant to our research, it addresses
a number of factors that are associated with obesity and the consequences of the ready availability of palatable food, including the
effects of palatable food on the brain, the role of food cues and the
rewarding aspects of food, in addition to the relationship between
stress and food intake.
2. Effect of obesogenic diets on reward processing
Many people report that they feel compelled to eat sweet foods,
even in the absence of metabolic requirements, and animal models have been developed that capture addiction-like aspects of
palatable high fat/high sugar food consumption that can lead to
obesity. This consumption is based largely on palatability rather

37

41
42
42

than nutrient requirements, and driven by the availability of such


foods (Lutter and Nestler, 2009; Meye and Adan, 2014; Saper et al.,
2002). The mesocortical dopamine system plays an important role
in reward-related processes. This system extends from the ventral
tegmental area (VTA) via the nucleus accumbens (NAc) to cortical regions (Di Chiara, 1998), and contains circuits that function to
motivate and reinforce food seeking and eating behaviors (Ikemoto
and Panksepp, 1999). This system also underlies seeking and taking psychoactive drugs (Ahmed and Koob, 2005; Koob and Le Moal,
2005). The fact that such drugs can result in addiction raises the
possibility that highly rewarding, sugar and fat rich foods might
also lead to a form of food addiction, as binging on such foods,
like taking psychoactive drugs, evoke a phasic increase in dopamine
in the NAc shell. This dopamine increase is also observed utilizing
NAc shell microdialysis in rats binging on sugar solution and high
fat foods, mimicking the pharmacological effects of many drugs of
abuse (Avena et al., 2006, 2008; Bocarsly et al., 2014; Rada et al.,
2005).
2.1. Intermittent access models of binge-like behavior
Rats provided with intermittent access to sugar, or sugar and
fat rich foods for a few hours each day over several weeks exhibit
behaviors characteristic of addiction. These behaviors include
binging, withdrawal, increased motivation or craving, and hyperactivity similar to that produced by stimulant drugs (Avena et al.,
2006; Rada et al., 2005). Consistent with the suggestion that
such foods have addictive-like properties, rats fed palatable high
fat/high sugar diets have reduced sensitivity to psychostimulant
rewards, for example, such rats are impaired in acquiring a cocainereinforced instrumental response (Carroll and Lac, 1998; Wellman
et al., 2007). Moreover, chronic exposure to such foods is associated with long-term alterations in brain reward regions including
the striatum (Furlong et al., 2014), again similar to the changes produced by chronic exposure to stimulant drugs, such as cocaine and
amphetamine (LeBlanc et al., 2013; Nelson and Killcross, 2006).
2.2. Incentive learning processes that may underpin
overconsumption
Food seeking behaviors are directed to enable the acquisition of these intrinsically rewarding outcomes. This can lead to
modication of neural processes that underpin appetitive (rewarddriven) learning processes attributing signicance to the cues
and responses that predict a signicant outcome, termed incentive salience (Robinson and Berridge, 1993). As such, both food and
drug associated cues have been shown to increase activity in brain
regions involved in learning, memory and motivation including the
amygdala, orbitofrontal cortex, and the striatum (Tang et al., 2012).
The incentive salience theory distinguishes between the hedonic/pleasure component (liking) of eating and the incentive to eat
(wanting or reward-seeking) when animals are allowed access
to palatable foods. Dysfunction of brain reward systems may lead
to abnormal responsiveness to palatable food and food-associated
cues. Environmental cues or stimuli that occur in conjunction with
highly rewarding, palatable foods become imbued with incentive
value. Over eating as a response to food-associated cues may therefore reect a distinction between wanting and liking (Robinson
and Berridge, 1993). Wanting is identied with the attribution of
incentive salience to cues associated with palatable food rewards

38

M.J. Morris et al. / Neuroscience and Biobehavioral Reviews 58 (2015) 3645

and liking is the pleasurable effects of the outcome. This model


views obesity as the product of incentive learning processes that
potentiate the effectiveness of food-related cues, promoting eating
behaviors.
Furthermore, changes in opioid receptors that regulate the
hedonic value of palatable foods may disrupt feedback signaling
resulting in enhanced non-homeostatic consumption of palatable
food (Kenny, 2011). This hypothesis is supported by studies in
rodents, including obesity-prone rats, which reveal addiction-like
neuroadaptive responses of the reward circuits following chronic
consumption of obesogenic foods. Johnson and Kenny (2010)
reported that extended exposure to palatable foods led to feeding
behaviors in rats that were resistant to disruption when an aversive
CS predicting electric footshock was presented, and proposed that
such exposure had led to compulsive-like feeding behaviors. The
compulsive nature of this feeding could mean that dietary obese
rats fail to modulate feeding based on negative consequences, or
that the incentive value of the palatable foods was increased to a
point where feeding overpowers aversive events.
In terms of behavioral changes in rodents following palatable food exposure, differences have been observed in producing
goal-directed responses for rewarding foods, altered craving-like
behaviors such as enhanced responding for sugar following cessation of the diet (Avena et al., 2005), and changed thresholds
in the self-administration of electrical stimulation of the lateral
hypothalamus (Johnson and Kenny, 2010; la Fleur et al., 2007;
Pickering et al., 2009). Furthermore, studies of withdrawal of palatable food access (Martire et al., 2014; South et al., 2012) indicate
that rats restrict their intake of standard laboratory chow following
cessation of palatable high fat/high sugar foods, suggestive of anhedonic or withdrawal-like responses. This suggestion is supported
by decreases in mu-opioid and cannabinoid CB1 receptor mRNA,
indicating changes in hedonic processing following cessation of
access to the palatable diet (Martire et al., 2014). Furthermore,
these behavioral changes may also be associated with reduced
dopamine levels and dopamine receptor expression following prolonged palatable food intake (Alsio et al., 2010; Di Chiara and Tanda,
1997; Geiger et al., 2009; Johnson and Kenny, 2010; Kenny et al.,
2013; Thanos et al., 2008).

2.3. Cue driven mechanisms of food intake


Unrestrained consumption of palatable food has been shown
to increase the reinforcing and rewarding value of foods, triggering learned associations between cues that predict the availability
of food rewards. These external, environmental cues, such as the
sight and smell of particular foods, or locations where certain foods
are procured, evoke expectations that a reward will be imminently
received. These cue-specic reactions are proposed to be responsible for continuing drug use in drug-addiction, and in a standard
population, can evoke cravings for particular foods (Jastreboff et al.,
2013; Meule et al., 2012, 2014).
Cue-evoked food seeking behaviors are consistent with the proposal that consumption of palatable foods enhances sensitivity to
cues associated with food reward and increases responsiveness of
reward circuitry to food consumption, potentiating feeding behaviors and leading to overconsumption (Volkow et al., 2012). Food
and food-related cues are capable of activating reward associated
brain circuits, and regions including the OFC, insula, amygdala,
hypothalamus, striatum, and midbrain regions including the VTA
and substantia nigra (SN) (Bragulat et al., 2010; Pelchat et al., 2004;
Schur et al., 2009; Simmons et al., 2005). The VTA plays a pivotal
role in cue-induced food seeking, and several neuromodulators
including neuropeptides and hormones involved in hedonic and
homeostatic control of feeding behavior appear to have critical

input to reward systems and neurotransmitters, including CB1R,


orexin and ghrelin (Meye and Adan, 2014).
Studies indicate that differences in food reward processing
exist between overweight and lean individuals and in the way
they respond to food associated cues (Burger and Stice, 2011;
Castellanos et al., 2009; Kenny, 2011; Volkow et al., 2012). These
cues can promote food consumption even in the absence of hunger
so-called cue-induced feeding (Petrovich et al., 2005, 2007, 2012;
Reppucci and Petrovich, 2012; Walker et al., 2012; Weingarten,
1983). In this paradigm, hungry rats or mice are trained to associate an auditory cue (CS+) or distinct context with deliveries of
a palatable food. Following training, the animals are permitted to
freely feed on chow in their home cages, so they become sated.
Rats are returned to the training chambers in a sated state and are
presented with the palatable food; greater consumption occurred
when the cue associated with the food (CS+) was presented as
opposed to a cue that was not paired with food (CS), indicating
that the food-associated cue was capable of evoking a specic motivational state, akin to craving (Petrovich et al., 2005, 2007, 2012;
Reppucci and Petrovich, 2012). Furthermore, following training on
a pavlovian discrimination task where CS1 (e.g., click) predicted
delivery of outcome 1 (cherry sucrose) and CS2 (e.g., tone) predicted
delivery of outcome 2 (grape maltodextrin), we demonstrated that
rats fed a high fat/high sugar cafeteria diet no longer exhibited CS
specic food-seeking responses (magazine entries) in a motivationally appropriate manner following devaluation of one outcome by
sensory specic satiety (Reichelt et al., 2014). This indicates that
access to high fat high/sugar diets impaired control of pavlovian
responding (Reichelt et al., 2014).
It has been proposed that the mesolimbic reward system
increases responsiveness to visual food cues and that the magnitude of this neural responsiveness is increased in obesity (Volkow
et al., 2012). However, decits in reward processing may also
be present in obese individuals in response to overconsumption
of palatable food during the development of obesity. A marked
decline in striatal activity in response to palatable food over this
time period was observed in women who gained weight over a
6-month period (mean BMI increase of 4.1%, range 2.58.2%) compared with women who did not gain weight and maintained a
stable BMI, and the increased BMI negatively correlated with striatal activity (Stice et al., 2010). This reduced striatal response to
the consumption of palatable foods may imply a reduction in sensitivity in reward neurocircuitry, leading to overconsumption to
reach a rewarding threshold. Recent work links the fat mass
and obesity associated gene (Fto) to the control of dopamine2 (D2R) and dopamine-3 receptor (D3R) dependent signaling,
suggesting that humans carrying FTO gene variants may have
alterations in D2R-and D3R-dependent behaviors (Hess et al.,
2013).
A large body of literature indicates that obese individuals can be
hyper-responsive to food cues, demonstrated by increased BOLD
response in reward processing brain regions (Davids et al., 2010;
Demos et al., 2012; Gearhardt et al., 2011; Rothemund et al.,
2007), indicating that obesity changes food-cue reactivity, particularly in individuals with increased genetic risk of obesity (Karra
et al., 2013; Velders et al., 2012). Furthermore, mesolimbic activity following the presentation of food cues is positively correlated
with BMI (Grosshans et al., 2012; Loeber et al., 2012). This hyperresponsiveness to food cues in obese people may be underpinned by
the assignment of an increased incentive value to highly palatable,
energy dense foods, driving overconsumption through augmented
activity within the mesocorticolimbic circuitry (Burger and Stice,
2011; Castellanos et al., 2009; Volkow et al., 2012). Furthermore,
negative affect, such as when under stress, can enhance responsiveness to food cues, evoking an attentional bias (Hepworth et al.,
2010). Thus, food-associated cues may promote overconsumption

M.J. Morris et al. / Neuroscience and Biobehavioral Reviews 58 (2015) 3645

in both normal weight and overweight subjects particularly when


experiencing a negative mood.
3. Stress and eating behavior
The neural circuits that regulate energy intake converge on
the paraventricular nucleus, which contains the cell bodies of
corticotrophin releasing hormone (CRH) neurons. The paraventricular nucleus regulates hypothalamopituitaryadrenal (HPA) axis
activity and responds to circulating glucocorticoids and insulin
(Dallman et al., 1995; Nieuwenhuizen and Rutters, 2008), thereby
providing overlap between the stress and feeding systems. Glucocorticoids, secreted by the adrenal gland, and insulin from
the pancreas, have opposite effects on feeding responses; the
former increasing and the latter reducing food intake (Strack
et al., 1995). Administration of synthetic glucocorticoid was shown
to promote hyperphagia, body weight gain and hyperinsulinemia (Veyrat-Durebex et al., 2012). Glucocorticoids also stimulate
insulin secretion but affect food intake via the orexigenic neuropeptide, neuropeptide Y (NPY) (Hanson and Dallman, 1995; Strack
et al., 1995). Glucocorticoids can affect the expression of other
hypothalamic feeding neuropeptides; adrenalectomy decreased
proopiomelanocortin (POMC) and agouti-related peptide (AgRP)
while replacement of glucocorticoids reversed these effects in
rats (Savontaus et al., 2002). A more detailed discussion of the
interactions between glucocorticoids, hypothalamic appetite regulatory neuropeptides, and feeding hormones is provided elsewhere
(Maniam and Morris, 2012). Exposure to chronic stress is also associated with the development of abdominal obesity in humans.
Bjorntorp (2001) proposed that chronic activation of the HPA
axis by stressors increases glucocorticoid binding to glucocorticoid
receptors (GR). These receptors are highly expressed in abdominal fat, and in the presence of insulin, stimulation of GR activates
lipoprotein lipase which leads to inhibition of lipid mobilization,
accumulation of triglyceride and retention of abdominal fat. In line
with the link between stress and obesity, those people reporting
a greater number of stressful events had higher BMI (Sinha and
Jastreboff, 2013).
Stress can increase or decrease food intake, in both rodents and
people (Foster et al., 2006; Groesz et al., 2012; Harris et al., 1998;
Marti et al., 1994; McIntosh et al., 1999; Pecoraro et al., 2004; Ryu
et al., 2008; Schulz and Laessle, 2012; Tomiyama et al., 2011). In
rodents, most studies demonstrate that stress reduces food intake,
unless palatable food is present during the stress period; this is
in line with human studies reporting comfort eating in response
to stressful situations (Adam and Epel, 2007; Dallman et al., 2006;
Pecoraro et al., 2004; Tomiyama et al., 2011). Such eating is thought
to involve subcortical areas controlling stress arousal and energy
storage (HPA axis and limbic areas) as strong motivational drive
and impulsivity (NAc) (Adam and Epel, 2007). The dopaminergic
system may have a direct inuence on the HPA axis. CRH release
following stress has been shown to increase dopamine activity in
the VTA. Wanat et al. (2008) reported that VTA dopamine neuron
ring was dose-dependently increased by CRH, and this ring was
inhibited with CRH-1 receptor antagonism. This effect of CRH on
dopamine suggests that stress evokes a response to avoid the stressor, promoting seeking and eating hedonically pleasant food (Bello
and Hajnal, 2010). The intake of palatable food (lard or sucrose)
was increased during restraint stress exposure and the physiological stress response was reduced, as evidenced by reduced
levels of plasma corticosterone, in rats consuming palatable food
relative to those only consuming chow (Bell et al., 2002; la Fleur
et al., 2005). Similar effects were also reported in human studies
(Macht and Mueller, 2007; Tomiyama et al., 2011). Moreover, the
increased anxiety and hyperactivity of HPA axis induced by early
life stress were reduced in rats provided with a highly palatable

39

diet (cafeteria style food) relative to rats provided with chow


(Maniam and Morris, 2010b). Eating hedonically pleasant food
during stress may dampen HPA axis activity through activation of
dopamine activity in the limbic area. Dopamine release is proposed
to act as negative feedback to the hypothalamus to inhibit further
release of CRH, a process involved in stress recovery in the HPA
axis that results in reduced corticosterone/cortisol levels.
Several studies have also demonstrated that increases in glucocorticoid levels promote the intake of palatable foods (Bell et al.,
2000; Bhatnagar et al., 2000). In a recent prospective study examining almost 7000 adolescents, greater stress-related eating was
observed in girls, and the so-called stress-driven eaters had a
higher prevalence of overweight, obesity and abdominal adiposity compared to those who did not eat in response to stress
(Jaaskelainen et al., 2014). A systematic review examining the
relationship between stress and obesity reported that positive associations between stress and body weight were more often identied
in females (Moore and Cunningham, 2012). The possibility of gender specic effects warrants further investigation. Another recent
study of more than 65,000 older (>50) adults reported that higher
levels of perceived stress were associated with greater intake of
high fat snacks and fast foods (Barrington et al., 2014). In summary,
it appears that stress induced positive energy intake is associated
with increased risk of developing obesity and its associated diseases
such as diabetes and cardiovascular disease, which have become a
primary public health concern.
3.1. Early life stress and other inuences on eating
behavior/obesity risk
In addition to the impact of stressful events in adulthood, growing evidence points to the possibility of long term metabolic effects
of an adverse early environment. Severe emotional or physical
stress early in life may have long lasting effects, increasing the risk
of depression and a range of psychiatric disorders. Early disruption
of the motherchild relationship, child abuse, neglect, or exposure
to war, modies stress pathways and elevates plasma corticosterone levels in later life (Cohen et al., 2006; Heim and Nemeroff,
1999). Adverse childhood experiences have also been associated
with increased risk of body weight gain across the lifespan. Cross
sectional analysis of a large UK sample showed that maternal stress
was associated with early childhood obesity (odds ratio 1.62 at age 3
years), which was taken to be mediated by an inuence on the early
life environment of the child (Ramasubramanian et al., 2013). Children who experienced many negative life events were more likely
to be obese at 15 years of age (Lumeng et al., 2013). A prospective cohort study of female victims of sexual abuse between the
ages of 6 and 27 years reported increased incidence of obesity during young adulthood (Noll et al., 2007). Adversity during early life
and adolescence in the form of parental conict or parental separation increased the risk of later life obesity (DArgenio et al., 2009;
Gunstad et al., 2006) and markers of metabolic risk (Danese et al.,
2009). Similarly, experience of a range of early life stressors was
positively correlated with increased adult BMI in men, independent
of mental health condition (Gunstad et al., 2006).
Alterations in diet during fetal development or the postnatal period can constitute a form of stress. Changes in overall
food availability (malnutrition or overnutrition), or limitations in
selected nutrients (macronutrient and mineral deciency) during fetal or early postnatal development alters cell structure and
metabolism, which reprograms stress responsiveness (HPA axis
activity) (Barker, 1998; Poore et al., 2010; Schmidt et al., 2008).
Early life stress induces hypersecretion of glucocorticoids via
hyperactivation of HPA axis (Lehmann and Feldon, 2000; Lippmann
et al., 2007; Macri et al., 2008; Maniam and Morris, 2010a,b). This
physiological response exposes peripheral tissues to high levels

40

M.J. Morris et al. / Neuroscience and Biobehavioral Reviews 58 (2015) 3645

of glucocorticoids. At pharmacological doses, glucocorticoids have


anti-inammatory actions. However, chronic exposure to high levels of glucocorticoids may elicit metabolic derangements including
accumulation of abdominal fat, dyslipidemia, and insulin/glucose
intolerance later in life (Maniam et al., 2014; Maniam and Morris,
2010a,b; Paternain et al., 2013). For example, chronic consumption of high fat diet post-weaning in rats previously exposed to
early life stress led to a doubling of plasma insulin levels versus
rats exposed to early life stress but consuming only chow (Maniam
and Morris, 2010a,b). Rats subjected to early life stress and consuming a diet decient in omega-3 had elevated plasma insulin levels
and impaired insulin sensitivity compared to rats exposed to early
life stress but fed a control diet (Bernardi et al., 2013). Despite the
known effects of early life stress on HPA axis activity in later life,
little is known regarding how early life stress exposure alters hypothalamic feeding neuropeptides. Feeding is regulated by orexigenic
and anorexigenic hypothalamic neuropeptides, and is also inuenced by HPA axis activity and circulating glucocorticoids (Krysiak
et al., 1999; Maniam and Morris, 2012). Stress has stimulatory
effects on hypothalamic NPY expression (Dean and White, 1990;
Goto et al., 2006; Sato et al., 2005). We recently proposed that early
life stress exposure may alter hypothalamic feeding neuropeptide
expression which may partly mediate increased food intake and
adiposity in later life (Maniam et al., 2014).
It is likely that other factors such as 11-hydroxysteroid dehydrogenase type I (11-HSD1) also play a role in mediating the
metabolic effects of early life stress (Maniam et al., 2014). In addition to circulating cortisol, tissue-specic glucocorticoids play an
important role in regulating metabolic function (Morgan et al.,
2014). Intracellular levels of glucocorticoids are tightly regulated
by 11-HSD1, which converts inactive glucocorticoid to active glucocorticoid in insulin sensitive tissues such as liver, adipose tissue
and skeletal muscle (Tomlinson et al., 2004). Chronically increased
glucocorticoid action as a result of elevated 11-HSD1 activity is
associated with obesity, insulin resistance, type 2 diabetes mellitus (Morton et al., 2004; Rask et al., 2001; Tomlinson et al., 2008).
Increased 11-HSD1 mRNA expression was observed in visceral
and subcutaneous fat of obese versus lean women and men (Alberti
et al., 2007; Paulsen et al., 2007). Taken together, these studies suggest a role of increased glucocorticoid availability, particularly in
adipose tissue, in promoting obesity.
3.2. Rodent model of early life stressmaternal separation
Rodent models have shown that early life stress induced by
maternal separation may also affect feeding behavior and inuence metabolic disease risk. In our hands rats appear to consume
similar amounts of energy, independent of diet, following early
life stress induced by maternal separation, but those consuming
a high fat/high sugar diet had increased circulating insulin relative
to non-stressed controls, suggesting long term metabolic consequences of early stress (Maniam et al., 2014; Maniam and Morris,
2010a,b). Others have reported changes in food intake following
maternal separation that were only apparent in the presence of a
stressor in later life, such as isolation stress or restricted feeding.
For example, post-weaning isolation following maternal separation
resulted in weight gain and increased food intake (Ryu et al., 2008).
In an earlier study, maternal separation was associated with greater
rebound hyperphagia following restricted feeding (2 h daily for a
period of 6 days) in female but not male rats (White et al., 1990).
Stress during pregnancy was also shown to affect metabolic function of offspring. Exposure to chronic mild stress during the 3rd
week of gestation increased fat mass in those offspring consuming
high fat/high sucrose diet, suggesting that maternal stress during
development enhances the predisposition to obesity induced by an
unhealthy diet (Paternain et al., 2013). Further work is required to

determine how exposure to adverse early life events may impose


metabolic decits on offspring.
In summary, stress in adulthood may lead to changes in feeding
behavior that increase the risk of metabolic disease, and emerging evidence points to impacts of early life experience, induced by
altered nutrition, or stress exposure in utero or early in life, on offspring food consumption or metabolism, that may increase the risk
of developing obesity in later years.
4. Effects of high energy diets on cognition
Cognitive impairments are one of the many adverse health
effects of the chronic consumption of high energy diets (Alencar
et al., 2010; Lu et al., 2012). However, an important new challenge
has emerged; these cognitive decits can occur rapidly and in some
cases, independently of the metabolic and cardiovascular disorders
associated with obesity.
A number of cross-sectional, prospective and longitudinal studies have found associations between diets rich in saturated fat
and/or sugar with cognitive decits after adjusting for demographic
and vascular factors. Higher saturated fat intake in mid and later life
has been associated with worse global cognitive function, impairments in prospective memory, memory retrieval and exibility,
as well as an increased vulnerability to both normal age related
cognitive decline and neurological diseases including dementia
and Alzheimers disease (Eskelinen et al., 2008; Grant et al., 2002;
Solfrizzi et al., 2010). Likewise, people whose diet contains excessive amounts of rened sugar are more likely to have lower overall
cognition in middle and older age (Ye et al., 2011). Roberts et al.
(2012) found that the risk of mild cognitive impairment or dementia was increased in elderly subjects who derived a high percentage
of their energy from carbohydrates, but was reduced in those who
derived a high percentage of energy from fat and protein. The effects
of diet on cognition are not conned to adults. Abargouei et al.
(2012) reported that higher sugar intake in school-aged children
was inversely related to non-verbal intelligence including spatial
memory as measured by Ravens Progressive Matrices
4.1. Evidence for effects of high energy diets on human cognition
Only a handful of studies have experimentally controlled their
participants diet and measured the effects on cognition. As accurate food consumption data is notoriously difcult to obtain outside
an experimental context, such studies are important as they allow
causal inferences to be drawn. In healthy, young adult males, a 5 day
high fat diet (75% of energy) was sufcient to impair attention and
speed of retrieval, and to depress mood, as measured by the Cognitive Drug Research assessment battery (Holloway et al., 2011).
Similarly, in sedentary male subjects (2545 years, <2 h/week physical exertion), 7 days of a high fat diet (74% of energy) decreased
reaction times and attention (Edwards et al., 2011). Moreover, eating a single high glycemic meal before testing impaired memory
performance, but increased vigilance, in normal weight undergraduate women (Nabb and Benton, 2006), school aged children (Micha
et al., 2011) and in adults with well-controlled type 2 diabetes
(Papanikolaou et al., 2006). A longer term study in children, however, found no behavioral or cognitive effects of 3 weeks of sucrose,
saccharin or aspartame sweetened diets, regardless of whether the
children were reported to respond adversely or normally to sugar
by their parents (Wolraich et al., 1994).
A different experimental approach was taken in a study by
Francis and Stevenson (2011). In Experiment 1, participants completed neuropsychological tests and questionnaires that assessed
their saturated fat and rened sugar intake. Food, not BMI or
general cognitive function, was found to be the best predictor

M.J. Morris et al. / Neuroscience and Biobehavioral Reviews 58 (2015) 3645

Fig. 1. High fat diet increases oxidative stress (OS) in the hippocampus, contributing
to decreased levels of neurotrophins such as BDNF, and downstream effects, including reduced phosphorylation of synapsin 1, growth-associated protein (GAP)-43 and
(cAMP response element-binding protein) CREB.
Taken from Wu et al. (2004) with permission.

of subsequent performance in the hippocampal-dependent verbal paired associate task and the logical memory subtests of the
Wechsler memory scale revised. Experiment 2 then compared a
subset of these participants, matched on factors such as BMI and
age, but who reported the highest and lowest intakes of saturated
fats and rened sugars. Individuals with higher saturated fat and
rened sugar consumption performed worse on the hippocampaldependent memory test (visual reproduction, logical memory, and
verbal paired associates) and were less accurate at recalling what
they had previously eaten in an experimental snack meal 60 min
prior than those who consumed low amounts of fat and sugar.
Importantly, these results were obtained in a young population
with a mainly BMI healthy range, no comorbid medical conditions,
such as diabetes or cardiovascular disease, and who did not differ on measures of general cognitive function such as attention
and concentration (digit span subtest of the Wechsler adult intelligence scale fourth edition) and general intelligence (National
Adult Reading Test). These experimental results in humans therefore suggest that the effects of fat and sugar on memory may occur
independently of any effects on body weight or general health.
4.2. Effects of high energy diets on learning and memory in
rodents
Studies with rodents have conrmed that the long-term intake
of high fat/high sugar diets impair cognition. These impairments
are especially evident in tasks which require the hippocampus and
surrounding cortices, such as the Morris water maze where rodents
must use spatial relations among cues to navigate to a hidden platform (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
maze (Murray et al., 2009) and the four arm maze (ValladolidAcebes et al., 2011), as well as hippocampal independent operant
lever pressing tasks (Farr et al., 2008) and conditioned discrimination (Greenwood and Winocur, 1990, 1996, 2005; Winocur and
Greenwood, 1999, 2005). Some of these studies have also identied
changes in the hippocampus which may mediate the link between
obesity and cognitive decits including oxidative stress, neuroinammation, as well as decreased levels of neurotrophic factors
(Fig. 1).
There is also emerging evidence that high energy diets can
impair memory in advance of substantial increases in body weight.

41

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 decits. Body weight differences had not
emerged after only 5 days of diet exposure when the impairments were rst evident and therefore, obesity was not a causal
factor. Hippocampal inammation (TNF- and IL-1 mRNA) and
oxidative stress (NRF-1 mRNA) markers were signicantly 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 recognition task (see Fig. 2). Hippocampal neurotrophic factors such as
brain-derived neurotrophic factor (BDNF) were not affected by the
month long exposure to these diets (Beilharz et al., 2014). Davidson
et al. (2013) demonstrated that rodents appear to recover from the
acute effects of western diet on cognition, and that high ketone
bodies maybe protective (Davidson et al., 2013). Interestingly, a
western diet was also shown to impact on hypothalamic inammation, eliciting early increases in inammatory markers in the
mediobasal hypothalamus, which subsided before returning after
one months exposure (Thaler et al., 2012). While several studies now indicate that cognitive decits may precede the onset of
obesity, identication of the relative impact and interrelationships
between adiposity, body weight and glucose regulation requires
more investigation. Taken together these results show that shortterm exposure to diets high in fat and sugar, or just high in
sugar, impairs aspects of cognition, suggesting that the modern
diet may rapidly produce impairments in people, including children
(Davidson et al., 2005).
These results are important because the hippocampus is not
just involved in learning and memory, but also in the control of
eating. The hippocampus contains receptors for hormones and adiposity signals, such as ghrelin and leptin, and hippocampal neurons
receive input from multiple brain regions critical for food intake.
Previous studies have shown that damage to the hippocampus
can increase food intake and result in body weight gain (Davidson
et al., 2009). Moreover, reversible inactivation of the hippocampus results in decreased latency in meal onset, increased meal size
and a disrupted relationship between meal size and the interval
to the next meal (Henderson et al., 2013). Given that overeating can impair hippocampal function, excessive food intake may
contribute to body weight gain by interfering with hippocampaldependent higher-order learning and memory processes. These
processes include episodic memory, for example, remembering
where, when and what we have previously eaten. This may create a
vicious cycle with high energy diets causing hippocampal dysfunction which leads to a further overeating of the foods that caused
the dysfunction in the rst place (Davidson et al., 2005, 2007).

5. Summary and future directions


Unless the current obesity trend is halted, the burden of chronic
disease in future generations will lead to a crisis in health and
economic systems around the world. It is of vital importance to
understand how the modern diet, the foods that we manufacture, plus the prevailing stressful environment inuence individual
food choices, potentially driving overconsumption of energy. The
omnipresence of cues promoting intake of palatable food, coupled
with the rewarding aspects of these appealing, heavily processed
foods may increase the motivation to eat to excess. Stress also
affects food choice. People may increase the amount of high fat/high

42

M.J. Morris et al. / Neuroscience and Biobehavioral Reviews 58 (2015) 3645

Fig. 2. Relationship between hippocampal TNF- mRNA expression and exploration ratios on the object (left) and place (right) recognition tasks after 2021 days on the
regular (open circle), cafeteria with sugar (closed circle), regular with sugar (open triangle) and cafeteria alone (solid square) diets. n = 58 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


a risk factor for the development of obesity in vulnerable individuals. Emerging evidence linking severe early life stress and
later metabolic disorders needs additional work to identify the
mechanisms underlying this association. Animal and human data
indicate that high fat/high sugar foods can also impair some forms
of hippocampal-dependent memories, which may further impair
appetite control by undermining the processes involved in seeking
and consuming food. Further research is also necessary to determine the mechanisms through which obesity and its comorbidities
may exacerbate cognitive impairments, particularly with aging,
where optimal nutrition is critical to preserve cognitive function.
Understanding the mechanisms underpinning the impact of other
preventive/supportive strategies such as increasing physical activity, should also be a priority. Such information is vital to provide
important insights into the drivers of obesity and to enable novel
interventions to ameliorate the epidemic.
In the 21st century, obesity should be viewed not only as a
metabolic disorder, but as a multifactorial disease. Thus, strategies that focus on modulation of the reward system, reduction of
stress reactivity and reverse compromised hippocampal function
may be more effective than the currently available pharmacological approaches typically targeting appetite suppression. Overall,
understanding the factors that increase the vulnerability of individuals to develop obesity is essential in developing more effective
treatments. Of course, providing education and support to individuals and populations in strategies for promoting healthy food
intake, and appropriate physical activity, are also of critical importance in the prevention of obesity.
Acknowledgement
This work was supported by NHMRC Project grant 1023073
awarded to MJM and RFW.
References
Abargouei, A.S., Kalantari, N., Omidvar, N., Rashidkhani, B., Rad, A.H., Ebrahimi, A.A.,
Khosravi-Boroujeni, H., Esmaillzadeh, A., 2012. Rened carbohydrate intake in
relation to non-verbal intelligence among Tehrani schoolchildren. Public Health
Nutr. 15, 19251931.
Adam, T.C., Epel, E.S., 2007. Stress, eating and the reward system. Physiol. Behav. 91,
449458.
Aggleton, J.P., Brown, M.W., 2005. Contrasting hippocampal and perirhinal cortex
function using immediate early gene imaging. Q. J. Exp. Psychol. B 58, 218233.

Ahmed, S.H., Koob, G.F., 2005. Transition to drug addiction: a negative reinforcement
model based on an allostatic decrease in reward function. Psychopharmacology
(Berl.) 180, 473490.
Alberti, L., Girola, A., Gilardini, L., Conti, A., Cattaldo, S., Micheletto, G., Invitti, C.,
2007. Type 2 diabetes and metabolic syndrome are associated with increased
expression of 11beta-hydroxysteroid dehydrogenase 1 in obese subjects. Int. J.
Obes. (Lond.) 31, 18261831.
Alencar, R.C., Cobas, R.A., Gomes, M.B., 2010. Assessment of cognitive status in
patients with type 2 diabetes through the mini-mental status examination: a
cross-sectional study. Diabetol. Metab. Syndr. 2, 10.
Alsio, J., Olszewski, P.K., Norback, A.H., Gunnarsson, Z.E., Levine, A.S., Pickering,
C., Schioth, H.B., 2010. Dopamine D1 receptor gene expression decreases in
the nucleus accumbens upon long-term exposure to palatable food and differs depending on diet-induced obesity phenotype in rats. Neuroscience 171,
779787.
Avena, N.M., Bocarsly, M.E., Rada, P., Kim, A., Hoebel, B.G., 2008. After daily bingeing on a sucrose solution, food deprivation induces anxiety and accumbens
dopamine/acetylcholine imbalance. Physiol. Behav. 94, 309315.
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.
Physiol. Behav. 84, 359362.
Avena, N.M., Rada, P., Hoebel, B.G., 2006. Sugar bingeing in rats. Curr. Protoc. Neurosci. 36, 23, C:29.23C.2129.23C.26.
Badland, H., Schoeld, G., 2006. Understanding the relationship between town size
and physical activity levels: a population study. Health Place 12, 538546.
Barker, D.J., 1998. In utero programming of chronic disease. Clin. Sci. (Lond.) 95,
115128.
Barrington, W.E., Beresford, S.A., McGregor, B.A., White, E., 2014. Perceived stress
and eating behaviors by sex, obesity status, and stress vulnerability: ndings from the vitamins and lifestyle (VITAL) study. J. Acad. Nutr. Diet 14,
17911799.
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, 134141.
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
behaviour, energy balance, autonomic outow and neuroendocrine responses
during chronic stress. J. Neuroendocrinol. 14, 330342.
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
response to acute stress. J. Neurosci. 20, 67076713.
Bello, N.T., Hajnal, A., 2010. Dopamine and binge eating behaviors. Pharmacol.
Biochem. Behav. 97, 2533.
Bernardi, J.R., Ferreira, C.F., Senter, G., Krolow, R., de Aguiar, B.W., Portella, A.K.,
Kauer-Santanna, M., Kapczinski, F., Dalmaz, C., Goldani, M.Z., Silveira, P.P., 2013.
Early life stress interacts with the diet deciency of omega-3 fatty acids during
the life course increasing the metabolic vulnerability in adult rats. PLOS ONE 8,
e62031.
Bhatnagar, S., Bell, M.E., Liang, J., Soriano, L., Nagy, T.R., Dallman, M.F., 2000.
Corticosterone facilitates saccharin intake in adrenalectomized rats: does corticosterone increase stimulus salience? J. Neuroendocrinol. 12, 453460.
Bjorntorp, P., 2001. Do stress reactions cause abdominal obesity and comorbidities?
Obes. Rev. 2, 7386.
Bocarsly, M.E., Hoebel, B.G., Paredes, D., von Loga, I., Murray, S.M., Wang, M., Arolfo,
M.P., Yao, L., Diamond, I., Avena, N.M., 2014. GS 455534 selectively suppresses
binge eating of palatable food and attenuates dopamine release in the accumbens of sugar-bingeing rats. Behav. Pharmacol. 25, 147157.

M.J. Morris et al. / Neuroscience and Biobehavioral Reviews 58 (2015) 3645


Bragulat, V., Dzemidzic, M., Bruno, C., Cox, C.A., Talavage, T., Considine, R.V., Kareken,
D.A., 2010. Food-related odor probes of brain reward circuits during hunger: a
pilot FMRI study. Obesity (Silver Spring) 18, 15661571.
Bruce-Keller, A.J., Keller, J.N., Morrison, C.D., 2009. Obesity and vulnerability of the
CNS. Biochim. Biophys. Acta 1792, 395400.
Burger, K.S., Stice, E., 2011. Variability in reward responsivity and obesity: evidence
from brain imaging studies. Curr. Drug Abuse Rev. 4, 182189.
Carroll, M.E., Lac, S.T., 1998. Dietary additives and the acquisition of cocaine selfadministration in rats. Psychopharmacology (Berl.) 137, 8189.
Castellanos, E.H., Charboneau, E., Dietrich, M.S., Park, S., Bradley, B.P., Mogg,
K., Cowan, R.L., 2009. Obese adults have visual attention bias for food cue
images: evidence for altered reward system function. Int. J. Obes. (Lond.) 33,
10631073.
Chaput, J.P., Perusse, L., Despres, J.P., Tremblay, A., Bouchard, C., 2014. Findings from
the Quebec family study on the etiology of obesity: genetics and environmental
highlights. Curr. Obes. Rep. 3, 5466.
Cohen, R.A., Grieve, S., Hoth, K.F., Paul, R.H., Sweet, L., Tate, D., Gunstad, J., Stroud, L.,
McCaffery, J., Hitsman, B., Niaura, R., Clark, C.R., McFarlane, A., Bryant, R., Gordon,
E., Williams, L.M., 2006. Early life stress and morphometry of the adult anterior
cingulate cortex and caudate nuclei. Biol. Psychiatry 59, 975982.
Cook, T., Rutishauser, I., Seelig, M., 2001. Comparable Measurements from the 1983,
1985 and 1995 National Nutrition Surveys. Australian Food and Nutrition Monitoring Unit, Canberra.
DArgenio, A., Mazzi, C., Pecchioli, L., Di Lorenzo, G., Siracusano, A., Troisi, A., 2009.
Early trauma and adult obesity: is psychological dysfunction the mediating
mechanism? Physiol. Behav. 98, 543546.
Dallman, M.F., Akana, S.F., Strack, A.M., Hanson, E.S., Sebastian, R.J., 1995. The neural network that regulates energy balance is responsive to glucocorticoids and
insulin and also regulates HPA axis responsivity at a site proximal to CRF neurons. Ann. N. Y. Acad. Sci. 771, 730742.
Dallman, M.F., Pecoraro, N.C., La Fleur, S.E., Warne, J.P., Ginsberg, A.B., Akana, S.F.,
Laugero, K.C., Houshyar, H., Strack, A.M., Bhatnagar, S., Bell, M.E., 2006. Glucocorticoids, chronic stress, and obesity. Prog. Brain Res. 153, 75105.
Danese, A., Moftt, T.E., Harrington, H., Milne, B.J., Polanczyk, G., Pariante, C.M., Poulton, R., Caspi, A., 2009. Adverse childhood experiences and adult risk factors for
age-related disease: depression, inammation, and clustering of metabolic risk
markers. Arch. Pediatr. Adolesc. Med. 163, 11351143.
Davids, S., Lauffer, H., Thoms, K., Jagdhuhn, M., Hirschfeld, H., Domin, M., Hamm,
A., Lotze, M., 2010. Increased dorsolateral prefrontal cortex activation in obese
children during observation of food stimuli. Int. J. Obes. (Lond.) 34, 94104.
Davidson, T.L., Chan, K., Jarrard, L.E., Kanoski, S.E., Clegg, D.J., Benoit, S.C., 2009. Contributions of the hippocampus and medial prefrontal cortex to energy and body
weight regulation. Hippocampus 19, 235252.
Davidson, T.L., Hargrave, S.L., Swithers, S.E., Sample, C.H., Fu, X., Kinzig, K.P., Zheng,
W., 2013. Inter-relationships among diet, obesity and hippocampal-dependent
cognitive function. Neuroscience 253, 110122.
Davidson, T.L., Kanoski, S.E., Schier, L.A., Clegg, D.J., Benoit, S.C., 2007. A potential role
for the hippocampus in energy intake and body weight regulation. Curr. Opin.
Pharmacol. 7, 613616.
Davidson, T.L., Kanoski, S.E., Walls, E.K., Jarrard, L.E., 2005. Memory inhibition and
energy regulation. Physiol. Behav. 86, 731746.
Dean, R.G., White, B.D., 1990. Neuropeptide Y expression in rat brain: effects of
adrenalectomy. Neurosci. Lett. 114, 339344.
Demos, K.E., Heatherton, T.F., Kelley, W.M., 2012. Individual differences in nucleus
accumbens activity to food and sexual images predict weight gain and sexual
behavior. J. Neurosci. 32, 55495552.
Di Chiara, G., 1998. A motivational learning hypothesis of the role of mesolimbic
dopamine in compulsive drug use. J. Psychopharmacol. 12, 5467.
Di Chiara, G., Tanda, G., 1997. Blunting of reactivity of dopamine transmission to
palatable food: a biochemical marker of anhedonia in the CMS model? Psychopharmacology (Berl.) 134, 351353.
Edwards, L.M., Murray, A.J., Holloway, C.J., Carter, E.E., Kemp, G.J., Codreanu, I.,
Brooker, H., Tyler, D.J., Robbins, P.A., Clarke, K., 2011. Short-term consumption of a high-fat diet impairs whole-body efciency and cognitive function in
sedentary men. FASEB J. 25, 10881096.
Eskelinen, M.H., Ngandu, T., Helkala, E.L., Tuomilehto, J., Nissinen, A., Soininen, H.,
Kivipelto, M., 2008. Fat intake at midlife and cognitive impairment later in life:
a population-based CAIDE study. Int. J. Geriatr. Psychiatry 23, 741747.
Farr, S.A., Yamada, K.A., Buttereld, D.A., Abdul, H.M., Xu, L., Miller, N.E., Banks,
W.A., Morley, J.E., 2008. Obesity and hypertriglyceridemia produce cognitive
impairment. Endocrinology 149, 26282636.
Finucane, M.M., Stevens, G.A., Cowan, M.J., Danaei, G., Lin, J.K., Paciorek, C.J., Singh,
G.M., Gutierrez, H.R., Lu, Y., Bahalim, A.N., Farzadfar, F., Riley, L.M., Ezzati, M.,
Global Burden of Metabolic Risk Factors of Chronic Diseases Collaborating, G.,
2011. National, regional, and global trends in body-mass index since 1980: systematic analysis of health examination surveys and epidemiological studies with
960 country-years and 9.1 million participants. Lancet 377, 557567.
Flegal, K.M., Graubard, B.I., Williamson, D.F., Gail, M.H., 2007. Cause-specic excess
deaths associated with underweight, overweight, and obesity. JAMA 298,
20282037.
Foster, M.T., Solomon, M.B., Huhman, K.L., Bartness, T.J., 2006. Social defeat increases
food intake, body mass, and adiposity in Syrian hamsters. Am. J. Physiol. Regul.
Integr. Comp. Physiol. 290, R1284R1293.
Francis, H.M., Stevenson, R.J., 2011. Higher reported saturated fat and rened sugar
intake is associated with reduced hippocampal-dependent memory and sensitivity to interoceptive signals. Behav. Neurosci. 125, 943955.

43

Furlong, T.M., Jayaweera, H.K., Balleine, B.W., Corbit, L.H., 2014. Binge-like consumption of a palatable food accelerates habitual control of behavior and is dependent
on activation of the dorsolateral striatum. J. Neurosci. 34, 50125022.
Gearhardt, A.N., Yokum, S., Orr, P.T., Stice, E., Corbin, W.R., Brownell, K.D., 2011.
Neural correlates of food addiction. Arch. Gen. Psychiatry 68, 808816.
Geiger, B.M., Haburcak, M., Avena, N.M., Moyer, M.C., Hoebel, B.G., Pothos, E.N.,
2009. Decits of mesolimbic dopamine neurotransmission in rat dietary obesity.
Neuroscience 159, 11931199.
Goto, M., Arima, H., Watanabe, M., Hayashi, M., Banno, R., Sato, I., Nagasaki, H.,
Oiso, Y., 2006. Ghrelin increases neuropeptide Y and agouti-related peptide
gene expression in the arcuate nucleus in rat hypothalamic organotypic cultures.
Endocrinology 147, 51025109.
Grant, W.B., Campbell, A., Itzhaki, R.F., Savory, J., 2002. The signicance of environmental factors in the etiology of Alzheimers disease. J. Alzheimers Dis. 4,
179189.
Greenwood, C.E., Winocur, G., 1990. Learning and memory impairment in rats fed a
high saturated fat diet. Behav. Neural Biol. 53, 7487.
Greenwood, C.E., Winocur, G., 1996. Cognitive impairment in rats fed high-fat diets:
a specic effect of saturated fatty-acid intake. Behav. Neurosci. 110, 451459.
Greenwood, C.E., Winocur, G., 2005. High-fat diets, insulin resistance and declining
cognitive function. Neurobiol. Aging 26 (Suppl. 1), 4245.
Groesz, L.M., McCoy, S., Carl, J., Saslow, L., Stewart, J., Adler, N., Laraia, B., Epel, E.,
2012. What is eating you? Stress and the drive to eat. Appetite 58, 717721.
Grosshans, M., Vollmert, C., Vollstadt-Klein, S., Tost, H., Leber, S., Bach, P., Buhler, M.,
von der Goltz, C., Mutschler, J., Loeber, S., Hermann, D., Wiedemann, K., MeyerLindenberg, A., Kiefer, F., 2012. Association of leptin with food cue-induced
activation in human reward pathways. Arch. Gen. Psychiatry 69, 529537.
Gunstad, J., Paul, R.H., Spitznagel, M.B., Cohen, R.A., Williams, L.M., Kohn, M., Gordon,
E., 2006. Exposure to early life trauma is associated with adult obesity. Psychiatry
Res. 142, 3137.
Hanson, E.S., Dallman, M.F., 1995. Neuropeptide Y (NPY) may integrate responses of
hypothalamic feeding systems and the hypothalamopituitaryadrenal axis. J.
Neuroendocrinol. 7, 273279.
Harris, R.B., Zhou, J., Youngblood, B.D., Rybkin, I.I., Smagin, G.N., Ryan, D.H., 1998.
Effect of repeated stress on body weight and body composition of rats fed lowand high-fat diets. Am. J. Physiol. 275, R1928R1938.
Haslam, D.W., James, W.P., 2005. Obesity. Lancet 366, 11971209.
Heim, C., Nemeroff, C.B., 1999. The impact of early adverse experiences on brain
systems involved in the pathophysiology of anxiety and affective disorders. Biol.
Psychiatry 46, 15091522.
Henderson, Y.O., Smith, G.P., Parent, M.B., 2013. Hippocampal neurons inhibit meal
onset. Hippocampus 23, 100107.
Hepworth, R., Mogg, K., Brignell, C., Bradley, B.P., 2010. Negative mood increases
selective attention to food cues and subjective appetite. Appetite 54, 134142.
Hess, M.E., Hess, S., Meyer, K.D., Verhagen, L.A., Koch, L., Bronneke, H.S., Dietrich,
M.O., Jordan, S.D., Saletore, Y., Elemento, O., Belgardt, B.F., Franz, T., Horvath, T.L.,
Ruther, U., Jaffrey, S.R., Kloppenburg, P., Bruning, J.C., 2013. The fat mass and
obesity associated gene (Fto) regulates activity of the dopaminergic midbrain
circuitry. Nat. Neurosci. 16, 10421048.
Heyward, F.D., Walton, R.G., Carle, M.S., Coleman, M.A., Garvey, W.T., Sweatt, J.D.,
2012. Adult mice maintained on a high-fat diet exhibit object location memory decits and reduced hippocampal SIRT1 gene expression. Neurobiol. Learn.
Mem. 98, 2532.
Holloway, C.J., Cochlin, L.E., Emmanuel, Y., Murray, A., Codreanu, I., Edwards, L.M.,
Szmigielski, C., Tyler, D.J., Knight, N.S., Saxby, B.K., Lambert, B., Thompson, C.,
Neubauer, S., Clarke, K., 2011. A high-fat diet impairs cardiac high-energy phosphate metabolism and cognitive function in healthy human subjects. Am. J. Clin.
Nutr. 93, 748755.
Ikemoto, S., Panksepp, J., 1999. The role of nucleus accumbens dopamine in
motivated behavior: a unifying interpretation with special reference to rewardseeking. Brain Res. Brain Res. Rev. 31, 641.
Jaaskelainen, A., Nevanpera, N., Remes, J., Rahkonen, F., Jarvelin, M.R., Laitinen, J.,
2014. Stress-related eating, obesity and associated behavioural traits in adolescents: a prospective population-based cohort study. BMC Public Health 14,
321.
James, W.P., Astrup, A., Finer, N., Hilsted, J., Kopelman, P., Rossner, S., Saris, W.H., Van
Gaal, L.F., 2000. Effect of sibutramine on weight maintenance after weight loss:
a randomised trial. STORM study group. Sibutramine trial of obesity reduction
and maintenance. Lancet 356, 21192125.
Jastreboff, A.M., Sinha, R., Lacadie, C., Small, D.M., Sherwin, R.S., Potenza, M.N., 2013.
Neural correlates of stress- and food cue-induced food craving in obesity: association with insulin levels. Diabetes Care 36, 394402.
Johnson, P.M., Kenny, P.J., 2010. Dopamine D2 receptors in addiction-like reward
dysfunction and compulsive eating in obese rats. Nat. Neurosci. 13, 635641.
Karra, E., ODaly, O.G., Choudhury, A.I., Yousseif, A., Millership, S., Neary, M.T., Scott,
W.R., Chandarana, K., Manning, S., Hess, M.E., Iwakura, H., Akamizu, T., Millet, Q.,
Gelegen, C., Drew, M.E., Rahman, S., Emmanuel, J.J., Williams, S.C., Ruther, U.U.,
Bruning, J.C., Withers, D.J., Zelaya, F.O., Batterham, R.L., 2013. A link between
FTO, ghrelin, and impaired brain food-cue responsivity. J. Clin. Invest. 123,
35393551.
Kenny, P.J., 2011. Reward mechanisms in obesity: new insights and future directions.
Neuron 69, 664679.
Kenny, P.J., Voren, G., Johnson, P.M., 2013. Dopamine D2 receptors and striatopallidal
transmission in addiction and obesity. Curr. Opin. Neurobiol. 23, 535538.
Koob, G.F., Le Moal, M., 2005. Plasticity of reward neurocircuitry and the dark side
of drug addiction. Nat. Neurosci. 8, 14421444.

44

M.J. Morris et al. / Neuroscience and Biobehavioral Reviews 58 (2015) 3645

Krysiak, R., Obuchowicz, E., Herman, Z.S., 1999. Interactions between the neuropeptide Y system and the hypothalamicpituitaryadrenal axis. Eur. J.
Endocrinol. 140, 130136.
la Fleur, S.E., Houshyar, H., Roy, M., Dallman, M.F., 2005. Choice of lard, but not total
lard calories, damps adrenocorticotropin responses to restraint. Endocrinology
146, 21932199.
la Fleur, S.E., Vanderschuren, L.J., Luijendijk, M.C., Kloeze, B.M., Tiesjema, B., Adan,
R.A., 2007. A reciprocal interaction between food-motivated behavior and dietinduced obesity. Int. J. Obes. (Lond.) 31, 12861294.
LeBlanc, K.H., Maidment, N.T., Ostlund, S.B., 2013. Repeated cocaine exposure facilitates the expression of incentive motivation and induces habitual control in rats.
PLOS ONE 8, e61355.
Lehmann, J., Feldon, J., 2000. Long-term biobehavioral effects of maternal separation
in the rat: consistent or confusing? Rev. Neurosci. 11, 383408.
Lippmann, M., Bress, A., Nemeroff, C.B., Plotsky, P.M., Monteggia, L.M., 2007. Longterm behavioural and molecular alterations associated with maternal separation
in rats. Eur. J. Neurosci. 25, 30913098.
Loeber, S., Grosshans, M., Korucuoglu, O., Vollmert, C., Vollstadt-Klein, S., Schneider,
S., Wiers, R.W., Mann, K., Kiefer, F., 2012. Impairment of inhibitory control in
response to food-associated cues and attentional bias of obese participants and
normal-weight controls. Int. J. Obes. (Lond.) 36, 13341339.
Lu, Y., Lu, J., Wang, S., Li, C., Liu, L., Zheng, R., Tian, H., Wang, X., Yang, L., Zhang, Y.,
Pan, C., 2012. Cognitive function with glucose tolerance status and obesity in
Chinese middle-aged and aged adults. Aging Ment. Health 16, 911914.
Lumeng, J.C., Wendorf, K., Pesch, M.H., Appugliese, D.P., Kaciroti, N., Corwyn, R.F.,
Bradley, R.H., 2013. Overweight adolescents and life events in childhood. Pediatrics 132, e1506e1512.
Lutter, M., Nestler, E.J., 2009. Homeostatic and hedonic signals interact in the regulation of food intake. J. Nutr. 139, 629632.
Macht, M., Mueller, J., 2007. Immediate effects of chocolate on experimentally
induced mood states. Appetite 49, 667674.
Macri, S., Chiarotti, F., Wurbel, H., 2008. Maternal separation and maternal care act
independently on the development of HPA responses in male rats. Behav. Brain
Res. 191, 227234.
Maniam, J., Antoniadis, C., Morris, M.J., 2014. Early-life stress, HPA axis adaptation, and mechanisms contributing to later health outcomes. Front. Endocrinol.
(Lausanne) 5, 73.
Maniam, J., Morris, M.J., 2010a. Long-term postpartum anxiety and depression-like
behavior in mother rats subjected to maternal separation are ameliorated by
palatable high fat diet. Behav. Brain Res. 208, 7279.
Maniam, J., Morris, M.J., 2010b. Palatable cafeteria diet ameliorates anxiety and
depression-like symptoms following an adverse early environment. Psychoneuroendocrinology 35, 717728.
Maniam, J., Morris, M.J., 2012. The link between stress and feeding behaviour. Neuropharmacology 63, 97110.
Marti, O., Marti, J., Armario, A., 1994. Effects of chronic stress on food intake in rats:
inuence of stressor intensity and duration of daily exposure. Physiol. Behav.
55, 747753.
Martire, S.I., Maniam, J., South, T., Holmes, N., Westbrook, R.F., Morris, M.J., 2014.
Extended exposure to a palatable cafeteria diet alters gene expression in
brain regions implicated in reward, and withdrawal from this diet alters gene
expression in brain regions associated with stress. Behav. Brain Res. 265,
132141.
McIntosh, J., Anisman, H., Merali, Z., 1999. Short- and long-periods of neonatal maternal separation differentially affect anxiety and feeding in adult rats:
gender-dependent effects. Brain Res. Dev. Brain Res. 113, 97106.
McNay, E.C., Ong, C.T., McCrimmon, R.J., Cresswell, J., Bogan, J.S., Sherwin, R.S., 2010.
Hippocampal memory processes are modulated by insulin and high-fat-induced
insulin resistance. Neurobiol. Learn. Mem. 93, 546553.
Mei, Z., Grummer-Strawn, L.M., Pietrobelli, A., Goulding, A., Goran, M.I., Dietz, W.H.,
2002. Validity of body mass index compared with other body-composition
screening indexes for the assessment of body fatness in children and adolescents. Am. J. Clin. Nutr. 75, 978985.
Meule, A., Lutz, A., Vogele, C., Kubler, A., 2012. Food cravings discriminate differentially between successful and unsuccessful dieters and non-dieters. Validation
of the Food Cravings Questionnaires in German. Appetite 58, 8897.
Meule, A., Lutz, A.P., Vogele, C., Kubler, A., 2014. Impulsive reactions to food-cues
predict subsequent food craving. Eat. Behav. 15, 99105.
Meye, F.J., Adan, R.A., 2014. Feelings about food: the ventral tegmental area in food
reward and emotional eating. Trends Pharmacol. Sci. 35, 3140.
Micha, R., Rogers, P.J., Nelson, M., 2011. Glycaemic index and glycaemic load of
breakfast predict cognitive function and mood in school children: a randomised
controlled trial. Br. J. Nutr. 106, 15521561.
Moore, C.J., Cunningham, S.A., 2012. Social position, psychological stress, and obesity: a systematic review. J. Acad. Nutr. Diet. 112, 518526.
Morgan, S.A., McCabe, E.L., Gathercole, L.L., Hassan-Smith, Z.K., Larner, D.P., Bujalska,
I.J., Stewart, P.M., Tomlinson, J.W., Lavery, G.G., 2014. 11-HSD1 is the major
regulator of the tissue-specic effects of circulating glucocorticoid excess. Proc.
Natl. Acad. Sci. 111, E2482E2491.
Morton, N.M., Paterson, J.M., Masuzaki, H., Holmes, M.C., Staels, B., Fievet, C., Walker,
B.R., Flier, J.S., Mullins, J.J., Seckl, J.R., 2004. Novel adipose tissue-mediated resistance to diet-induced visceral obesity in 11 beta-hydroxysteroid dehydrogenase
type 1-decient mice. Diabetes 53, 931938.
Murray, A.J., Knight, N.S., Cochlin, L.E., McAleese, S., Deacon, R.M., Rawlins, J.N.,
Clarke, K., 2009. Deterioration of physical performance and cognitive function
in rats with short-term high-fat feeding. FASEB J. 23, 43534360.

Nabb, S., Benton, D., 2006. The inuence on cognition of the interaction between the
macro-nutrient content of breakfast and glucose tolerance. Physiol. Behav. 87,
1623.
Nelson, A., Killcross, S., 2006. Amphetamine exposure enhances habit formation. J.
Neurosci. 26, 38053812.
Nieuwenhuizen, A.G., Rutters, F., 2008. The hypothalamicpituitaryadrenal-axis in
the regulation of energy balance. Physiol. Behav. 94, 169177.
Noll, J.G., Zeller, M.H., Trickett, P.K., Putnam, F.W., 2007. Obesity risk for female
victims of childhood sexual abuse: a prospective study. Pediatrics 120, e61e67.
Ogden, C.L., Yanovski, S.Z., Carroll, M.D., Flegal, K.M., 2007. The epidemiology of
obesity. Gastroenterology 132, 20872102.
Papanikolaou, Y., Palmer, H., Binns, M.A., Jenkins, D.J., Greenwood, C.E., 2006. Better
cognitive performance following a low-glycaemic-index compared with a highglycaemic-index carbohydrate meal in adults with type 2 diabetes. Diabetologia
49, 855862.
Paternain, L., de la Garza, A.L., Batlle, M.A., Milagro, F.I., Martinez, J.A., Campion, J.,
2013. Prenatal stress increases the obesogenic effects of a high-fat-sucrose diet
in adult rats in a sex-specic manner. Stress 16, 220232.
Paulsen, S.K., Pedersen, S.B., Fisker, S., Richelsen, B., 2007. 11Beta-HSD type 1 expression in human adipose tissue: impact of gender, obesity, and fat localization.
Obesity (Silver Spring) 15, 19541960.
Pecoraro, N., Reyes, F., Gomez, F., Bhargava, A., Dallman, M.F., 2004. Chronic stress
promotes palatable feeding, which reduces signs of stress: feedforward and
feedback effects of chronic stress. Endocrinology 145, 37543762.
Pelchat, M.L., Johnson, A., Chan, R., Valdez, J., Ragland, J.D., 2004. Images of desire:
food-craving activation during fMRI. Neuroimage 23, 14861493.
Petrovich, G.D., Hobin, M.P., Reppucci, C.J., 2012. Selective Fos induction in hypothalamic orexin/hypocretin, but not melanin-concentrating hormone neurons,
by a learned food-cue that stimulates feeding in sated rats. Neuroscience 224,
7080.
Petrovich, G.D., Holland, P.C., Gallagher, M., 2005. Amygdalar and prefrontal pathways to the lateral hypothalamus are activated by a learned cue that stimulates
eating. J. Neurosci. 25, 82958302.
Petrovich, G.D., Ross, C.A., Gallagher, M., Holland, P.C., 2007. Learned contextual cue
potentiates eating in rats. Physiol. Behav. 90, 362367.
Pickering, C., Alsio, J., Hulting, A.L., Schioth, H.B., 2009. Withdrawal from free-choice
high-fat high-sugar diet induces craving only in obesity-prone animals. Psychopharmacology (Berl.) 204, 431443.
Pistell, P.J., Morrison, C.D., Gupta, S., Knight, A.G., Keller, J.N., Ingram, D.K., BruceKeller, A.J., 2010. Cognitive impairment following high fat diet consumption is
associated with brain inammation. J. Neuroimmunol. 219, 2532.
Poore, K.R., Boullin, J.P., Cleal, J.K., Newman, J.P., Noakes, D.E., Hanson, M.A., Green,
L.R., 2010. Sex- and age-specic effects of nutrition in early gestation and
early postnatal life on hypothalamo-pituitaryadrenal axis and sympathoadrenal function in adult sheep. J. Physiol. 588, 22192237.
Putnam, J.J., Allshouse, J.E., 1999. Food Consumption, Prices, and Expenditures,
197097. E.R.S. Food and Consumers Economics Division, U.S. Department of
Agriculture, Washington, DC.
Rada, P., Avena, N.M., Hoebel, B.G., 2005. Daily bingeing on sugar repeatedly releases
dopamine in the accumbens shell. Neuroscience 134, 737744.
Ramasubramanian, L., Lane, S., Rahman, A., 2013. The association between maternal serious psychological distress and child obesity at 3 years: a cross-sectional
analysis of the UK Millennium Cohort Data. Child Care Health Dev. 39, 134140.
Rangan, A.M., Randall, D., Hector, D.J., Gill, T.P., Webb, K.L., 2008. Consumption of
extra foods by Australian children: types, quantities and contribution to energy
and nutrient intakes. Eur. J. Clin. Nutr. 62, 356364.
Rangan, A.M., Schindeler, S., Hector, D.J., Gill, T.P., Webb, K.L., 2009. Consumption of
extra foods by Australian adults: types, quantities and contribution to energy
and nutrient intakes. Eur. J. Clin. Nutr. 63, 865871.
Rask, E., Olsson, T., Soderberg, S., Andrew, R., Livingstone, D.E., Johnson, O., Walker,
B.R., 2001. Tissue-specic dysregulation of cortisol metabolism in human obesity. J. Clin. Endocrinol. Metab. 86, 14181421.
Reichelt, A.C., Morris, M.J., Westbrook, R.F., 2014. Cafeteria diet impairs expression of
sensory-specic satiety and stimulus-outcome learning. Front. Psychol. 5, 852.
Reppucci, C.J., Petrovich, G.D., 2012. Learned food-cue stimulates persistent feeding
in sated rats. Appetite 59, 437447.
Roberts, R.O., Roberts, L.A., Geda, Y.E., Cha, R.H., Pankratz, V.S., OConnor, H.M., Knopman, D.S., Petersen, R.C., 2012. Relative intake of macronutrients impacts risk of
mild cognitive impairment or dementia. J. Alzheimers Dis. 32, 329339.
Robinson, T.E., Berridge, K.C., 1993. The neural basis of drug craving: an incentivesensitization theory of addiction. Brain Res. Brain Res. Rev. 18, 247291.
Rothemund, Y., Preuschhof, C., Bohner, G., Bauknecht, H.C., Klingebiel, R., Flor, H.,
Klapp, B.F., 2007. Differential activation of the dorsal striatum by high-calorie
visual food stimuli in obese individuals. Neuroimage 37, 410421.
Ryu, V., Lee, J.H., Yoo, S.B., Gu, X.F., Moon, Y.W., Jahng, J.W., 2008. Sustained hyperphagia in adolescent rats that experienced neonatal maternal separation. Int. J.
Obes. (Lond.) 32, 13551362.
Saper, C.B., Chou, T.C., Elmquist, J.K., 2002. The need to feed: homeostatic and hedonic
control of eating. Neuron 36, 199211.
Sato, I., Arima, H., Ozaki, N., Watanabe, M., Goto, M., Hayashi, M., Banno, R., Nagasaki,
H., Oiso, Y., 2005. Insulin inhibits neuropeptide Y gene expression in the arcuate
nucleus through GABAergic systems. J. Neurosci. 25, 86578664.
Savontaus, E., Conwell, I.M., Wardlaw, S.L., 2002. Effects of adrenalectomy on AGRP,
POMC, NPY and CART gene expression in the basal hypothalamus of fed and
fasted rats. Brain Res. 958, 130138.

M.J. Morris et al. / Neuroscience and Biobehavioral Reviews 58 (2015) 3645


Schmidt, M.V., Liebl, C., Sterlemann, V., Ganea, K., Hartmann, J., Harbich,
D., Alam, S., Muller, M.B., 2008. Neuropeptide Y mediates the initial
hypothalamicpituitaryadrenal response to maternal separation in the neonatal mouse. J. Endocrinol. 197, 421427.
Schulz, S., Laessle, R.G., 2012. Stress-induced laboratory eating behavior in obese
women with binge eating disorder. Appetite 58, 457461.
Schur, E.A., Kleinhans, N.M., Goldberg, J., Buchwald, D., Schwartz, M.W., Maravilla,
K., 2009. Activation in brain energy regulation and reward centers by food cues
varies with choice of visual stimulus. Int. J. Obes. (Lond.) 33, 653661.
Simmons, W.K., Martin, A., Barsalou, L.W., 2005. Pictures of appetizing foods activate
gustatory cortices for taste and reward. Cereb. Cortex 15, 16021608.
Sinha, R., Jastreboff, A.M., 2013. Stress as a common risk factor for obesity and
addiction. Biol. Psychiatry 73, 827835.
Solfrizzi, V., Frisardi, V., Capurso, C., DIntrono, A., Colacicco, A.M., Vendemiale, G.,
Capurso, A., Panza, F., 2010. Dietary fatty acids in dementia and predementia syndromes: epidemiological evidence and possible underlying mechanisms. Ageing
Res. Rev. 9, 184199.
South, T., Westbrook, F., Morris, M.J., 2012. Neurological and stress related effects of
shifting obese rats from a palatable diet to chow and lean rats from chow to a
palatable diet. Physiol. Behav. 105, 10521057.
Stice, E., Yokum, S., Blum, K., Bohon, C., 2010. Weight gain is associated with reduced
striatal response to palatable food. J. Neurosci. 30, 1310513109.
Strack, A.M., Sebastian, R.J., Schwartz, M.W., Dallman, M.F., 1995. Glucocorticoids and insulin: reciprocal signals for energy balance. Am. J. Physiol. 268,
R142R149.
Swinburn, B., Sacks, G., Ravussin, E., 2009. Increased food energy supply is more
than sufcient to explain the US epidemic of obesity. Am. J. Clin. Nutr. 90,
14531456.
Swinburn, B.A., Caterson, I., Seidell, J.C., James, W.P., 2004. Diet, nutrition and the
prevention of excess weight gain and obesity. Public Health Nutr. 7, 123146.
Tang, D.W., Fellows, L.K., Small, D.M., Dagher, A., 2012. Food and drug cues activate
similar brain regions: a meta-analysis of functional MRI studies. Physiol. Behav.
106, 317324.
Thaler, J.P., Yi, C.X., Schur, E.A., Guyenet, S.J., Hwang, B.H., Dietrich, M.O., Zhao, X.,
Sarruf, D.A., Izgur, V., Maravilla, K.R., Nguyen, H.T., Fischer, J.D., Matsen, M.E.,
Wisse, B.E., Morton, G.J., Horvath, T.L., Baskin, D.G., Tschop, M.H., Schwartz, M.W.,
2012. Obesity is associated with hypothalamic injury in rodents and humans. J.
Clin. Invest. 122, 153162.
Thanos, P.K., Michaelides, M., Piyis, Y.K., Wang, G.J., Volkow, N.D., 2008. Food restriction markedly increases dopamine D2 receptor (D2R) in a rat model of obesity
as assessed with in-vivo muPET imaging ([11C] raclopride) and in-vitro ([3H]
spiperone) autoradiography. Synapse 62, 5061.
Tomiyama, A.J., Dallman, M.F., Epel, E.S., 2011. Comfort food is comforting to those
most stressed: evidence of the chronic stress response network in high stress
women. Psychoneuroendocrinology 36, 15131519.
Tomlinson, J.W., Finney, J., Gay, C., Hughes, B.A., Hughes, S.V., Stewart, P.M., 2008.
Impaired glucose tolerance and insulin resistance are associated with increased
adipose 11beta-hydroxysteroid dehydrogenase type 1 expression and elevated
hepatic 5alpha-reductase activity. Diabetes 57, 26522660.

45

Tomlinson, J.W., Walker, E.A., Bujalska, I.J., Draper, N., Lavery, G.G., Cooper, M.S.,
Hewison, M., Stewart, P.M., 2004. 11beta-hydroxysteroid dehydrogenase type
1: a tissue-specic regulator of glucocorticoid response. Endocr. Rev. 25,
831866.
Valladolid-Acebes, I., Stucchi, P., Cano, V., Fernandez-Alfonso, M.S., Merino, B., GilOrtega, M., Fole, A., Morales, L., Ruiz-Gayo, M., Del Olmo, N., 2011. High-fat diets
impair spatial learning in the radial-arm maze in mice. Neurobiol. Learn. Mem.
95, 8085.
Velders, F.P., De Wit, J.E., Jansen, P.W., Jaddoe, V.W., Hofman, A., Verhulst, F.C.,
Tiemeier, H., 2012. FTO at rs9939609, food responsiveness, emotional control
and symptoms of ADHD in preschool children. PLoS ONE 7, e49131.
Veyrat-Durebex, C., Deblon, N., Caillon, A., Andrew, R., Altirriba, J., Odermatt,
A., Rohner-Jeanrenaud, F., 2012. Central glucocorticoid administration promotes weight gain and increased 11beta-hydroxysteroid dehydrogenase type
1 expression in white adipose tissue. PLoS ONE 7, e34002.
Volkow, N.D., Wang, G.J., Fowler, J.S., Tomasi, D., Baler, R., 2012. Food and drug
reward: overlapping circuits in human obesity and addiction. Curr. Top. Behav.
Neurosci. 11, 124.
Walker, A.K., Ibia, I.E., Zigman, J.M., 2012. Disruption of cue-potentiated feeding in
mice with blocked ghrelin signaling. Physiol. Behav. 108, 3443.
Wanat, M.J., Hopf, F.W., Stuber, G.D., Phillips, P.E., Bonci, A., 2008. Corticotropinreleasing factor increases mouse ventral tegmental area dopamine neuron
ring through a protein kinase C-dependent enhancement of Ih. J. Physiol. 586,
21572170.
Weingarten, H.P., 1983. Conditioned cues elicit feeding in sated rats: a role for
learning in meal initiation. Science 220, 431433.
Wellman, P.J., Nation, J.R., Davis, K.W., 2007. Impairment of acquisition of cocaine
self-administration in rats maintained on a high-fat diet. Pharmacol. Biochem.
Behav. 88, 8993.
White, B.D., Dean, R.G., Martin, R.J., 1990. Adrenalectomy decreases neuropeptide Y
mRNA levels in the arcuate nucleus. Brain Res. Bull. 25, 711715.
World Health Organization. (2000). Obesity: preventing and managing the global
epidemic (No. 894). World Health Organization.
Winocur, G., Greenwood, C.E., 1999. The effects of high fat diets and environmental inuences on cognitive performance in rats. Behav. Brain Res. 101,
153161.
Winocur, G., Greenwood, C.E., 2005. Studies of the effects of high fat diets on cognitive function in a rat model. Neurobiol Aging 26 (Suppl. 1), 4649.
Wolraich, M.L., Lindgren, S.D., Stumbo, P.J., Stegink, L.D., Appelbaum, M.I., Kiritsy,
M.C., 1994. Effects of diets high in sucrose or aspartame on the behavior and
cognitive performance of children. N. Engl. J. Med. 330, 301307.
Wu, A., Ying, Z., Gomez-Pinilla, F., 2004. The interplay between oxidative stress and
brain-derived neurotrophic factor modulates the outcome of a saturated fat diet
on synaptic plasticity and cognition. Eur. J. Neurosci. 19, 16991707.
Ye, X., Gao, X., Scott, T., Tucker, K.L., 2011. Habitual sugar intake and cognitive function among middle-aged and older Puerto Ricans without diabetes. Br. J. Nutr.
106, 14231432.

You might also like