You are on page 1of 10

Asian Journal of Psychiatry 25 (2017) 91–100

Contents lists available at ScienceDirect

Asian Journal of Psychiatry


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

Review article

Neurobiology of eating disorders - an overview


Anand Mishra, MBBS, Junior resident Psychiatrya,* ,
Manu Anand, MBBS, Junior resident Psychiatrya ,
Shreekantiah Umesh, MD, DPM, Senior resident Psychiatryb
a
Central Institute of Psychiatry, Ranchi, Jharkhand, India
b
K.S. Mani Centre for Cognitive Neurosciences, Central Institute of Psychiatry, Ranchi, Jharkhand, India

A R T I C L E I N F O

Article history:
Received 29 January 2016
Received in revised form 3 September 2016
Accepted 9 October 2016
Available online xxx

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
2. Neurobiology of appetite . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92
2.1. Homeostatic regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93
2.2. Non homeostatic regulation . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93
3. Neurobiology of ED . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
3.1. Genetic evidences . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
3.2. Neuropsychological evidences . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
3.3. Interoceptive awareness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
3.4. Obsessisonality . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
3.5. Neuroimaging evidences . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
4. Neurotransmitters and neuropeptides . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
4.1. Serotonin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
4.2. Dopamine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
4.3. Others . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
5. Clinical perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96
6. The research domain criteria (RDoC) framework for research in ED . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96
7. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96

1. Introduction that have identifiable causes, clear symptomatology, predictable


outcomes, affect people regardless of gender ethnicity or age, and
Eating disorders (ED) are serious illnesses that usually involve respond to treatment (Kramer and Kittleson, 2005). They most
eating way too little or way too much, and can seriously jeopardize commonly occur among late adolescent and young adult women
one’s health. At one time, rarely mentioned and poorly understood, (Giordano, 2007). These may be chronic, relapsing and are often
today ED are one of the most researched and discussed illness associated with psychiatric comorbidity and medical sequelae.
throughout the world. We know now that ED are mental disorders Among the ED, anorexia nervosa (AN) and bulimia nervosa (BN) are
the most common, but DSM 5 also recognized other clinically
significant eating disturbances like binge-eating disorder (BED),
* Corresponding author. pica, Avoidant/Restrictive food intake disorder (ARFID),
E-mail address: anandmishraxxxxxxx@gmail.com (A. Mishra).

http://dx.doi.org/10.1016/j.ajp.2016.10.009
1876-2018/ã 2016 Elsevier B.V. All rights reserved.
92 A. Mishra et al. / Asian Journal of PsychiatryAJP 25 (2017) 91–100

rumination disorder and other specified eating or feeding and Piran, 2007) highlights the possible existence of differences in
disorders (OSFED) in its formal diagnostic category after merging neural processes of obesity and addiction.
feeding and eating disorders in a single chapter (American Although the development of a neurobiological model of ED is
Psychiatric Association, 2013). Despite of the immense amount still in a nascent stage, there are enough evidences to warrant an
of research the pathophysiology of ED is still poorly understood. understanding of the neurobiology as an important approach for
Though mostly attributed to psychosocial factors, there is growing further improvement in treatment of ED.
agreement on the fact that neurobiological vulnerabilities contrib-
ute to the pathogenesis of ED (Treasure and Campbell, 1994). 2. Neurobiology of appetite
Recent studies have shown that genetic factors account for
approximately 50–80% of the risk of developing eating disorders The sensation of hunger (physiological drive) is associated with
(Bulik et al., 2006) and contribute to neurobiological factors a craving for food and several other physiological effects, which
underlying ED (Kaye and Strober, 2009). The ED display a unique cause the person to seek an adequate food supply. While appetite
and complex appetitive symptomatology which is not shared by (psychological drive) is a desire for food, often of a particular type,
other psychiatric disorders which suggests that they possibly and is useful in helping the individual to choose the quality of food
reflect some aberration in the appetitive pathways (Kaye et al., he eats. If the quest for food is successful, the feeling of satiety
2011). In addition, many individuals with ED tend to express occurs (Hall, 2010). Both metabolic and non-metabolic factors are
behaviors like inhibition/disinhibition, anxiety, depression, obses- involved in the initiation and maintenance of eating behavior.
sionality, body image distortion, perfectionism and anhedonia in Particularly in humans, non-metabolic factors like cues, reward,
concert because they are likely to be encoded in limbic and cognitive and emotional factors play a major role. The brain regions
cognitive circuits known to regulate neurological processes related regulating appetite may be considered as either homeostatic or
to appetite, emotionality, and cognitive control (Kaye et al., 2011). hedonic (Gibson et al., 2010). If one has not eaten for long,
Studies on cortico-limbic neural processes in obesity (Berthoud, homeostatic processes would induce hunger perception and if one
2006), brain mechanisms in appetite (Rolls, 2006), gustatory is not hungry but presented with food stimuli like smell or sight,
processing (Small, 2006) suggest that individuals can overeat even then hunger perception is elicited concomitantly with bodily
when satiated and with replete energy stores. It has also been responses like salivation. They can be ‘intrinsic’ or ‘extrinsic’
suggested that ED and addiction share overlapping neurobiology factors as homeostatic induced hunger mainly arises from
(Volkow et al., 2012). However, contrasting symptoms such as processes within the individual, while food preferences and
underconsumption of food despite of emaciation and decreased associated rewards are learnt from prior experiences (Arana
rates of substance abuse in AN (Calero-Elvira et al., 2009; Gadalla et al., 2003). Hypothalamus (Arora, 2006) and brainstem as part of

Fig. 1. Brain regions involved in feeding behaviour. Acb, nucleus accumbens; BLA, basolateral amygdala; CEA, central nucleus of the amygdala; DMH, dorsomedial
hypothalamus; LH, lateral hypothalamus; MD, mediodorsal thalamic nucleus; NTS, nucleus of the solitary tract; PB, parabrachial nucleus; PV, paraventricular; PVN,
paraventricular nucleus of the hypothalamus; VMH, ventromedial nucleus of the hypothalamus (Kelley et al., 2005). “Reprinted from Physiology & behavior, 86(5), Kelley, A.
E., Baldo, B. A., Pratt, W. E., & Will, M. J., Corticostriatal-hypothalamic circuitry and food motivation: integration of energy, action and reward, 773–795, Copyright (2005), with
permission from Elsevier [OR APPLICABLE SOCIETY COPYRIGHT OWNER]”.
A. Mishra et al. / Asian Journal of PsychiatryAJP 25 (2017) 91–100 93

the homeostatic system regulate food intake based on caloric need


and energy balance (Berthoud and Morrison, 2008; Nisbett, 1972).
They integrate inputs from cortical regions involving reward value
of food (Kampe et al., 2009; Schwartz, 2006) indicating that
‘hedonic’ hunger may be neurally mediated (Berridge, 2009; Haase
et al., 2009; Hinton et al., 2004). The neural mechanisms in the
hedonic network may override homeostatic signals and contribute
to ED (Fig. 1).

2.1. Homeostatic regulation

The lateral nuclei of the hypothalamus perform the role of


feeding center by initiating the motor drives to search for food. The
ventromedial nuclei of the hypothalamus perform the role of
satiety center.
Other regions like paraventricular, dorsomedial and arcuate
nuclei of hypothalamus also affect the regulation of food intake in
important ways. The arcuate nuclei are the sites in the
hypothalamus where several hormones released from the gastro-
intestinal tract and adipose tissue converge to regulate food intake,
as well as energy expenditure. The hypothalamus receives neural
signals from the gastrointestinal tract that provide sensory Fig. 2. Brain network for appetitive behavior. PFC, prefrontal cortex; OFC,
orbitofrontal cortex; VTA, ventral tegmental area; DA, dopamine (Dagher, 2009).
information about the filling of stomach; chemical signals
Reprinted by permission from Macmillan Publishers Ltd: [INTERNATIONAL
signaling satiety from nutrients in the blood; signals from JOURNAL OF OBESITY] (Dagher, A. (2009). The neurobiology of appetite: hunger
gastrointestinal hormones; hormonal signals from adipose tissue; as addiction.International journal of obesity, 33, S30-S33.), copyright (2009).
and signals from the cerebral cortex (taste, smell and sight of the
food) that influence feeding behavior. The receptors for various 2001). In addition to integrating the homeostatic information, as a
neurotransmitters that modulate feeding behavior are highly group these structures are involved in learning, liking and wanting
expressed in the hypothalamic feeding and satiety centers. These component of reward in respect of food by allocating attention,
neurotransmitters are broadly categorized as (1) orexigenic effort, and motivation for it (Berridge and Robinson, 2003). They all
substances that stimulate feeding (example – Neuropeptide Y generally respond to food cues which are conditioned stimuli
(NPY), Agouti-related protein (AGRP), Melatonin concentrating predictive of reward. The cognitive control of appetite is mediated
hormone (MCH), Orexin A & B, Endorphins, Galanin (Gal), by the modulatory control of prefrontal cortex over the appetitive
glutamate & GABA, Ghrelin, Cortisol, endocannabinoids etc.) or regions.
(2) anorexigenic substances that inhibit feeding (Example – Based on neuroanatomical observations and findings (Mogen-
a-MSH, leptin, serotonin, CRH, norepinephrine, insulin, gluca- son et al., 1980; Otake and Nakamura, 2000; Usuda et al., 1998;
gon-like peptide (GLP), cholecystokinine (CCK), cocaine and Zahm, 2000) and various receptor studies in nucleus accumbens
amphetamine regulated transcript (CART), peptide YY (PYY)). (Basso and Kelley, 1999; Maldonado-Irizarry et al., 1995; Stratford
Gastrointestinal Filling, CCK, PYY, GLP, Ghrelin and various “oral and Kelley, 1997, 1999; Will et al., 2003; Zhang et al., 1998, 2003;
factors” (chewing, salivation, swallowing and tasting) are opera- Zhang and Kelley, 1997, 2000; Zheng et al., 2003) and (Berthoud
tive in short term regulation of food intake. While neuropsycho- and Morrison, 2008) suggested a role for accumbens-hypothalamic
logical studies of functions in specific areas of brain support the pathways for the cognitive and emotional brain to override
glucostatic, aminostatic and lipostatic theories in long term homeostatic regulation (Fig. 3).
regulation of food intake. Also interaction within the hypothala- Several neurotransmitters are involved in the normal feeding
mus between the temperature regulating and food intake behavior (Kalra et al., 1999). Serotonin has a hypophagic effect
regulating system has its effect on the homeostatic regulation of probably mediated by postsynaptic 5-HT2C receptors (Leibowitz
food intake. Leptin, a hormone secreted by adipocytes, stimulates and Alexander, 1998; Nonogaki et al., 1998). Also 5-HT1A and 5-
actions at multiple sites in hypothalamus that help in decreasing HT1 B receptors are considered to have opposing effects on feeding
fat storage, which also includes decreased production of orexi- behavior.
genics and increased production of anorexigenics in the hypothal- Dopamine, via its action in the nucleus accumbens, seems to be
amus. associated with the reinforcement effect in feeding (food cues) (Di
Chiara and Imperato, 1988; Wise and Rompré, 1989). Its release in
2.2. Non homeostatic regulation hypothalamus seems to be associated with the duration of meal
consumption, thus associated with meal size (Meguid et al., 2000).
In humans the meal can be started even in the absence of any There are evidences supporting a stimulatory role of GABA and
depletion signal, purely as an executive cortical decision. de Castro glutamate in several species (Ebenezer and Baldwin, 1990; Meister,
and Plunkett, 2002 showed that food intake level change 2007; Parker and Bloom, 2012; Stanley et al., 1993, 1996; Stratford
depending on external environment (palatability, dietary and Kelley, 1997; Van Den Pol, 2003). Brain noradrenaline
restraints, social facilitation etc.) (Fig. 2). alteration can increase or decrease eating (Wellman, 2000).
Four heavily interconnected structures, the amygdala/hippo- Several studies show that acetylcholine receptor ligand nicotine
campus, orbitofrontal cortex (OFC), insula, and striatum are the reduces appetite and alters feeding patterns (Grunberg et al., 1986;
main elements in the control of appetitive behavior (Dagher, Jo et al., 2005; Levin et al., 1987; Miyata et al., 1999).
2009). Especially fMRI studies have shown that anterior insula is Brain derived Neurotropic Factor (BDNF) has also been shown to
most probably the primary gustatory cortex and a portion of the influence both the homeostatic and hedonic mechanisms of
orbitofrontal cortex being the secondary gustatory cortex as both appetite. Its role in decreasing food intake, increasing energy
seem to respond to taste (Francis et al., 1999; O'Doherty et al.,
94 A. Mishra et al. / Asian Journal of PsychiatryAJP 25 (2017) 91–100

Fig. 3. Schematic diagram depicting possible interactions between “cognitive” and “emotional” brain with the “metabolic” brain (Berthoud, 2006). “Copyright (2006) Wiley.
Used with permission from (Berthoud, Homeostatic and Non-homeostatic Pathways Involved in the Control of Food Intake and Energy Balance, Obesity, The Obesity Society.".

expenditure, reducing body weight, along with increased expres- regulation as the possible candidates in the neurobiology of ED
sion in brain centers for appetite has been demonstrated in animals (Frey et al., 2000; Hebebrand et al., 1997; Hebebrand and
(Nakagawa et al., 2000; Tsuchida et al., 2001). It is also highly Remschmidt, 1995; Kaye et al., 1990, 1998a, 1998b; Koronyo-
expressed in the central reward pathways which hints at a role in Hamaoui et al., 2002). Although genome wide linkage studies have
modulation of the positive hedonic eating with respect to palatable been equivocal, still variations in the 5-HT2A receptor gene
food (Cordeira et al., 2010; Numan and Seroogy, 1999), as shown in (Gorwood et al., 2003) and the Val66Met variant in BDNF (Ribases
animal studies. et al., 2003) have good evidence (Scherag et al., 2010).
High levels of endocannabinoids (anandamide and 2-arach-
idonoylglycerol (2-AG)) and cannabinoid receptors have also been 3.2. Neuropsychological evidences
demonstrated in the limbic systems of food deprived animals
(Kirkham et al., 2002). The endocannabinoids affect feeding In a first of its kind study in which healthy men from military
behavior and the reward processing associated with it. Following services military service were starved, Keys et al. found neuro-
short term food deprivation, they regulate the actions of orexigenic psychological changes similar to ED (Keys et al., 1950). Studies have
and anorexigeic substances and by their effect on the mesolimbic also shown that patients with AN have specific thinking styles with
dopaminergic pathway, increase the level of motivation to search characteristics of seeing things in detail, perfectionism, difficulties
and consume more rewarding food items (Cota et al., 2003). The in ‘set shifting’ and weak ‘central coherence’ (having difficulty in
cannabinoid induced wanting and liking for food has been shown seeing the larger picture due to focusing on minute details)
to be mediated by increased dopaminergic activity in the nucleus (Gillberg et al., 1996; Keys et al., 1950; Tchanturia et al., 2005).
accumbens (Solinas et al., 2006), which can be reduced by opioid Difficulties in emotional processing and altered reward sensitivity
antagonists (Williams and Kirkham, 2002) and thus hinting at a are also seen (Oldershaw et al., 2012). Though these symptoms
potential interactive role of endogenous opioids in the reward disappear with refeeding in many, they do persist in some post
aspect of feeding behavior. recovery and are also seen in non suffering first degree relatives
(Holliday et al., 2014; Rose et al., 2012). Superior working memory
3. Neurobiology of ED performance in ED helps in practicing strategies to avoid food cues
which are considered irrelevant or unwanted by the individual
Despite of the obstacles like the confounding effect of (Brooks et al., 2012a, 2012b, 2012c). Inadequate self perception and
malnourishment, difficulty in premorbid identification of probable ruminations about body weight and shape suggest of a possible
patients, tendency to cross between subtypes and lack of association between psychological profile and neurobiology in the
consensus on definition for recovery (Kaye et al., 2011), studies pathogenesis of cognitive aberrations in ED (Shafran et al., 2004).
in neuropsychology, neuroimaging and receptor functions have Amygdala due to its involvement in anxiety and fear may be
contributed to a growing body of knowledge on the neurobiologi- associated with the learned fear of food in ED (Costafreda et al.,
cal aspect of ED. Hence a constantly strengthening consensus is 2013).
there about the neurobiological vulnerabilities making substantial
contribution to the pathogenesis of ED (Hausswolff-Juhlin et al., 3.3. Interoceptive awareness
2015).
Altered interoceptive awareness seems to be a precipitating and
3.1. Genetic evidences reinforcing factor in AN (Bruch, 1962; Fassino et al., 2004; Garner
et al., 1983; Kaye et al., 2011). Interoception is the awareness of the
Several studies have implicate genes involved in the serotoner- physiological condition of the complete body (Craig, 2002). The
gic & dopaminergic system along with those involved in weight insula is important in interoceptive monitoring of sensations vital
A. Mishra et al. / Asian Journal of PsychiatryAJP 25 (2017) 91–100 95

for the integrity of the internal body state and connects to systems recovering ED (Brewerton et al., 1990; Engel et al., 2005; Jimerson
responsible for attention, planning and action, through dorsolat- et al., 1997; Kaye and Strober, 2009; Walsh and Devlin, 1998). There
eral striatal pathways (Chikama et al., 1997; Craig, 2002; Paulus are evidences (Anderson et al., 1990; Biggio et al., 1974; Fernstrom
and Stein, 2006). Studies have found altered activity in insula of ill and Wurtman, 1971, 1972; Gibbons et al., 1979; Messing et al., 1976;
AN patients (Nozoe et al., 1993). Studies have also found elevated Young and Gauthier, 1981) showing that plasma tryptophan levels
pain thresholds in ED (Papežová et al., 2005), which persists post are lowered by a restricted diet, leading to decreased 5-HT
recovery (Stein et al., 2003) and is suggestive of altered production (Goodwin et al., 1987a, 1987b), which downregulates 5-
interoceptive awareness. Recent fMRI studies(Haase et al., 2009; HT transporter density (Huether et al., 1997) along with
Vocks et al., 2011; Wagner et al., 2008) focussing on the appetitive supersensitivity of postsynaptic receptors. Data showing reduced
circuitry using tastants, also suggest that a dysfunctional plasma tryptophan in emaciated AN (Schweiger et al., 1986) has led
interoceptive processing characterized by altered sensitivity to the idea that dietary restraint has anxiety reducing character in
and/or setpoint for sensory-interoceptive-reward processes in AN (Kaye et al., 2003; Strober, 1995; Vitousek and Manke, 1994).
response to palatable food (Small, 2009) might be a trait CSF 5-HIAA levels are significantly reduced in AN while normal in B
characteristic of AN. (Jimerson et al., 1992; Kaye et al., 1984, 1988). Various studies have
shown dysregulated binding potentials in 5-HT1A, 5-HT1B and 5-
3.4. Obsessisonality HT2A receptors in ED (Bailer et al., 2007a, 2007b; Becker et al.,
2007; Frank et al., 2002; Tammela et al., 2003). Harm avoidance
Research has found the possibility of phenomenological and and 5-HT2A binding in lateral and medial OFC, cingulate cortex and
functional overlap between ED and obsessive – compulsive parietal cortex show correlation in patients with AN (Bailer et al.,
disorder (OCD) (Altman and Shankman, 2009; Murphy et al., 2007b, 2005), who in general score high on harm avoidance and
2004) which has led to the suggestion of a shared etiopathogenesis low on novelty seeking and reward dependence (Klump et al.,
between the two disorders (Bellodi et al., 2001; Robbins et al., 2004). Studies also suggest a possibility of mesial temporal
2012). Many studies have shown dysfunctional limbic and (amygdala)-cingulate 5HT1A/2A imbalance as a common trait in
cognitive neural networks both in ED (Uher et al., 2003) and AN subgroups in relation to behavioral inhibition, anticipatory
OCD (Insel, 1992; Saxena, 2003). Elevated levels of impulsivity is anxiety or integration of mood and cognition (Charney and Deutch,
seen in both ED (Boisseau et al., 2009, 2012) and OCD (Lochner and 1996; Freedman et al., 2000).
Stein, 2006; Morein-Zamir et al., 2010). Behavioral and cognitive
inhibition impairment similar to OCD has also been seen in ED 4.2. Dopamine
(Galimberti et al., 2012; Murphy et al., 2004).
Dopaminergic dysfunction in fronto-striatal circuit can be
3.5. Neuroimaging evidences responsible for the failure to form hedonic association with
rewarding stimuli in ED (Frank et al., 2005; Steinglass and Walsh,
In general all neuroimaging show cerebral atrophy and enlarged 2006; Wagner et al., 2007). In AN reduced CSF DA metabolites
ventricles in patients with AN (Titova et al., 2013) and to a lesser (Kaye et al., 1984) which persists post recovery (Kaye et al., 1999),
extent in BN (Krieg et al., 1989). Presence of enlarged ventricles altered frequency of functional polymorphism of D2 receptor
despite normal weight in ED suggests there possibility as a genes (Bergen et al., 2005) and increased D2/D3 receptor binding
predisposing disturbance in the development of an ED (Kiriike in anteroventral striatum (Frank et al., 2005). In BN the dopamine
et al., 1990).Studies in brain lesions have found diagnostic EDs to be receptors are reduced and dopamine release is decreased in the
associated with right frontal and temporal lobe damage (Uher and striatum (Broft et al., 2012). Surprisingly people with BED have
Treasure, 2005). Parietal cortex studies (Delvenne et al., 1997; increased dopamine release in the striatum (Wang et al., 2011).
McGlynn and Schacter, 1989; Nozoe et al., 1995) suggest parietal Overall in people with ED it seems there is an imbalance in the
dysfunction (Delvenne et al., 1997) and its effect on body image antagonistically acting 5-HT and DA neurocircuitry (Daw et al.,
distortion in ED. Dorsolateral prefrontal cortex is implicated as an 2002; Duvvuri et al., 2009).
indicator of cognitive control in AN while as vulnerabilities in
cognitive control in BN (Brooks et al., 2011; Titova et al., 2013; Uher 4.3. Others
et al., 2003, 2004). The basal ganglia, the striatum in particular,
(dorsal with excessive inhibition of appetite and ventral involving A recent meta-analysis of several studies has shown that BDNF
parietal cortex, with dysfunctional sense of self and body image of levels are reduced in individuals with AN or BN (Brandys et al.,
distortion), is associated with abnormal responses to food and 2011) which could be an adaptive way of improving food
altered motivation in those with ED (Hausswolff-Juhlin et al., consumption in persistent starvation (Monteleone and Maj, 2013).
2015). fMRI studies on restrictive AN have shown hyper-reactivity Similarly increased endocannabinoids (specifically ananda-
of amygdala (Pietrini et al., 2011). AN has fronto-striatal circuitry mide) have been seen in AN and BED patients, but not in BN
involvement similar to autism as seen in excessive appetitive and (Monteleone et al., 2005). Also CB1 receptor markers are seen in
emotional restraint in both (Fonville et al., 2014; Martinez et al., higher amount in the bloods of AN and BN patients (Frieling et al.,
2014; Pepin and Stagnitti, 2012). Changes similar to anxiety 2009) which can been correlated with their increased number in
disorder in the frontal and temporal cortex have been seen in ED the insula in these patients as shown in a PET study (Gérard et al.,
(Gordon et al., 2001; Nozoe et al., 1993; Schulte-Rüther et al., 2012; 2011). Thus the dysregulation of the endocannabinoid system may
Zald and Kim, 1996). also reflect an adaptive response to improve hunger and food
intake (Monteleone and Maj, 2013).
4. Neurotransmitters and neuropeptides Lastly several peripheral neuropeptides such as leptin(Bara-
nowska et al., 2008), ghrelin (Monteleone et al., 2003; Otto et al.,
4.1. Serotonin 2001; Palmiter, 2007; Tanaka et al., 2002), CCK(Philipp et al., 1991),
PYY (Prince et al., 2009), oxyntomodulin(Cohen et al., 2003) etc.
5-HT abnormalities can lead to appetite dysregulation (Blun- are also found dysregulated in people with AN and BN (Bailer and
dell, 1984; Leibowitz and Shor-Posner, 1986). There are studies Kaye, 2003). This leads to dysfunctional DA activation and
suggesting presence of monoamine dysfunction both in ill and disturbances in normal appetite (Chattopadhyaya et al., 2007).
96 A. Mishra et al. / Asian Journal of PsychiatryAJP 25 (2017) 91–100

5. Clinical perspective 7. Conclusion

Early intervention appears to be the most effective treatment The DSM 5 reorganized its classification system for eating
for those with EDs (Fisher et al., 2010). 5-HT2A receptor due to disorder with the idea of improving the clinical utility. In
its response to SSRI is of interest in ED as it is associated with addition to introduction of some new and reintroduction of
modulation of appetite and mood (Bailer et al., 2004; some old diagnostic entities it specifically decided not to
Bonhomme and Esposito, 1998; De Vry and Schreiber, 2000; consider obesity as a psychiatric diagnosis (Attia et al., 2013).
Simansky, 1995; Stockmeier, 1997). For AN in adults, there is no The evidences on the neurobiological studies focussing on the
consistent evidence to date for an effective treatment of choice disorders other than AN, BN and to some extent BED, is largely
(Hausswolff-Juhlin et al., 2015). AN do not respond well to lacking. Still from the available studies it would be safe to
Selective Serotonin Reuptake Inhibitors (SSRIs) (Walsh et al., conclude that neurobiology has an important role in the
2006) because there is dysfunction in 5-HT neurocircuitry pathogenesis of ED. Owing to the inconsistencies across studies
leading to reduced 5-HT release at the synapse while SSRI rely our current knowledge at best might be called nascent, but
on 5-HT release (Duvvuri et al., 2009). Also increased 5-HT1A nevertheless considering the associated morbidity and the fact
receptor activity in AN accounts for poor response to that AN has the highest mortality rate (Arcelus et al., 2011;
medications (Bailer et al., 2007a, 2007b). A WFSBP task force Harris and Barraclough, 1998) among all psychiatry illnesses,
on ED found grade B evidence for use of olanzapine, grade C the neurobiological aspect of ED definitely warrants further
evidence for quetiapine and risperidone and no conclusive research. The current treatment modalities from pharmacother-
evidence for antidepressants (Aigner et al., 2011) in AN as apy to psychotherapy being not very promising, further
characterized by weight gain. In BN treatment there is grade A refinement in the neurobiological model may guide us in a
evidence for fluoxetine in high dosage, and also for topiramate better targeted approach to treatment.
and TCA, as characterized by reduced episodes of binge eating
(Mitchell et al., 2003). Though relapse during treatment is quite References
common (Walsh and Devlin, 1995). From their clinical experi-
ence, Kaye et al. suggested better response to SSRI in RAN Aigner, M., Treasure, J., Kaye, W., Kasper, S., 2011. World Federation of Societies of
Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of
compared to BAN and poor response in some BN even to high eating disorders. World J. Biol. Psychiatry 12 (6), 400–443.
dosage of SSRI (Kaye et al., 2001, 1991; Walsh et al., 2006). Altman, S.E., Shankman, S.A., 2009. What is the association between obsessive–
compulsive disorder and eating disorders? Clin. Psychol. Rev. 29 (7), 638–646.
American Psychiatric Association, 2013. Diagnostic and Statistical Manual of Mental
6. The research domain criteria (RDoC) framework for research Disorders. DSM-5: ManMag.
in ED Anderson, I., Parry-Billings, M., Newsholme, E., Fairburn, C.G., Cowen, P., 1990.
Dieting reduces plasma tryptophan and alters brain 5-HT function in women.
Psychol. Med. 20 (04), 785–791.
The RDoC in its bid to make psychiatry more precise, tries to Arana, F.S., Parkinson, J.A., Hinton, E., Holland, A.J., Owen, A.M., Roberts, A.C., 2003.
incorporate research in terms of various units of analysis (genes, Dissociable contributions of the human amygdala and orbitofrontal cortex to
molecules, cells, circuits, physiology, behavior, and self-reports) incentive motivation and goal selection. J. Neurosci. 23 (29), 9632–9638.
Arcelus, J., Mitchell, A.J., Wales, J., Nielsen, S., 2011. Mortality rates in patients with
into the identified dimensions of behavior (negative valence
anorexia nervosa and other eating disorders: a meta-analysis of 36 studies.
system, positive valence system, cognitive systems, systems for Arch. Gen. Psychiatry 68 (7), 724–731.
social processes and arousal & regulatory systems) (Insel et al., Arora, S., 2006. Role of neuropeptides in appetite regulation and obesity–a review.
2010) for established psychiatric illnesses. In contrast to the Neuropeptides 40 (6), 375–401.
Attia, E., Becker, A.E., Bryant-Waugh, R., Hoek, H.W., Kreipe, R.E., Marcus, M.D., et al.,
categorical approach, it provides a unique way to investigate the 2013. Feeding and eating disorders in DSM-5. Am. J. Psychiatry 170 (11), 1237–
association between neural processes, etiology and outcome in 1239.
individuals with ED (Wildes and Marcus, 2015). Also considering Bailer, U.F., Kaye, W.H., 2003. A review of neuropeptide and neuroendocrine
dysregulation in anorexia and bulimia nervosa. Curr. Drug Targets-CNS &
the diagnostic instability, high rate of comorbidity and a Neurol. Disord. 2 (1), 53–59.
predominance of “other specified feeding or eating disorder” Bailer, U.F., Price, J.C., Meltzer, C.C., Mathis, C.A., Frank, G.K., Weissfeld, L., et al., 2004.
diagnosis, a utilization of a broader inclusion and exclusion criteria Altered 5-HT2A receptor binding after recovery from bulimia-type anorexia
nervosa: relationships to harm avoidance and drive for thinness.
improves the validity of studies in the RDoC approach (Sanislow Neuropsychopharmacology 29 (6), 1143.
et al., 2010). Bailer, U.F., Frank, G.K., Henry, S.E., Price, J.C., Meltzer, C.C., Weissfeld, L., et al., 2005.
The interest in the dimensional approach in classifying eating Altered brain serotonin 5-HT1A receptor binding after recovery from anorexia
nervosa measured by positron emission tomography and [carbonyl11C] WAY-
disorders is increasing steadily (Brooks et al., 2012c; Wildes and
100635. Arch. Gen. Psychiatry 62 (9), 1032–1041.
Marcus, 2013). Several studies as discussed in preceding sections Bailer, U.F., Frank, G.K., Henry, S.E., Price, J.C., Meltzer, C.C., Becker, C., et al., 2007a.
have provided support for various RDoC dimensions in eating Serotonin transporter binding after recovery from eating disorders.
Psychopharmacology 195 (3), 315–324.
disorders. For example – Hyper-responsiveness in fear circuits to
Bailer, U.F., Frank, G.K., Henry, S.E., Price, J.C., Meltzer, C.C., Mathis, C.A., et al., 2007b.
provoking stimuli (Friederich et al., 2013) (negative valence Exaggerated 5-HT1A but normal 5-HT2A receptor activity in individuals ill with
system), dysregulated reward processing (Manwaring et al., anorexia nervosa. Biol. Psychiatry 61 (9), 1090–1099.
2011; Steinglass et al., 2012) (positive valence), fronto-striatal Balodis, I.M., Molina, N.D., Kober, H., Worhunsky, P.D., White, M.A., Sinha, R., et al.,
2013. Divergent neural substrates of inhibitory control in binge eating disorder
circuit hypoactivity in eating disorder during performance on tasks relative to other manifestations of obesity. Obesity 21 (2), 367–377.
involving response inhibition (Balodis et al., 2013; Marsh et al., Baranowska, B., Baranowska-Bik, A., Bik, W., Martynska, L., 2008. The role of leptin
2011; Oberndorfer et al., 2011), and impaired attachment and and orexins in the dysfunction of hypothalamo-pituitary-gonadal regulation
and in the mechanism of hyperactivity in patients with anorexia nervosa. Neuro
social communication (Caglar-Nazali et al., 2014) (social processes) Endocrinol. Lett. 29 (1), 37–40.
etc. A detailed discussion of these studies is beyond the scope of Basso, A.M., Kelley, A.E., 1999. Feeding induced by GABA A receptor stimulation
this text and interested readers are advised to refer to relevant within the nucleus accumbens shell: regional mapping and characterization of
macronutrient and taste preference. Behav. Neurosci. 113 (2), 324.
studies as mentioned. Overall, an RDoC approach to studies in Bellodi, L., Cavallini, M.C., Bertelli, S., Chiapparino, D., Riboldi, C., Smeraldi, E., 2001.
future can prove beneficial to the advancement of our under- Morbidity risk for obsessive-compulsive spectrum disorders in first-degree
standing of ED. relatives of patients with eating disorders. Am. J. Psychiatry 158 (4), 563–569.
Bergen, A.W., Yeager, M., Welch, R.A., Haque, K., Ganjei, J.K., van den Bree, M.B., et al.,
2005. Association of multiple DRD2 polymorphisms with anorexia nervosa.
Neuropsychopharmacology 30 (9), 1703–1710.
A. Mishra et al. / Asian Journal of PsychiatryAJP 25 (2017) 91–100 97

Berridge, K.C., Robinson, T.E., 2003. Parsing reward. Trends Neurosci. 26 (9), 507– Delvenne, V., Goldman, S., Biver, F., De Maertalaer, V., Wikler, D., Damhaut, P.,
513. Lotstra, F., 1997. Brain hypometabolism of glucose in low-weight depressed
Berridge, K.C., 2009. ‘Liking’and ‘wanting’ food rewards: brain substrates and roles patients and in anorectic patients: a consequence of starvation? J. Affect. Disord.
in eating disorders. Physiol. Behav. 97 (5), 537–550. 44 (1), 69–77.
Berthoud, H.-R., Morrison, C., 2008. The brain, appetite, and obesity. Annu. Rev. Di Chiara, G., Imperato, A., 1988. Drugs abused by humans preferentially increase
Psychol. 59, 55–92. synaptic dopamine concentrations in the mesolimbic system of freely moving
Berthoud, H.R., 2006. Homeostatic and non-homeostatic pathways involved in the rats. Proc. Natl. Acad. Sci. 85 (14), 5274–5278.
control of food intake and energy balance. Obesity 14 (S8), 197S–200S. Duvvuri, V., Risbrough, V.B., Kaye, W.H., Geyer, M.A., 2009. 5-HT 1A receptor
Biggio, G., Fadda, F., Fanni, P., Tagliamonte, A., Gessa, G.L., 1974. Rapid depletion of activation is necessary for 5-MeODMT-dependent potentiation of feeding
serum tryptophan, brain tryptophan, serotonin and 5-hydroxyindoleacetic acid inhibition. Pharmacol. Biochem. Behav. 93 (3), 349–353.
by a tryptophan-free diet. Life Sci. 14 (7), 1321–1329. Ebenezer, I., Baldwin, B., 1990. Effect of intracerebroventricular administration of
Blundell, J., 1984. Serotonin and appetite. Neuropharmacology 23 (12), 1537–1551. the GABAB-receptor agonist baclofen on operant feeding in satiated pigs. Br. J.
Boisseau, C.L., Thompson-Brenner, H., Eddy, K.T., Satir, D.A., 2009. Impulsivity and Pharmacol. 101 (3), 559–562.
personality variables in adolescents with eating disorders. J. Nerv. Mental Dis. Engel, S.G., Corneliussen, S.J., Wonderlich, S.A., Crosby, R.D., le Grange, D., Crow, S., et
197 (4), 251. al., 2005. Impulsivity and compulsivity in bulimia nervosa. Int. J. Eat. Disord. 38
Boisseau, C.L., Thompson-Brenner, H., Caldwell-Harris, C., Pratt, E., Farchione, T., (3), 244–251.
Barlow, D.H., 2012. Behavioral and cognitive impulsivity in obsessive– Fassino, S., Pierò, A., Gramaglia, C., Abbate-Daga, G., 2004. Clinical,
compulsive disorder and eating disorders. Psychiatry Res. 200 (2), 1062–1066. psychopathological and personality correlates of interoceptive awareness in
Bonhomme, N., Esposito, E., 1998. Involvement of serotonin and dopamine in the anorexia nervosa, bulimia nervosa and obesity. Psychopathology 37 (4), 168–
mechanism of action of novel antidepressant drugs: a review. J. Clin. 174.
Psychopharmacol. 18 (6), 447–454. Fernstrom, J.D., Wurtman, R.J., 1971. Brain serotonin content: increase following
Brandys, M.K., Kas, M.J., van Elburg, A.A., Campbell, I.C., Adan, R.A., 2011. A meta- ingestion of carbohydrate diet. Science 174 (4013), 1023–1025.
analysis of circulating BDNF concentrations in anorexia nervosa. World J. Biol. Fernstrom, J.D., Wurtman, R.J., 1972. Brain serotonin content: physiological
Psychiatry 12 (6), 444–454. regulation by plasma neutral amino acids. Science 178 (4059), 414–416.
Brewerton, T., Brandt, H., Lesem, M., Murphy, D., Jimerson, D., 1990. Serotonin in Fisher, C.A., Hetrick, S.E., Rushford, N., 2010. Family therapy for anorexia nervosa.
eating disorders. Serotonin in Major Psychiatric Disorders, Vol. 21. American Cochrane Database Syst. Rev. 4.
Psychiatric Press, Washington, DC, pp. 155–184. Fonville, L., Giampietro, V., Williams, S., Simmons, A., Tchanturia, K., 2014.
Broft, A., Shingleton, R., Kaufman, J., Liu, F., Kumar, D., Slifstein, M., et al., 2012. Alterations in brain structure in adults with anorexia nervosa and the impact of
Striatal dopamine in bulimia nervosa: a PET imaging study. Int. J. Eat. Disord. 45 illness duration. Psychol. Med. 44 (09), 1965–1975.
(5), 648–656. Francis, S., Rolls, E.T., Bowtell, R., McGlone, F., O'Doherty, J., Browning, A., Smith, E.,
Brooks, S.J., Barker, G.J., O'Daly, O.G., Brammer, M., Williams, S.C., Benedict, C., et al., 1999. The representation of pleasant touch in the brain and its relationship with
2011. Restraint of appetite and reduced regional brain volumes in anorexia taste and olfactory areas. Neuroreport 10 (3), 453–459.
nervosa: a voxel-based morphometric study. BMC Psychiatry 11 (1), 179. Frank, G.K., Kaye, W.H., Meltzer, C.C., Price, J.C., Greer, P., McConaha, C., Skovira, K.,
Brooks, S.J., O’Daly, O., Uher, R., Friederich, H.-C., Giampietro, V., Brammer, M., et al., 2002. Reduced 5-HT2A receptor binding after recovery from anorexia nervosa.
2012a. Thinking about eating food activates visual cortex with reduced bilateral Biol. Psychiatry 52 (9), 896–906.
cerebellar activation in females with anorexia nervosa: an fMRI study. PLoS One Frank, G.K., Bailer, U.F., Henry, S.E., Drevets, W., Meltzer, C.C., Price, J.C., et al., 2005.
7 (3), e34000. Increased dopamine D2/D3 receptor binding after recovery from anorexia
Brooks, S.J., O’Daly, O.G., Uher, R., Schiöth, H.B., Treasure, J., Campbell, I.C., 2012b. nervosa measured by positron emission tomography and [11C] raclopride. Biol.
Subliminal food images compromise superior working memory performance in Psychiatry 58 (11), 908–912.
women with restricting anorexia nervosa. Conscious. Cogn. 21 (2), 751–763. Freedman, L.J., Insel, T.R., Smith, Y., 2000. Subcortical projections of area 25
Brooks, S.J., Rask-Andersen, M., Benedict, C., Schiöth, H.B., 2012c. A debate on (subgenual cortex) of the macaque monkey. J. Comp. Neurol. 421 (2), 172–188.
current eating disorder diagnoses in light of neurobiological findings: is it time Frey, J., Hebebrand, J., Müller, B., Ziegler, A., Blum, W.F., Remschmidt, H., Herpertz-
for a spectrum model? BMC Psychiatry 12 (1), 76. Dahlmann, B.M., 2000. Reduced body fat in long-term followed-up female
Bruch, H., 1962. Perceptual and conceptual disturbances in anorexia nervosa. patients with anorexia nervosa. J. Psychiatr. Res. 34 (1), 83–88.
Psychosom. Med. 24 (2), 187–194. Friederich, H.C., Wu, M., Simon, J.J., Herzog, W., 2013. Neurocircuit function in eating
Bulik, C.M., Sullivan, P.F., Tozzi, F., Furberg, H., Lichtenstein, P., Pedersen, N.L., 2006. disorders. Int. J. Eat. Disord. 46 (5), 425–432.
Prevalence, heritability, and prospective risk factors for anorexia nervosa. Arch. Frieling, H., Albrecht, H., Jedtberg, S., Gozner, A., Lenz, B., Wilhelm, J., Bleich, S., 2009.
Gen. Psychiatry 63 (3), 305–312. Elevated cannabinoid 1 receptor mRNA is linked to eating disorder related
Caglar-Nazali, H.P., Corfield, F., Cardi, V., Ambwani, S., Leppanen, J., Olabintan, O., et behavior and attitudes in females with eating disorders.
al., 2014. A systematic review and meta-analysis of ‘Systems for Social Psychoneuroendocrinology 34 (4), 620–624.
Processes’ in eating disorders. Neurosci. Biobehav. Rev. 42, 55–92. Gérard, N., Pieters, G., Goffin, K., Bormans, G., Van Laere, K., 2011. Brain type 1
Calero-Elvira, A., Krug, I., Davis, K., López, C., Fernández-Aranda, F., Treasure, J., 2009. cannabinoid receptor availability in patients with anorexia and bulimia nervosa.
Meta-analysis on drugs in people with eating disorders. Eur. Eat. Disord. Rev. 17 Biol. Psychiatry 70 (8), 777–784.
(4), 243–259. Gadalla, T., Piran, N., 2007. Co-occurrence of eating disorders and alcohol use
Charney, D.S., Deutch, A., 1996. A functional neuroanatomy of anxiety and fear: disorders in women: a meta analysis. Arch. Women's Mental Health 10 (4), 133–
implications for the pathophysiology and treatment of anxiety disorders. Crit. 140.
Rev.TM Neurobiol. 10, 3–4. Galimberti, E., Martoni, R.M., Cavallini, M.C., Erzegovesi, S., Bellodi, L., 2012. Motor
Chattopadhyaya, B., Di Cristo, G., Wu, C.Z., Knott, G., Kuhlman, S., Fu, Y., et al., 2007. inhibition and cognitive flexibility in eating disorder subtypes. Prog. Neuro-
GAD67-mediated GABA synthesis and signaling regulate inhibitory synaptic Psychopharmacol. Biol. Psychiatry 36 (2), 307–312.
innervation in the visual cortex. Neuron 54 (6), 889–903. Garner, D.M., Olmstead, M.P., Polivy, J., 1983. Development and validation of a
Chikama, M., McFarland, N.R., Amaral, D.G., Haber, S.N., 1997. Insular cortical multidimensional eating disorder inventory for anorexia nervosa and bulimia.
projections to functional regions of the striatum correlate with cortical Int. J. Eat. Disord. 2 (2), 15–34.
cytoarchitectonic organization in the primate. J. Neurosci. 17 (24), 9686–9705. Gibbons, J.L., Barr, G.A., Bridger, W.H., Liebowitz, S.F., 1979. Manipulations of dietary
Cohen, M.A., Ellis, S.M., Le Roux, C.W., Batterham, R.L., Park, A., Patterson, M., et al., tryptophan: effects on mouse killing and brain serotonin in the rat. Brain Res.
2003. Oxyntomodulin suppresses appetite and reduces food intake in humans. 169 (1), 139–153.
J. Clin. Endocrinol. Metab. 88 (10), 4696–4701. Gibson, C., Carnell, S., Ochner, C., Geliebter, A., 2010. Neuroimaging, gut peptides and
Cordeira, J.W., Frank, L., Sena-Esteves, M., Pothos, E.N., Rios, M., 2010. Brain-derived obesity: novel studies of the neurobiology of appetite. J. Neuroendocrinol. 22
neurotrophic factor regulates hedonic feeding by acting on the mesolimbic (8), 833–845.
dopamine system. J. Neurosci. 30 (7), 2533–2541. Gillberg, I.C., Gillberg, C., Råstam, M., Johansson, M., 1996. The cognitive profile of
Costafreda, S.G., McCann, P., Saker, P., Cole, J.H., Cohen-Woods, S., Farmer, A.E., et al., anorexia nervosa: a comparative study including a community-based sample.
2013. Modulation of amygdala response and connectivity in depression by Compr. Psychiatry 37 (1), 23–30.
serotonin transporter polymorphism and diagnosis. J. Affect. Disord. 150 (1), Giordano, S., 2007. Understanding Eating Disorders: Conceptual and Ethical Issues
96–103. in the Treatment of Anorexia and Bulimia Nervosa. Clarendon Press.
Cota, D., Marsicano, G., Lutz, B., Vicennati, V., Stalla, G., Pasquali, R., Pagotto, U., 2003. Goodwin, G., Fairburn, C.G., Cowen, P., 1987a. Dieting changes serotonergic function
Endogenous cannabinoid system as a modulator of food intake. Int. J. Obes. 27 in women, not men: implications for the aetiology of anorexia nervosa? Psychol.
(3), 289–301. Med. 17 (04), 839–842.
Craig, A.D., 2002. How do you feel? Interoception: the sense of the physiological Goodwin, G., Fraser, S., Stump, K., Fairburn, C., Elliott, J., Cowen, P., 1987b. Dieting
condition of the body. Nat. Rev. Neurosci. 3 (8), 655–666. and weight loss in volunteers increases the number of a 2-adrenoceptors and 5-
Dagher, A., 2009. The neurobiology of appetite: hunger as addiction. Int. J. Obes. 33, HT receptors on blood platelets without effect on [3 H] imipramine binding. J.
S30–S33. Affect. Disord. 12 (3), 267–274.
Daw, N.D., Kakade, S., Dayan, P., 2002. Opponent interactions between serotonin and Gordon, C.M., Dougherty, D.D., Fischman, A.J., Emans, S.J., Grace, E., Lamm, R., et al.,
dopamine. Neural Netw. 15 (4), 603–616. 2001. Neural substrates of anorexia nervosa: a behavioral challenge study with
De Vry, J., Schreiber, R., 2000. Effects of selected serotonin 5-HT 1 and 5-HT 2 positron emission tomography. J. Pediatr. 139 (1), 51–57.
receptor agonists on feeding behavior: possible mechanisms of action. Neurosci. Gorwood, P., Kipman, A., Foulon, C., 2003. The human genetics of anorexia nervosa.
Biobehav. Rev. 24 (3), 341–353. Eur. J. Pharmacol. 480 (1), 163–170.
98 A. Mishra et al. / Asian Journal of PsychiatryAJP 25 (2017) 91–100

Grunberg, N.E., Bowen, D.J., Winders, S.E., 1986. Effects of nicotine on body weight satiation: stimulation of eating by 2-arachidonoyl glycerol. Br. J. Pharmacol. 136
and food consumption in female rats. Psychopharmacology 90 (1), 101–105. (4), 550–557.
Haase, L., Cerf-Ducastel, B., Murphy, C., 2009. Cortical activation in response to pure Klump, K.L., Strober, M., Bulik, C.M., Thornton, L., Johnson, C., Devlin, B., et al., 2004.
taste stimuli during the physiological states of hunger and satiety. Neuroimage Personality characteristics of women before and after recovery from an eating
44 (3), 1008–1021. disorder. Psychol. Med. 34 (08), 1407–1418.
Hall, J.E., 2010. Guyton and Hall Textbook of Medical Physiology. Elsevier Health Koronyo-Hamaoui, M., Danziger, Y., Frisch, A., Stein, D., Leor, S., Laufer, N., et al.,
Sciences. 2002. Association between anorexia nervosa and the hsKCa3 gene: a family-
Harris, E.C., Barraclough, B., 1998. Excess mortality of mental disorder. Br. J. based and case control study. Mol. Psychiatry 7 (1), 82–85.
Psychiatry 173 (1), 11–53. Kramer, G.F., Kittleson, M.J., 2005. The Truth About Eating Disorders. Infobase
Hausswolff-Juhlin, Y., Brooks, S., Larsson, M., 2015. The neurobiology of eating Publishing.
disorders—a clinical perspective. Acta Psychiatr. Scand. 131 (4), 244–255. Krieg, J.-C., Lauer, C., Pirke, K.-M., 1989. Structural brain abnormalities in patients
Hebebrand, J., Remschmidt, H., 1995. Anorexia nervosa viewed as an extreme weight with bulimia nervosa. Psychiatry Res. 27 (1), 39–48.
condition: genetic implications. Hum. Genet. 95 (1), 1–11. Leibowitz, S.F., Alexander, J.T., 1998. Hypothalamic serotonin in control of eating
Hebebrand, J., Blum, W., Barth, N., Coners, H., Englaro, P., Juul, A., et al., 1997. With behavior, meal size, and body weight. Biol. Psychiatry 44 (9), 851–864.
anorexia nervosa are. Mol. Psychiatry 2, 330–334. Leibowitz, S.F., Shor-Posner, G., 1986. Brain serotonin and eating behavior. Appetite
Hinton, E.C., Parkinson, J.A., Holland, A.J., Arana, F.S., C Roberts, A., Owen, A.M., 2004. 7, 1–14.
Neural contributions to the motivational control of appetite in humans. Eur. J. Levin, E.D., Morgan, M.M., Galvez, C., Ellison, G.D., 1987. Chronic nicotine and
Neurosci. 20 (5), 1411–1418. withdrawal effects on body weight and food and water consumption in female
Holliday, J., Tchanturia, K., Landau, S., Collier, D., Treasure, J., 2014. Is impaired set- rats. Physiol. Behav. 39 (4), 441–444.
shifting an endophenotype of anorexia nervosa? Am. J. Psychiatry 16 (12), Lochner, C., Stein, D.J., 2006. Does work on obsessive–compulsive spectrum
2269–2275. disorders contribute to understanding the heterogeneity of obsessive–
Huether, G., Zhou, D., Rüther, E., 1997. Long-term modulation of presynaptic 5-HT- compulsive disorder? Prog. Neuro-Psychopharmacol. Biol. Psychiatry 30 (3),
output: experimentally induced changes in cortical 5-HT-transporter density, 353–361.
tryptophan hydroxylase content and 5-HT innervation density. J. Neural Maldonado-Irizarry, C.S., Swanson, C.J., Kelley, A.E., 1995. Glutamate receptors in the
Transm. 104 (10), 993–1004. nucleus accumbens shell control feeding behavior via the lateral hypothalamus.
Insel, T., Cuthbert, B., Garvey, M., Heinssen, R., Pine, D.S., Quinn, K., et al., 2010. J. Neurosci. 15 (10), 6779–6788.
Research domain criteria (RDoC): toward a new classification framework for Manwaring, J.L., Green, L., Myerson, J., Strube, M.J., Wilfley, D.E., 2011. Discounting of
research on mental disorders. Am. J. Psychiatry 167 (7), 748–751. various types of rewards by women with and without binge eating disorder:
Insel, T.R., 1992. Toward a neuroanatomy of obsessive-compulsive disorder. Arch. evidence for general rather than specific differences. Psychol. Rec. 61 (4), 561.
Gen. Psychiatry 49 (9), 739–744. Marsh, R., Horga, G., Wang, Z., Wang, P., Klahr, K.W., Berner, L.A., Peterson, B.S., 2011.
Jimerson, D.C., Lesem, M.D., Kaye, W.H., Brewerton, T.D., 1992. Low serotonin and An FMRI study of self-regulatory control and conflict resolution in adolescents
dopamine metabolite concentrations in cerebrospinal fluid from bulimic with bulimia nervosa. Am. J. Psychiatry 168, 1210–1220.
patients with frequent binge episodes. Arch. Gen. Psychiatry 49 (2), 132–138. Martinez, G., Cook-Darzens, S., Chaste, P., Mouren, M., Doyen, C., 2014. [Anorexia
Jimerson, D.C., Wolfe, B.E., Metzger, E.D., Finkelstein, D.M., Cooper, T.B., Levine, J.M., nervosa in the light of neurocognitive functioning: new theoretical and
1997. Decreased serotonin function in bulimia nervosa. Arch. Gen. Psychiatry 54 therapeutic perspectives]. L'Encephale 40 (2), 160–167.
(6), 529–534. McGlynn, S.M., Schacter, D.L., 1989. Unawareness of deficits in neuropsychological
Jo, Y.-H., Wiedl, D., Role, L.W., 2005. Cholinergic modulation of appetite-related syndromes. J. Clin. Exp. Neuropsychol. 11 (2), 143–205.
synapses in mouse lateral hypothalamic slice. J. Neurosci. 25 (48), 11133–11144. Meguid, M.M., Fetissov, S.O., Varma, M., Sato, T., Zhang, L., Laviano, A., Rossi-Fanelli,
Kalra, S.P., Dube, M.G., Pu, S., Xu, B., Horvath, T.L., Kalra, P.S., 1999. Interacting F., 2000. Hypothalamic dopamine and serotonin in the regulation of food intake.
appetite-regulating pathways in the hypothalamic regulation of body weight 1. Nutrition 16 (10), 843–857.
Endocr. Rev. 20 (1), 68–100. Meister, B., 2007. Neurotransmitters in key neurons of the hypothalamus that
Kampe, J., Tschöp, M., Hollis, J., Oldfield, B.J., 2009. An anatomic basis for the regulate feeding behavior and body weight. Physiol. Behav. 92 (1), 263–271.
communication of hypothalamic, cortical and mesolimbic circuitry in the Messing, R.B., Fisher, L.A., Phebus, L., Lytle, L.D., 1976. Interaction of diet and drugs in
regulation of energy balance. Eur. J. Neurosci. 30 (3), 415–430. the regulation of brain 5-hydroxyindoles and the response to painful electric
Kaye, W., Strober, M., 2009. Neurobiology of eating disorders, The American shock. Life Sci. 18 (7), 707–714.
Psychiatric Publishing Textbook of Psychopharmacology. 4th edition Am Mitchell, J., De Zwaan, M., Roerig, J., 2003. Drug therapy for patients with eating
Psychiatric Assoc. disorders. Curr. Drug Targets-CNS Neurol. Disord. 2 (1), 17–29.
Kaye, W.H., Ebert, M.H., Raleigh, M., Lake, C.R., 1984. Abnormalities in CNS Miyata, G., Meguid, M.M., Fetissov, S.O., Torelli, G.F., Kim, H.-J., 1999. Nicotine's effect
monoamine metabolism in anorexia nervosa. Arch. Gen. Psychiatry 41 (4), 350– on hypothalamic neurotransmitters and appetite regulation. Surgery 126 (2),
355. 255–263.
Kaye, W.H., Gwirtsman, H.E., George, D.T., Jimerson, D.C., Ebert, M.H., 1988. CSF 5- Mogenson, G.J., Jones, D.L., Yim, C.Y., 1980. From motivation to action: functional
HIAA concentrations in anorexia nervosa: reduced values in underweight interface between the limbic system and the motor system. Prog. Neurobiol. 14
subjects normalize after weight gain. Biol. Psychiatry 23 (1), 102–105. (2), 69–97.
Kaye, W.H., Berrettini, W., Gwirtsman, H., George, D.T., 1990. Altered cerebrospinal Monteleone, P., Maj, M., 2013. Dysfunctions of leptin, ghrelin, BDNF and
fluid neuropeptide Y and peptide YY immunoreactivity in anorexia and bulimia endocannabinoids in eating disorders: beyond the homeostatic control of food
nervosa. Arch. Gen. Psychiatry 47 (6), 548–556. intake. Psychoneuroendocrinology 38 (3), 312–330.
Kaye, W.H., Weltzin, T.E., Hsu, L.G., Bulik, C.M., 1991. An open trial of fluoxetine in Monteleone, P., Fabrazzo, M., Martiadis, V., Fuschino, A., Serritella, C., Milici, N., Maj,
patients with anorexia nervosa. J. Clin. Psychiatry 52 (November (11)), 464–471. M., 2003. Opposite changes in circulating adiponectin in women with bulimia
Kaye, W., Greeno, C.G., Moss, H., Fernstrom, J., Lilenfeld, L.R., Weltzin, T.E., Mann, J.J., nervosa or binge eating disorder. J. Clin. Endocrinol. Metab. 88 (11), 5387–5391.
1998a. Altreation in serotonin activity and psychiatric symptoms after recovery Monteleone, P., Matias, I., Martiadis, V., De Petrocellis, L., Maj, M., Di Marzo, V., 2005.
from bulimia nevosa. Arch. Gen. Psychitry 55, 927–993\. Blood levels of the endocannabinoid anandamide are increased in anorexia
Kaye, W.H., Greeno, C.G., Moss, H., Fernstrom, J., Fernstrom, M., Lilenfeld, L.R., et al., nervosa and in binge-eating disorder, but not in bulimia nervosa.
1998b. Alterations in serotonin activity and psychiatric symptoms after Neuropsychopharmacology 30 (6), 1216–1221.
recovery from bulimia nervosa. Arch. Gen. Psychiatry 55 (10), 927–935. Morein-Zamir, S., Fineberg, N., Robbins, T., Sahakian, B., 2010. Inhibition of thoughts
Kaye, W.H., Frank, G.K., McConaha, C., 1999. Altered dopamine activity after recovery and actions in obsessive-compulsive disorder: extending the endophenotype?
from restricting-type anorexia nervosa. Neuropsychopharmacology 21 (4), Psychol. Med. 40 (02), 263–272.
503–506. Murphy, R., Nutzinger, D., Paul, T., Leplow, B., 2004. Conditional-associative learning
Kaye, W.H., Nagata, T., Weltzin, T.E., Hsu, L.G., Sokol, M.S., McConaha, C., Deep, D., in eating disorders: a comparison with OCD. J. Clin. Exp. Neuropsychol. 26 (2),
2001. Double-blind placebo-controlled administration of fluoxetine in 190–199.
restricting-and restricting-purging-type anorexia nervosa. Biol. Psychiatry 49 Nakagawa, T., Tsuchida, A., Itakura, Y., Nonomura, T., Ono, M., Hirota, F., Noguchi, H.,
(7), 644–652. 2000. Brain-derived neurotrophic factor regulates glucose metabolism by
Kaye, W.H., Barbarich, N.C., Putnam, K., Gendall, K.A., Fernstrom, J., Fernstrom, M., et modulating energy balance in diabetic mice. Diabetes 49 (3), 436–444.
al., 2003. Anxiolytic effects of acute tryptophan depletion in anorexia nervosa. Nisbett, R.E., 1972. Eating behavior and obesity in men and animals1. In Hunger and
Int. J. Eat. Disord. 33 (3), 257–267. Satiety in Health and Disease. Karger Publishers, pp. 173–193.
Kaye, W.H., Wagner, A., Fudge, J.L., Paulus, M., 2011. Neurocircuity of eating Nonogaki, K., Strack, A.M., Dallman, M.F., Tecott, L.H., 1998. Leptin-independent
disorders. Behavioral Neurobiology of Eating Disorders. Springer, pp. 37–57. hyperphagia and type 2 diabetes in mice with a mutated serotonin 5-HT2C
Kelley, A.E., Baldo, B.A., Pratt, W.E., Will, M.J., 2005. Corticostriatal-hypothalamic receptor gene. Nat. Med. 4 (10), 1152–1156.
circuitry and food motivation: integration of energy, action and reward. Physiol. Nozoe, S.-i., Naruo, T., Nakabeppu, Y., Soejima, Y., Nakajo, M., Tanaka, H., 1993.
Behav. 86 (5), 773–795. Changes in regional cerebral blood flow in patients with anorexia nervosa
Keys, A., Brožek, J., Henschel, A., Mickelsen, O., Taylor, H.L., 1950. The Biology of detected through single photon emission tomography imaging. Biol. Psychiatry
Human Starvation, 2 vols. . 34 (8), 578–580.
Kiriike, N., Nishiwaki, S., Nagata, T., Inoue, Y., Inoue, K., Kawakita, Y., 1990. Nozoe, S.-I., Naruo, T., Yonekura, R., Nakabeppu, Y., Soejima, Y., Nagai, N., et al., 1995.
Ventricular enlargement in normal weight bulimia. Acta Psychiatr. Scand. 82 Comparison of regional cerebral blood flow in patients with eating disorders.
(3), 264–266. Brain Res. Bull. 36 (3), 251–255.
Kirkham, T.C., Williams, C.M., Fezza, F., Marzo, V.D., 2002. Endocannabinoid levels in
rat limbic forebrain and hypothalamus in relation to fasting, feeding and
A. Mishra et al. / Asian Journal of PsychiatryAJP 25 (2017) 91–100 99

Numan, S., Seroogy, K.B., 1999. Expression of trkB and trkC mRNAs by adult midbrain Steinglass, J., Walsh, B.T., 2006. Habit learning and anorexia nervosa: a cognitive
dopamine neurons: a double-label in situ hybridization study. J. Comp. Neurol. neuroscience hypothesis. Int. J. Eat. Disord. 39 (4), 267–275.
403 (3), 295–308. Steinglass, J.E., Figner, B., Berkowitz, S., Simpson, H.B., Weber, E.U., Walsh, B.T., 2012.
O'Doherty, J., Rolls, E.T., Francis, S., Bowtell, R., McGlone, F., 2001. Representation of Increased capacity to delay reward in anorexia nervosa. J. Int. Neuropsychol.
pleasant and aversive taste in the human brain. J. Neurophysiol. 85 (3), 1315– Soc. 18 (04), 773–780.
1321. Stockmeier, C.A., 1997. Neurobiology of serotonin in depression and suicidea. Ann. N.
Oberndorfer, T.A., Kaye, W.H., Simmons, A.N., Strigo, I.A., Matthews, S.C., 2011. Y. Acad. Sci. 836 (1), 220–232.
Demand-specific alteration of medial prefrontal cortex response during an Stratford, T.R., Kelley, A.E., 1997. GABA in the nucleus accumbens shell participates in
inhibition task in recovered anorexic women. Int. J. Eat. Disord. 44 (1), 1–8. the central regulation of feeding behavior. J. Neurosci. 17 (11), 4434–4440.
Oldershaw, A., DeJong, H., Hambrook, D., Broadbent, H., Tchanturia, K., Treasure, J., Stratford, T.R., Kelley, A.E., 1999. Evidence of a functional relationship between the
Schmidt, U., 2012. Emotional processing following recovery from anorexia nucleus accumbens shell and lateral hypothalamus subserving the control of
nervosa. Eur. Eat. Disord. Rev. 20 (6), 502–509. feeding behavior. J. Neurosci. 19 (24), 11040–11048.
Otake, K., Nakamura, Y., 2000. Possible pathways through which neurons of the Strober, M., 1995. Family-genetic perspectives on anorexia nervosa and bulimia
shell of the nucleus accumbens influence the outflow of the core of the nucleus nervosa. Eat. Disord. Obes. 212–218.
accumbens. Brain Dev. 22, 17–26. Tammela, L.I., Rissanen, A., Kuikka, J.T., Karhunen, L.J., Bergström, K.A., Repo-
Otto, B., Cuntz, U., Fruehau, E., 2001. col. Weight gain decreases elevated plasma Tiihonen, E., et al., 2003. Treatment improves serotonin transporter binding and
concentrations of patients with anorexia nervosa. Eur. J. Endocrinol. 145, 669– reduces binge eating. Psychopharmacology 170 (1), 89–93.
673. Tanaka, M., Naruo, T., Muranaga, T., Yasuhara, D., Shiiya, T., Nakazato, M., et al., 2002.
Palmiter, R.D., 2007. Is dopamine a physiologically relevant mediator of feeding Increased fasting plasma ghrelin levels in patients with bulimia nervosa. Eur. J.
behavior? Trends Neurosci. 30 (8), 375–381. Endocrinol. 146 (6), R1–R3.
Papežová, H., Yamamotova, A., Uher, R., 2005. Elevated pain threshold in eating Tchanturia, K., Campbell, I.C., Morris, R., Treasure, J., 2005. Neuropsychological
disorders: physiological and psychological factors. J. Psychiatr. Res. 39 (4), 431– studies in anorexia nervosa. Int. J. Eat. Disord. 37 (S1), S72–S76.
438. Titova, O.E., Hjorth, O.C., Schiöth, H.B., Brooks, S.J., 2013. Anorexia nervosa is linked
Parker, J.A., Bloom, S.R., 2012. Hypothalamic neuropeptides and the regulation of to reduced brain structure in reward and somatosensory regions: a meta-
appetite. Neuropharmacology 63 (1), 18–30. analysis of VBM studies. BMC Psychiatry 13 (1), 110.
Paulus, M.P., Stein, M.B., 2006. An insular view of anxiety. Biol. Psychiatry 60 (4), Treasure, J., Campbell, I., 1994. The case for biology in the aetiology of anorexia
383–387. nervosa. Psychol. Med. 24 (01), 3–8.
Pepin, G., Stagnitti, K., 2012. Come play with me: an argument to link autism Tsuchida, A., Nonomura, T., Ono-Kishino, M., Nakagawa, T., Taiji, M., Noguchi, H.,
spectrum disorders and anorexia nervosa through early childhood pretend play. 2001. Acute effects of brain-derived neurotrophic factor on energy expenditure
Eat. Disord. 20 (3), 254–259. in obese diabetic mice. Int. J. Obes. Relat. Metab. Disord. 25 (9) .
Philipp, E., Pirke, K.-M., Kellner, M.B., Krieg, J.-C., 1991. Disturbed cholecystokinin Uher, R., Treasure, J., 2005. Brain lesions and eating disorders. J. Neurol. Neurosurg.
secretion in patients with eating disorders. Life Sci. 48 (25), 2443–2450. Psychiatry 76 (6), 852–857.
Pietrini, F., Castellini, G., Ricca, V., Polito, C., Pupi, A., Faravelli, C., 2011. Functional Uher, R., Brammer, M.J., Murphy, T., Campbell, I.C., Ng, V.W., Williams, S.C., Treasure,
neuroimaging in anorexia nervosa: a clinical approach. Eur. Psychiatry 26 (3), J., 2003. Recovery and chronicity in anorexia nervosa: brain activity associated
176–182. with differential outcomes. Biol. Psychiatry 54 (9), 934–942.
Prince, A.C., Brooks, S.J., Stahl, D., Treasure, J., 2009. Systematic review and meta- Uher, R., Murphy, T., Brammer, M.J., Dalgleish, T., Phillips, M.L., Ng, V.W., Andrew, C.
analysis of the baseline concentrations and physiologic responses of gut M., Williams, S.C., Campbell, I.C., Treasure, J., 2004. Medial prefrontal cortex
hormones to food in eating disorders. Am. J. Clin. Nutr. 27056 ajcn. activity associated with symptom provocation in eating disorders. Am. J.
Ribases, M., Gratacos, M., Armengol, L., De Cid, R., Badia, A., Jimenez, L., et al., 2003. Psychiatry 161 (7), 1238–1246.
Met66 in the brain-derived neurotrophic factor (BDNF) precursor is associated Usuda, I., Tanaka, K., Chiba, T., 1998. Efferent projections of the nucleus accumbens
with anorexia nervosa restrictive type. Mol. Psychiatry 8 (8), 745–751. in the rat with special reference to subdivision of the nucleus: biotinylated
Robbins, T.W., Gillan, C.M., Smith, D.G., de Wit, S., Ersche, K.D., 2012. Neurocognitive dextran amine study. Brain Res. 797 (1), 73–93.
endophenotypes of impulsivity and compulsivity: towards dimensional Van Den Pol, A., 2003. Weighing the role of hypothalamic feeding
psychiatry. Trends Cogn. Sci. 16 (1), 81–91. neurotransmitters. Neuron 40 (6), 1059–1061.
Rolls, E.T., 2006. Brain mechanisms underlying flavour and appetite. Phil. Trans. R. Vitousek, K., Manke, F., 1994. Personality variables and disorders in anorexia
Soc. B : Biol. Sci. 361 (1471), 1123–1136. nervosa and bulimia nervosa. J. Abnorm. Psychol. 103 (1), 137.
Rose, M., Frampton, I., Lask, B., 2012. A case series investigating distinct Vocks, S., Herpertz, S., Rosenberger, C., Senf, W., Gizewski, E.R., 2011. Effects of
neuropsychological profiles in children and adolescents with anorexia nervosa. gustatory stimulation on brain activity during hunger and satiety in females
Eur. Eat. Disord. Rev. 20 (1), 32–38. with restricting-type anorexia nervosa: an fMRI study. J. Psychiatr. Res. 45 (3),
Sanislow, C.A., Pine, D.S., Quinn, K.J., Kozak, M.J., Garvey, M.A., Heinssen, R.K., et al., 395–403.
2010. Developing constructs for psychopathology research: research domain Volkow, N., Wang, G., Fowler, J., Tomasi, D., Baler, R., 2012. Food and drug reward:
criteria. J. Abnorm. Psychol. 119 (4), 631. overlapping circuits in human obesity and addiction. Brain Imaging in
Saxena, S., 2003. Neuroimaging and the pathophysiology of obsessive compulsive Behavioral Neuroscience. Springer, pp. 1–24.
disorder. Neuroimaging in Psychiatry. Martin Dunitz, London (UK), pp. 191–224. Wagner, A., Aizenstein, H., Venkatraman, V.K., Fudge, J., May, J.C., Mazurkewicz, L., et
Scherag, S., Hebebrand, J., Hinney, A., 2010. Eating disorders: the current status of al., 2007. Altered reward processing in women recovered from anorexia
molecular genetic research. Eur. Child Adolesc. Psychiatry 19 (3), 211–226. nervosa. Am. J. Psychiatry 164 (12), 1842–1849.
Schulte-Rüther, M., Mainz, V., Fink, G.R., Herpertz-Dahlmann, B., Konrad, K., 2012. Wagner, A., Aizenstein, H., Mazurkewicz, L., Fudge, J., Frank, G.K., Putnam, K., et al.,
Theory of mind and the brain in anorexia nervosa: relation to treatment 2008. Altered insula response to taste stimuli in individuals recovered from
outcome. J. Am. Acad. Child Adolesc. Psychiatry 51 (8), 832–841 (e811). restricting-type anorexia nervosa. Neuropsychopharmacology 33 (3), 513–523.
Schwartz, G.J., 2006. Integrative capacity of the caudal brainstem in the control of Walsh, B.T., Devlin, M.J., 1995. Pharmacotherapy of bulimia nervosa and binge eating
food intake. Phil. Trans. R. Soc. B : Biol. Sci. 361 (1471), 1275–1280. disorder. Addict. Behav. 20 (6), 757–764.
Schweiger, U., Warnhoff, M., Pahl, J., Pirke, K.M., 1986. Effects of carbohydrate and Walsh, B.T., Devlin, M.J., 1998. Eating disorders: progress and problems. Science 280
protein meals on plasma large neutral amino acids, glucose, and insulin plasma (5368), 1387–1390.
levels of anorectic patients. Metab.: Clin. Exp. 35 (10), 938–943. Walsh, B.T., Kaplan, A.S., Attia, E., Olmsted, M., Parides, M., Carter, J.C., et al., 2006.
Shafran, R., Fairburn, C.G., Robinson, P., Lask, B., 2004. Body checking and its Fluoxetine after weight restoration in anorexia nervosa: a randomized
avoidance in eating disorders. Int. J. Eat. Disord. 35 (1), 93–101. controlled trial. JAMA 295 (22), 2605–2612.
Simansky, K.J., 1995. Serotonergic control of the organization of feeding and satiety. Wang, G.J., Geliebter, A., Volkow, N.D., Telang, F.W., Logan, J., Jayne, M.C., et al., 2011.
Behav. Brain Res. 73 (1), 37–42. Enhanced striatal dopamine release during food stimulation in binge eating
Small, D., 2006. Central gustatory processing in humans. In Taste and Smell, Vol. 63. disorder. Obesity 19 (8), 1601–1608.
Karger Publisher, pp. 191–220. Wellman, P.J., 2000. Norepinephrine and the control of food intake. Nutrition 16
Small, D.M., 2009. Individual differences in the neurophysiology of reward and the (10), 837–842.
obesity epidemic. Int. J. Obes. 33, S44–S48. Wildes, J.E., Marcus, M.D., 2013. Incorporating dimensions into the classification of
Solinas, M., Justinova, Z., Goldberg, S.R., Tanda, G., 2006. Anandamide eating disorders: three models and their implications for research and clinical
administration alone and after inhibition of fatty acid amide hydrolase (FAAH) practice. Int. J. Eat. Disord. 46 (5), 396–403.
increases dopamine levels in the nucleus accumbens shell in rats. J. Neurochem. Wildes, J.E., Marcus, M.D., 2015. Application of the Research Domain Criteria (RDoC)
98 (2), 408–419. framework to eating disorders: emerging concepts and research. Curr.
Stanley, B.G., Willett, V.L., Donias, H.W., Ha, L.H., Spears, L.C., 1993. The lateral Psychiatry Rep. 17 (5), 1–10.
hypothalamus: a primary site mediating excitatory amino acid-elicited eating. Will, M., Franzblau, E., Kelley, A., 2003. Nucleus accumbens m-opioids regulate
Brain Res. 630 (1), 41–49. intake of a high-fat diet via activation of a distributed brain network. J. Neurosci.
Stanley, B., Willett, V., Donias, H., Dee, M., Duva, M., 1996. Lateral hypothalamic 23 (7), 2882–2888.
NMDA receptors and glutamate as physiological mediators of eating and weight Williams, C.M., Kirkham, T.C., 2002. Observational analysis of feeding induced by D
control. Am. J. Physiol.-Regul. Integr Comp. Physiol. 270 (2), R443–R449. 9-THC and anandamide. Physiol. Behav. 76 (2), 241–250.
Stein, D., Kaye, W.H., Matsunaga, H., Myers, D., Orbach, I., Har-Even, D., et al., 2003. Wise, R.A., Rompré, P.-P., 1989. Brain dopamine and reward. Annu. Rev. Psychol. 40
Pain perception in recovered bulimia nervosa patients. Int. J. Eat. Disord. 34 (3), (1), 191–225.
331–336.
100 A. Mishra et al. / Asian Journal of PsychiatryAJP 25 (2017) 91–100

Young, S.N., Gauthier, S., 1981. Effect of tryptophan administration on tryptophan, 5- Zhang, M., Gosnell, B.A., Kelley, A.E., 1998. Intake of high-fat food is selectively
hydroxyindoleacetic acid and indoleacetic acid in human lumbar and cisternal enhanced by muopioid receptor stimulation within the nucleus accumbens. J.
cerebrospinal fluid. J. Neurol. Neurosurg. Psychiatry 44 (4), 323. Pharmacol. Exp. Ther. 285 (2), 908–914.
Zahm, D.S., 2000. An integrative neuroanatomical perspective on some subcortical Zhang, M., Balmadrid, C., Kelley, A.E., 2003. Nucleus accumbens opioid, GABaergic,
substrates of adaptive responding with emphasis on the nucleus accumbens. and dopaminergic modulation of palatable food motivation: contrasting effects
Neurosci. Biobehav. Rev. 24 (1), 85–105. revealed by a progressive ratio study in the rat. Behav. Neurosci. 117 (2), 202.
Zald, D.H., Kim, S.W., 1996. Anatomy and function of the orbital frontal cortex: i. Zheng, H., Corkern, M., Stoyanova, I., Patterson, L.M., Tian, R., Berthoud, H.-R., 2003.
Anatomy, neurocircuitry, and obsessive-compulsive disorder. J. Appetite-inducing accumbens manipulation activates hypothalamic orexin
Neuropsychiatry Clin. Neurosci. 8 (3), 249–261. neurons and inhibits POMC neurons. J. Physiol.-Regulatory Integrative and
Zhang, M., Kelley, A.E., 1997. Opiate agonists microinjected into the nucleus Comparative Physiol. 284 (6), R1436–R1444.
accumbens enhance sucrose drinking in rats. Psychopharmacology 132 (4), de Castro, J.M., Plunkett, S., 2002. A general model of intake regulation. Neurosci.
350–360. Biobehav. Rev. 26 (5), 581–595.
Zhang, M., Kelley, A., 2000. Enhanced intake of high-fat food following striatal mu-
opioid stimulation: microinjection mapping and fos expression. Neuroscience
99 (2), 267–277.

You might also like