You are on page 1of 15

At the Cutting Edge

Neuroendocrinology 2018;106:292–306 Received: June 19, 2017


Accepted after revision: October 21, 2017
DOI: 10.1159/000484525
Published online: October 24, 2017

An Evolutionary Genetic Perspective of


Eating Disorders
Alexandra J. Mayhew a Marie Pigeyre a, b Jennifer Couturier c David Meyre a, d
       

a Department
of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada;
b Department
of Nutrition, Lille University, Lille, France; Departments of c Psychiatry and Behavioral Neurosciences,
 

d
and Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
 

Keywords ping characteristics of the subtypes of AN (binge/purge and


Eating disorders · Binge eating disorder · Anorexia restrictive), BN, and BED. For instance, mutations/structural
nervosa · Bulimia nervosa · Heritability · Molecular gene variants in the same gene causing opposite effects or
genetics · Evolution mutations in nearby genes resulting in partial disequilibrium
for the genes causing AN (restrictive) and BED may explain
the overlap of phenotypes seen in AN (binge/purge).
Abstract © 2017 S. Karger AG, Basel
Eating disorders (ED) including anorexia nervosa (AN), buli-
mia nervosa (BN), and binge eating disorder (BED) affect up
to 5% of the population in Western countries. Risk factors for Introduction
developing an ED include personality traits, family environ-
ment, gender, age, ethnicity, and culture. Despite being The Diagnostic and Statistical Manual of Mental Dis-
moderately to highly heritable with estimates ranging from orders, version 5 (DSM-5) describes three eating disor-
28 to 83%, no genetic risk factors have been conclusively ders (EDs): anorexia nervosa (AN), bulimia nervosa
identified. Our objective was to explore evolutionary theo- (BN), and binge eating disorder (BED). EDs affect around
ries of EDs to provide a new perspective on research into 5% of the population in Western countries, and are more
novel biological mechanisms and genetic causes of EDs. We prevalent in females than males [1]. EDs develop from a
developed a framework that explains the possible interac- complex interaction of psychological risk factors, socio-
tions between genetic risk and cultural influences in the de- cultural influences, and biological and genetic predisposi-
velopment of ED. The framework includes three genetic pre- tions [2, 3]. Although heritability estimates range from 28
disposition categories (people with mainly AN restrictive to 78% for AN, 30 to 83% for BN [4], and 41 to 57% for
gene variants, people with mainly BED variants, and people BED [5], there is limited evidence of robust associations
with gene variants predisposing to both diseases) and a bi- between EDs and candidate genes or associations detect-
nary variable of either the presence or absence of pressure ed through genome-wide association studies (GWAS) or
to be thin. We propose novel theories to explain the overlap- whole-exome/genome sequencing [6]. The high herita-

© 2017 S. Karger AG, Basel David Meyre


Department of Health Research Methods, Evidence, and Impact
McMaster University, 1280 Main St.
E-Mail karger@karger.com
Hamilton, ON L8S 4K1 (Canada)
www.karger.com/nen
E-Mail meyred @ mcmaster.ca
bility and ethnic-dependent prevalence of EDs suggests a Prevalence of EDs
possible influence of natural selection; however, theories The lifetime prevalence of AN is 0.3–3.0% for females
examining EDs from an evolutionary perspective are lim- and 0.24–0.30% for males with a peak incidence between
ited. The purpose of this article is to investigate the epi- 10 and 19 years of age [1, 9–21]. BN is similarly more
demiological, genetic, and biological evidence about ED prevalent in females, with 0.88–4.6% of females suffering
and to use this information to inform new evolutionary from the disease compared to 0.10–1.5% of males [1, 10,
genetic theories of ED. These theories can be used to stim- 11, 13, 15, 16, 22, 23]. There is greater variability in the
ulate research into novel biological mechanisms and ge- estimates of peak incidence of BN, which in part could be
netic causes of ED. A qualitative review of the literature because of the use of different age cut-offs used in studies
related to the epidemiology (diagnostic criteria, preva- as well as the relatively recent changes in diagnostic cri-
lence, time trends, risk factors), neurotransmitters, and teria. The peak incidence is estimated to be as low as 10–
genetic determinants (heritability, linkage studies, candi- 20 years of age, or as high as 25–29 years [11, 12, 14, 16,
date gene studies, GWAS, and next-generation sequenc- 22, 24]. The lifetime prevalence of BED is higher in fe-
ing) of EDs was conducted (November 2016, no restric- males (2.5–3.5%) compared to males (1.5–2.0%) [1, 24–
tion on publication date of articles) using PubMed, 27]. Three studies have found that the incidence of BED
Google Scholar, and the MEDLINE databases. is relatively equally distributed across the lifespan [24, 28,
29], while another indicated that the average age at onset
of BED ranges from 18.3 to 25.4 years [10].
Epidemiology of EDs Ethnicity and culture may alter the prevalence of ED.
The prevalence of AN in non-Western countries is estimat-
Diagnostic Criteria of EDs ed to be 0.002–0.9% compared to 0.3–3.0% for females and
People with AN are significantly underweight, have an 0.24–0.30% in males in Western countries [30]. A study in
intense fear of gaining weight or becoming fat, a distur- Curacao found the incidence of AN is 1.82 cases per 100,000
bance in body image, a heavy emphasis on body shape for persons per year without any cases in Black females [31],
self-evaluation, and/or denial of the seriousness of their and in the United States, 0% of Black females were diag-
current weight loss [7]. There are two subtypes of AN: the nosed with AN in contrast to 1.5% of White females [32].
restricting type (ANR), involving food restriction with- Also in the United States, the lifetime prevalence of AN in
out binging or purging, and the binge-eating/purging the adult population was 0.15% for African-Americans and
type (ANBP), involving overeating and/or purging. Sim- 0% for Caribbean Blacks [33]. BN is also less prevalent in
ilar to AN, people with BN place a strong emphasis on non-Western countries compared to Western countries. A
body shape for self-evaluation, but are not underweight. meta-analysis found that there are no studies in non-West-
BN also involves recurrent episodes of binge eating dur- ern countries reporting the presence of BN without expo-
ing which large quantities of food are consumed during a sure to Western ideals [34] and in the United States, 2.3%
discrete period of time. During these episodes, people feel of White females versus 0.4% of Black females received a
a lack of control and are unable to stop eating. After bing- diagnosis of BN [32]. In contrast, BED appears to be more
es, inappropriate compensatory behaviors including self- prevalent in African-Americans and Caribbean Blacks
induced vomiting, laxative misuse, diuretics, enemas, ex- compared to Whites. Some studies conducted in Latino
cessive exercise, or abuse of medications are used to pre- populations suggest that the prevalence for Latinos is also
vent weight gain. This behavior must occur a minimum higher than for Whites [17, 28, 33, 35, 36].
of once a week for 3 months to be diagnosed as BN. Indi- It is unclear if the prevalence of EDs has changed over
viduals with BED do not engage in compensatory behav- time. Most studies indicate that the prevalence of AN,
ior and are at high risk of being obese [7]. Over self-eval- BN, and BED has increased [12, 34, 37]; however, there
uation of shape and weight is not a diagnostic criterion are studies that have observed declines in prevalence [12,
for BED, but can be present in individuals with this dis- 38]. Changes in the prevalence may not reflect true chang-
order [8]. It is noteworthy that some overlapping EDs are es in the number of people with EDs but may instead rep-
described in the DSM-5 under the “Other Specified Feed- resent changes in diagnostic criteria, shifting attitudes
ing or Eating Disorders” but will not be covered in this about mental health disorders, the challenge of identify-
review [7]. ing patients who often try to keep their disease hidden,
differing methodologies [34], and small sample sizes
from groups that are not generalizable.

Genes, Evolution, and Eating Disorders Neuroendocrinology 2018;106:292–306 293


DOI: 10.1159/000484525
Risk Factors of EDs ing, as well as hunger regulation [40, 50, 51]. Disturbanc-
es of brain serotonin activity have been described in
Personality Traits acutely ill as well as long-term recovered patients with
Personality characteristics are associated with EDs, in- both AN and BN [40]. It is hypothesized that a distur-
cluding perfectionism, obsessive compulsive personality, bance of serotonin activity may create vulnerability for
neuroticism, negative emotionality, harm avoidance, low the development of EDs. Alterations in central nervous
self-directedness, low cooperativeness, and features of system serotonin function may contribute to other psy-
avoidant personality disorder [39, 40]. Low novelty seek- chological symptoms associated with EDs, such as the
ing, low emotional responsiveness, decreased pleasure, high prevalence of depressive disorders, the impulsive-
decreased seeking of pleasure, and reduced social sponta- aggressive behavior observed in patients with BN, and the
neity are common characteristics in individuals with the obsessive-compulsive symptomatology observed in AN.
restrictive type of AN, while those with BN tend to be Serotonin activity may also contribute to symptoms such
impulsive, seek out new experiences, and have character- as anxiety, perfectionism, obsessions with symmetry and
istics of borderline personality disorder [39, 40]. BED is exactness, and harm avoidance, which, when coupled
associated with perfectionism and sensation seeking, and with psychosocial influences, make people vulnerable to
both AN and BN are associated with obsessive compul- developing AN [52]. Dysfunction of the neural network
sive personality disorder [39]. Though causality has not involved in reward, punishment, and motivational pro-
been established, psychiatric comorbidities are frequent cesses is also described in ED patients [40, 53]. Several
in all EDs with an estimated prevalence between 31.0 and studies have shown that dopamine system disturbance
88.9% [1, 19, 21, 41]. Depression and anxiety disorders contributes to the development and maintenance of AN
are most prevalent [16, 21, 41–43]; however, bipolar af- and other EDs [54, 55].
fective disorder, obsessive compulsive disorder, and sub-
stance use are also common [21].
Genetic Determinants of EDs
Environmental Exposures
Individuals diagnosed with EDs are more likely to Different strategies have been used to determine the
have experienced abuse or trauma than the general popu- genetic and epigenetic architecture of EDs including
lation. Specifically, sexual abuse has been reported in 20– heritability studies, linkage studies, candidate gene ap-
50% of individuals with AN and BN. Women with EDs proaches, GWAS, next-generation sequencing, candidate
who have suffered from sexual abuse also demonstrate gene and whole-genome methylation studies. Linkage
higher rates of comorbid psychiatric conditions [7]. In- studies use related individuals to identify genomic re-
adequate coping mechanisms are common in those with gions containing genes that predispose individuals to a
disordered eating and may explain an individual’s adop- disease [56]. Candidate gene studies investigate genes
tion of maladaptive eating patterns in response to trauma that have been selected based on their physiological, bio-
[44]. Studies have also noted a higher prevalence of EDs chemical, and functional aspects [57]. GWAS are hypoth-
among athletes, models, dancers, and performers, activi- esis-free study attempts to identify common genetic vari-
ties which emphasize dietary restraint and idealize a thin ants that contribute to disease risk using markers cover-
physique [2]. Family dynamics that involve overprotec- ing the entire genome [58]. Next-generation sequencing
tive, intrusive, controlling, or emotionally unresponsive approaches target rare genetic variants and short dele-
parents, as well as enmeshment of parents and children tion/duplication that contribute to ED etiology. Methyla-
and poor conflict resolution have been associated with tion arrays enable the quantitative interrogation of meth-
EDs, though evidence is limited [45–48]. Having a preoc- ylation sites across the genome at single-nucleotide reso-
cupation with weight in the form of family dieting or re- lution [59].
ceiving criticism about weight, eating, or body shape, is
also associated with the risk of EDs [49]. Heritability of EDs
EDs are moderately to highly heritable. Heritability es-
Neurobiological Regulation timates from twin studies range from 28 to 74% for AN,
Several neural systems have been implicated in the eti- 54 to 83% for BN, and 41 to 57% for BED [5]. Most of the
ology of EDs, including those associated with cognitive individual studies contributing to these estimates have
self-regulatory control, motivation and reward process- wide confidence intervals because of the low statistical

294 Neuroendocrinology 2018;106:292–306 Mayhew/Pigeyre/Couturier/Meyre


DOI: 10.1159/000484525
power caused by relatively small sample sizes due to the include: (a) homeostatic pathways (leptin melanocortin
low prevalence of EDs [5]. The range of estimates also re- pathway genes, i.e. LEP, LEPR, POMC, AGRP, MC4R,
flects that heritability estimates can include just additive BDNF genes, as well as GHRL, FTO genes) and (b) re-
genetic effects or all genetic effects including gene × gene ward-related pathways including central neurotransmis-
interactions and dominant effects [60, 61]. One of the sion of serotonin (i.e., HTR1D, HTR2A, HTR2C, SLC6A4
limitations of the heritability estimates is that the studies genes), dopamine (i.e., DRD2, DRD4, ANKK1 genes),
are predominantly conducted in Europeans. However, a noradrenaline (COMT gene), opioid (OPRD1, OPRM
study conducted in both European American and Afri- genes) and the cannabinoid endogenous system (CNR1
can-American female twins suggests that the contribu- gene). These pathways are relevant to candidate gene
tion of additive genes to BN and BED are similar [62, 63]. studies because healthy eating behavior is achieved
through the balance of homeostatic controls and reward
Linkage Studies processes. Dysregulation of these systems is observed in
Few linkage studies have been performed for ED traits, EDs, but it is currently unknown if alterations to these
and none of them used a logarithm of odds (LOD) score pathways are a cause or an effect [73]. Overall, these stud-
of 3.6 as the threshold for significant linkage [64]. In a ies do not show a conclusive association between a com-
genome-wide linkage analysis of 192 families with at least mon single nucleotide polymorphism (SNP) and EDs
one affected relative pair with AN and related EDs, only [74]. The contribution of common variants associated
a modest evidence of linkage with AN was reported on with body mass index (BMI) variation to EDs has been
chromosome 4 (LOD of 1.80). However, the analysis of recently tested in 3 studies, but the results are conflicting
the subset of patients with the restrictive subtype of AN [75–77]. However, a recent atlas of genetic correlations
led to the identification of a linkage peak close to signifi- across human diseases supports a shared molecular basis
cance on chromosome 1p36-34 (LOD of 3.03 and of 3.45 for AN and obesity [78].
after genotyping additional microsatellite markers) [65].
Devlin et al. [66] developed an original methodology Genome-Wide Association Studies
based on the linkage study of AN incorporating behav- Unsurprisingly, several modestly powered GWAS
ioral covariates. Applying this method to 196 multiplex studies for ED-related traits reported negative findings af-
families, they identified a linkage close to significance ter correcting for multiple testing [79, 80]. A recent nega-
(LOD of 3.46) for AN, obsessionality and drive-for-thin- tive two-stage GWAS meta-analysis for AN in 5,551 cases
ness. The analysis of 308 families with BN led to the iden- and 21,080 controls concluded that the accrual of large
tification of a suggestive peak of linkage on chromosome genotyped AN case-control samples should be an imme-
10p13-12 (LOD of 2.92). When the analysis was restricted diate priority for the field to identify AN-predisposing
to 133 families with BN and self-induced vomiting, the genes [81]. Although there were no genome-wide signifi-
linkage value increased at the same location (LOD of cant hits reported, 72 independent markers with the low-
3.39) [67]. Interestingly, significant linkage for obesity est p values were selected for replication in an indepen-
has been identified in the same chromosomal region [68– dent sample, and 76% of these loci produced results direc-
70]. Though noteworthy, these findings are not statisti- tionally consistent with the discovery sample [82]. Two
cally significant and are from insufficiently powered AN loci have successfully been identified by GWAS. A
studies, increasing the risk of false positive results [71]. study including 692 female AN cases and 2,570 controls
Furthermore, these linkage experiments have not been found a significant association approaching genome wide
confirmed by positional cloning, and no strong predis- significant (p = 2.04 × 10–7) between a variant in the Early
posing genes have emerged to date [4]. B-Cell Factor 1 (EBF1) gene (rs929626). The inactivation
of EBF1 in mice leads to a decrease in circulating leptin
Candidate Gene Studies levels, in line with the observations of very low leptin lev-
Hundreds of candidate gene studies of common vari- els made in human AN patients. In addition, another SNP
ants have been published, but have provided few defini- (rs4704963) in EBF1 showed genome-wide significant in-
tive conclusions [72]. Regardless of the type of ED, most teraction with psychosocial stress on obesity traits [83]. In
candidate gene studies have focused on genes related to a sample of 3,495 AN cases and 10,982 controls, rs4622308
homeostatic control and reward systems due to the hy- near the v-erb-b2 avian erythroblastic leukemia viral on-
pothesized shared roles of these pathways to all ED. The cogene homolog 3 (ERBB3) gene was significantly associ-
main pathways explored in candidate gene studies of ED ated with AN [84]. Further replication studies, larger sam-

Genes, Evolution, and Eating Disorders Neuroendocrinology 2018;106:292–306 295


DOI: 10.1159/000484525
ple sizes, and experiments on expression and function are Epigenetics
required to gain a better understanding of the role of these Epigenetic mechanisms provide a substrate for gene-
genes in AN. Thus far, there have not been any GWAS environment interactions. Starvation is a key clinical fea-
conducted in BN or BED, though a large GWAS in 2,564 ture of patients with AN, and a strong environmental ex-
female twins investigating ED-related phenotypes did not posure can be expected to trigger epigenetic alterations
find any significant associations (p < 10–8) with BN spec- [98]. A main epigenetic mechanism is DNA methylation.
trum, purging via substances, disordered eating behav- Hypermethylation of the alpha-synuclein (SNCA) gene
iors, or AN spectrum [85]. (linked to sensitivity to dietary folate) [99], the atrial na-
triuretic peptide (NPPA) gene, implicated in anxiety, de-
Next-Generation Sequencing pression, and stress responses [100], and the oxytocin re-
A recent whole-exome/-genome sequencing study in ceptor (OXT) gene [101] have been associated with AN.
2 large multigenerational pedigrees affected by multiple Weight loss can also alter methylation and expression of
EDs identified the estrogen-related receptor-α (ESRRA) the pro-opiomelanocortin (POMC) gene, implicated in
and the histone deacetylase 4 (HDAC4) genes as promis- hunger, satiety, and energy homeostasis [102]. AN and
ing candidates to further investigate in additional studies BN subjects showed increased methylation of dopamine
[86]. HDAC4 is expressed in the brain, including the cor- system genes [103]. Taken together, these findings sug-
tical region implicated in EDs [87] and has a known role gest that alterations in DNA methylation can lead to EDs
in synaptic plasticity [88]. These findings support that a by influencing a wide variety of systems implicated in
decrease in ESRRA activity may impact the neuronal dys- behavioral/affective regulation, sensitivity to nutritional
function in ED patients. Additionally, ESRRA has a well- insufficiencies, and body-weight maintenance [99, 104,
established role in mitochondrial regulation of neuronal 105]. Genome-wide methylation profiles in women with
plasticity [89]. Finally, ESRRA induces the expression of AN-restrictive type, and women with AN-binge/purge
both monoamine oxidase A and B, suggesting a potential type compared with normal-weight/normal-eater con-
role in the metabolism of monoamine neurotransmitters trol women, showed higher and less variable global meth-
such as serotonin and dopamine [90]. Moreover, given ylation patterns in AN than controls [59]. Hypermeth­
the female predominance of EDs, it is possible that estro- ylation concerned 11 genes (PRDM16, HDAC4, TNXB,
gen signaling may mediate the risk of EDs by altering FTSJD2, PXDNL, DLGAP2, FAM83A, NR1H3, DDX10,
ESRRA-HDAC4 activity [91]. ARHGAP1, PIWIL1) involved in histone acetylation,
RNA modification, cholesterol storage and lipid trans-
Copy Number Variants port, and dopamine and glutamate signaling [59]. Fur-
Copy number variants (CNV) consist of small dele- ther confirmation of these initial findings is needed at this
tions or duplications in the genome. The only genome- stage. For instance, the hypermethylation of TNXB has
wide CNV study of EDs reported a 1.4-Mb deletion on been recently confirmed by high-throughput DNA meth-
13q12 in 2 AN cases [80]. A genome-wide search for co- ylation analysis in AN [106]. Developmental experiences
py-number variants associated with BMI, found a 600-Kb and early adversity can also impact methylation levels of
deletion on chromosome 16p11.2 associated with a high- neuropsychiatric genes and, in turn, create variations in
ly penetrant form of obesity accompanied by hyperphagia phenotypes related to stress reactivity and mood regula-
and increased ad libitum food intake [92, 93]. SH2B1, one tion [107]. DNA hypermethylation of the glucocorticoid
of the genes deleted, encodes a protein involved in leptin receptor gene (NR3C1) has been observed on individuals
signaling and is the best candidate to account for the hy- with a history of childhood abuse, compared to individu-
perphagic obesity phenotype observed in deletion carri- als who did not report experiences of maltreatment [107].
ers [94]. Deletion carriers also exhibited subjective altera-
tion of reward, eating in the absence of hunger and sen- Gender Influences on Genetic and Environmental Risk
sitivity to external cues [95], but do not present with BN Factors of ED
or BED [96]. Conversely, a duplication at the same chro- Genetic and environmental influences on EDs appear
mosome 16p11.2 locus has been associated with failure to to be moderated by sex and developmental stage. The fe-
thrive in children and underweight in adults, with a high male to male ratio for EDs is between 4:1 and 10:1 [7], and
frequency of selective and restrictive eating behaviors there is a significant increase in EDs in females during
[97]. Unfortunately, all these findings have not yet been puberty. When investigating heritability in males and fe-
replicated. males separately, heritability appears higher in females

296 Neuroendocrinology 2018;106:292–306 Mayhew/Pigeyre/Couturier/Meyre


DOI: 10.1159/000484525
(51–61%) than in males (0–40%) for intentional weight and ANR subtypes may also help detect associations if the
loss, body dissatisfaction, drive for thinness [108–110], SNPs contributing to these AN subtypes are unique. The
and weight concerns [111]. A moderate overlap is also more promising results that emerged from recent epigen-
suggested in the genetic factors influencing ED symp- etic studies deserve further confirmation in additional
toms including body dissatisfaction, drive for thinness samples.
[112], and behaviors such as binge eating [42, 113] be-
tween genders. This moderate overlap may be explained
by sexual dimorphism and different levels of sex hor- Evolutionary Theories of EDs
mones. In Mendelian disorders, an exceptionally high fe-
male to male ratio suggests a higher rate of mutation of A complex and dynamic interplay between personality,
disease-causing genes located on the X chromosome in life experiences, shared and unique environmental fac-
sperm cells or a high rate of X inactivation of one gene tors, epigenetic marks, and genetic predisposition (Fig. 1)
copy or a dominant inheritance mode [114]. It is also is involved in the development of EDs. Examining the
possible that some Y chromosome genes are protective evolutionary theories of EDs can help future gene identi-
against EDs. Together, these may account for the in- fication efforts by focusing on novel candidate genes as
creased genetic risk of EDs in females versus males. The well as guiding the development of biological evidence to
genetic and biological differences between males and fe- support the findings of GWAS. With the increasing use of
males may also interact with environmental factors to al- GWAS approaches, evolutionary theories of EDs can also
ter the risk for developing EDs. Adolescence is also a key help determine new phenotypes to investigate. The lim-
developmental period for EDs. Longitudinal twin studies ited success of genetic studies so far may also result from
have found that the genetic influences on EDs increase a focus on symptoms of the disorders, rather than the
from early to late adolescence [115–117]. One possible causes of them. For AN and BN, abnormal neurotransmit-
explanation is that hormonal changes occur at puberty ter and hormone levels have been widely observed, but
including an increase in estradiol. Estradiol regulates genetic association studies have been inconsistent. If the
gene transcription within the central nervous system altered neurotransmitter and hormone levels are a conse-
[118] and during puberty may activate genetic factors for quence of a physiological adaptation to low food con-
EDs. This theory is supported by recent ED-related ge- sumption rather than one of the causes of the low food
netic variants found to be involved in estrogen metabo- consumption as we propose, then we would not expect
lism [86]. Estrogens are also involved in the regulation of genes involved with the production and reception of those
appetite during the ovarian cycle and between puberty neurotransmitters and hormones to be altered in EDs.
and reproductive senescence in women [119]. Other sex There is also the possibility that hormones relevant to EDs
hormones can influence orosensory capacity, hedonics, are yet to be discovered. Investigating metabolites, tran-
and homeostasis of food intake via a modulation of gas- scription factors, and protein profiles in people with EDs
tric mechanoreception, and hormones (i.e., ghrelin, CCK, and comparing these to levels after recovery of the disease
glucagon-like peptide-1, insulin, amylin, and leptin) as or between episodes as well as with controls may help to
well as central neurochemical signaling (i.e., serotonin, identify novel biomarkers and pathways in the etiology of
dopamine, and leptin melanocortin pathway) [119]. EDs. Using personal characteristics and environmental
factors which have been identified as risk factors for EDs,
Limitations of Genetic and Epigenetic Approaches we investigate previously identified evolutionary theories
Across all methodologies, a common limitation to of EDs as well as discuss novel hypotheses.
identification of genetic variants for EDs is sample size.
Only 2 studies to date have identified SNPs significantly Previously Identified Theories of EDs
associated with AN [83, 84]. To gain the statistical power Previous evolutionary theories of EDs include the sup-
necessary for further genome-wide significant results, re- pression of reproduction theory [120], the sexual compe-
search efforts should focus on the recruitment of large tition theory [121], the adapted to flee famine hypothesis
case-control cohorts. While boosting sample size, future [122], the thrifty gene hypothesis [123], and dual inter-
GWAS efforts should also include BN, BED, as well as vention point model [124]. Briefly, the suppression of re-
analysis of large male cohorts alongside females in order production theory states that AN was a method through
to make meaningful discoveries about the genetic risk which females could suppress reproduction when off-
factors for EDs in men. Differentiating between ANBP spring survival was threatened. Females who were able to

Genes, Evolution, and Eating Disorders Neuroendocrinology 2018;106:292–306 297


DOI: 10.1159/000484525
Biological factors Environmental and social
experiences
Age Family interaction Genetics
Gender Adverse life events Epigenetics
Puberty Sexual abuse
Race Athletic competition
Culture

Psychological factors Behaviors and


perceptions
General psychiatric
disturbance
Negative emotionality
Neuroticism Weight concern
Impulsivity Dieting
Cognitive flexibility Negative body image
Inhibitory control
Perfectionism

Eating disorder

Fig. 1. The relationship between biological factors, environmental and social experience, genetic influences, psy-
chological factors, and behaviors and perceptions in the development of eating disorders.

suppress reproduction until more favorable times are hy- symptoms. The leading theory for these EDs is the thrifty
pothesized to have higher overall reproductive success, genotype hypothesis stating that extra fat stores were pro-
therefore passing on the genes for AN characteristics tective because they played a role in helping to avoid mal-
[120]. The sexual competition theory suggests that AN nutrition, regulate reproduction, and allow for survival
was a method through which females could attract poten- during variations in the energy supply [123]. An elabora-
tial high-quality partners by maintaining a nubile shape tion on the set point theory called the dual intervention
and consequently increase their reproductive success point model may also explain excessive food consump-
[121]. The adapted to flee famine hypothesis explains a tion. This theory states that the body has upper and lower
possible adaptive advantage to what are now known as limits for weight, which when exceeded triggers physio-
symptoms of AN. It proposes that food restricting behav- logical adaptations. The lower limit is set at the minimal
ior, hyperactivity, and denial of seriousness of weight loss weight necessary to avoid starvation while the upper
previously enabled migration during periods of local food weight previously was set to limit predation. As humans
insecurity allowing individuals to reach more food-se- have evolved and predatory pressures have been reduced,
cure areas. Individuals who survived the journey would it is theorized that the genes coding for a higher upper
be able to later reproduce explaining why the genes may limit have become more common, and fewer people ex-
have been passed on [122]. The previous theories provide perience the physiological adaptations such as reducing
potential explanations for AN but not for BN or BED caloric intake, that prevent weight gain [124]. These exist-

298 Neuroendocrinology 2018;106:292–306 Mayhew/Pigeyre/Couturier/Meyre


DOI: 10.1159/000484525
ing theories are not widely accepted and though they pro- Accumulation of Independent Genes Leading to
vide interesting perspectives on the potential historical Binge Eating and ED
development of EDs, their applicability is limited espe- Accounting for the limitations of the previous theory
cially considering that most people with BN are not over- explaining the coexistence of binge/purging behavior with
weight [125]. AN, we hypothesize that independent genes may lead to
the binge/purging behavior seen in BN and the extreme
Novel Theories of EDs weight loss seen in AN. Through random segregation
The evolutionary theories of EDs clearly distinguish within populations, some people may cumulate both the
between AN and the other two EDs, BN and BED. How- genes predisposing to AN and the genes predisposing to
ever, the disorders have a significant amount of overlap. BN. This theory is unlikely because both AN and BN are
The diagnostic criteria for ANR includes weighing less relatively rare diseases. Using our estimated prevalence of
than one should and consuming a limited amount of cal- 0.3–3.0% for AN and 0.88–4.6% for BN in females, the
ories per day, while ANBP replaces the limited calories ANBP subtype should be between 0.0026 and 0.138%,
criteria with cycles of binging and purging [7]. ANBP is while the actual prevalence of ANBP is somewhere be-
very similar to BN, except that individuals with BN are tween 0.15 and 1.5% of women based on half of the people
not underweight. There is a significant amount of cross- with AN having the binge/purge subtype [20]. Therefore,
over between those with ANR, ANBP, and BN. In 7 years we can conclude that because the actual prevalence of
of follow-up, 49% of women initially diagnosed with AN ANBP is higher than would be expected from accumula-
crossed over between AN subtypes, 34% crossed over tion of independent genes leading to BN and AN, the genes
from AN to BN, and 18% of women initially diagnosed associated with the individual diseases are either in linkage
with BN crossed over to AN [20]. Below, we propose the- disequilibrium (i.e., nonrandom combination of alleles at
ories as to why the traits seen in BN, ANR, and ANBP different loci which could result from evolutionary selec-
overlap from a genetic perspective. tion, population ancestry history, and genetic linkage), or
the same genes are causing the two different diseases.
Binge Eating as Protection against ED
The denial of nutrients to the body seen in ANR could Mutations/Structural Gene Variations with Opposite
switch on a pathway encouraging the consumption of cal- Effects in the Same Gene Lead to ED or ED
ories, explaining the binging behavior seen in people with Mutations or structural variants such as deletion/du-
the binge/purge subtype of AN. This fits with the adapted plication, gain of function/loss of function, gain of ex-
to flee famine hypothesis as people should resume eating pression/loss of expression in the same genes may lead to
when food is available again. The cross-over of patients AN and binging behavior. Genetic correlation analyses,
from the subcategories of AN supports this theory when conducted from GWAS for BMI and AN, provided evi-
considering that more people cross over from the restric- dence for a shared polygenic etiology between these two
tive subtype to the binge/purge subtype than the contrary traits, such as overall genetic determinants for higher
[20]. Approximately 25–30% of people receiving treat- BMI decreases risk of AN [78]. However, this study does
ment for BN were previously diagnosed with AN, while not exclude that some of BMI-increasing variants can also
only 5% of people initially diagnosed with BN were later be associated with higher risk of AN, especially when
diagnosed with either subtype of AN [126]. The restric- stressed by environmental changes. This ties in with the
tive behavior seen in ANR may be difficult to maintain evolutionary theories as both the genes allowing for the
and consequently cross over to ANBP when the drive to consumption of large amounts of food when available
eat becomes overwhelming. The cross-over of individuals and the ability to be highly functioning with minimal ca-
from ANBP to BN shows that the binging behavior may loric intake may have been historically positively selected
slowly lead to weight gain, as the only diagnostic differ- for under different environmental conditions and people
ence between ANBP and BN is that people with AN weigh with both types of mutations may have been favored [122,
significantly less than ideal [7]. However, this theory ig- 123, 127–130]. In the past, people possessing both genet-
nores that binge eating is seen outside of the context of ic characteristics may not have suffered from any negative
AN in both BN and BED and that some people with ANR side effects allowing for the transmission of the genes on
do not cross over to ANBP during the duration of their to future generations. It is reasonable to think that the
disease. mixed phenotypes (i.e., ANBP) are now only problem-
atic in the obesogenic environment. Weight concerns are

Genes, Evolution, and Eating Disorders Neuroendocrinology 2018;106:292–306 299


DOI: 10.1159/000484525
Table 1. Framework for genetic and environmental predisposition to eating disorders

Genes predisposing to Genes predisposing to Genes predisposing to


anorexia nervosa anorexia nervosa (restrictive binge eating
(restrictive subtype) subtype) and genes
predisposing to binge eating

Presence of pressure Anorexia nervosa Anorexia nervosa (binge/ Bulimia nervosa


to be thin (restrictive subtype) purge subtype)
Absence of pressure No disease – unless Binge eating disorder Binge eating disorder
to be thin anorexia nervosa is
triggered by factors
other than weight loss

a relatively modern concept and dieting to lose weight serotonin transporter and the norepinephrine transport-
could trigger the metabolic changes allowing for survival er genes [138] as well as the monoamine oxidase A genes
despite low caloric consumption. However, if the same in AN [139], and between the dopaminergic genes
genes are also causing binging behavior, individuals may (DRD2/DRD4 and CMOT/DAT1), influencing both eat-
purge in order to balance the conflicting biological drives ing- and personality-related psychopathology [140].
to survive with minimal caloric consumption and to take
advantage of the food available to them. The possibility of
a single gene having both loss of function and gain of A Framework for EDs Integrating Genetic
function mutations is supported by recent findings on Evolutionary Theories and Epidemiological Evidence
MC4R and the 16p locus where loss of function muta-
tions/deletions lead to hyperphagic obesity while gain of We combined the evolutionary perspective of EDs
function mutations/duplications lead to leanness and se- with the possibility of the genetic overlap causing mixed
lective/restrictive eating behaviors [93, 97, 131–133]. phenotypes, and the epidemiological evidence of differ-
However, several studies have reported contradictory re- ences in prevalence of EDs in different ethnicities and cul-
sults on the association between binge eating behavior tures to create a framework suggesting under which con-
and loss of function variants in MC4R [134–136]; and ditions each ED will occur (Table 1).
gain of function variants in MC4R are not more frequent
in AN patients than in the general population [137]. An- Genetic Component
alyzing whole-genome SNP array and whole-exome se- The genes predisposing individuals to ANR and to
quencing data and searching for complex patterns of mir- binge eating are likely to overlap because they are in par-
ror CNVs or enrichment in loss of function and gain of tial linkage disequilibrium, either because mutations/
function mutations in ANBP cases may identify promis- structural variants within the same gene have opposite
ing candidate genes that may be further validated in ANR, functional effects or because the genes are located close to
BN, or BED cases. each other. Therefore, a greater number of people would
have characteristics of both diseases than would be ex-
Mutations in Nearby Genes Result in Partial Linkage pected if the gene variants were independent. Thus, 3 ge-
Disequilibrium of ED and Binge Eating Traits netic predisposition categories were created, people with
Similar to the theory that the same genes may contain mainly ANR gene variants, people with mainly binge eat-
loss of function/gain of function mutations causing the ing gene variants, and people with gene variants predis-
cross-over of traits seen in EDs is the theory that the genes posing to both diseases. There are other genetic factors
coding for the different characteristics of EDs are in genes that contribute to the risk of developing EDs not captured
located near each other and in partial linkage disequilib- in the model such as epigenetics, gene by gene interac-
rium. The co-occurrence of two distinct phenotypes be- tions, gene by environment interactions in addition to the
cause of the linkage disequilibrium of mutations located pressure of being thin, and any potential genetic factors
in different genes has been suggested in few studies on which contribute to the predisposition of being more sen-
EDs. Epistatic interactions have been found between the sitive to the pressure of being thin.

300 Neuroendocrinology 2018;106:292–306 Mayhew/Pigeyre/Couturier/Meyre


DOI: 10.1159/000484525
Pressure to Be Thin Component sion shows was associated with disordered eating habits
Body dissatisfaction is one of the most consistent and such as self-induced vomiting [147]. The fact that White
robust risk and maintenance factors for EDs [141]. The women are more concerned about weight and have lower
sociocultural model of EDs [2] asserts that exposure to weight ideals in comparison to Black women [143–145]
the Western concept of the ideal body type, often via mag- may explain why White women in the United States have
azines, television, and the internet, promotes internaliza- a higher prevalence of BN than Black women [32]. These
tion of a thin body ideal. Body dissatisfaction ensues studies suggest that the culture may play an important role
when individuals evaluate their own body size negatively in the development of BN by causing an idealization of a
because it is thought to vary from the ideal. Subsequently, thin body type which drives people to use inappropriate
elevated BMI and increased awareness of body size have compensatory methods after binging episodes.
been linked to the onset of dieting and body dissatisfac- Though there is a modest quantity of evidence, studies
tion, both of which being prominent risk factors for EDs in the United States have consistently shown that the
[2, 141, 142]. prevalence of BED is higher in African-American and Ca-
ribbean Blacks compared to Whites [17, 28, 33, 35, 36].
Ethnicity and Culture The higher prevalence of BED in Blacks compared to
Based on the epidemiological evidence, it appears that Whites may be explained by the lack of weight preoccu-
the racial/ethnic and cultural origins of an individual may pation. Even within the BED population, White females
be an important driver of the desire to be thin. However, are significantly more concerned about eating, dietary re-
because the terms ethnicity, race, and culture are often straint, body shape, and weight compared to Black fe-
used interchangeably and because of the strong associa- males, despite on average being less overweight [143]. Be-
tion between biological backgrounds versus cultural cause a smaller proportion of African-Americans have
backgrounds in many people, it is impossible to untangle weight concerns, the population may be less likely to en-
the genetic versus social contributions of race/ethnicity/ gage in purging behavior compared to their White Amer-
culture. Despite these challenges, it is well established that ican counterparts. Therefore, they receive a diagnosis of
White women are more concerned about weight and have BED rather than BN. In line with these data, a study found
lower weight ideals in comparison to Black women [143– that when weight preoccupation was removed as a crite-
145]. These concerns about weight may explain the high- rion, the prevalence of BN in African-Americans and Ca-
er prevalence of AN in Western countries as well as White ribbean Blacks increased [33].
individuals compared to African-Americans and Carib-
bean Blacks [31–33]. Interestingly, when preoccupation Biological Factors
with weight is removed as a diagnostic criteria for AN, Biological factors such as age, gender, and puberty sta-
there was an increase in prevalence of AN of 0.21% in the tus may also contribute to the risk of feeling pressure to
African-American population, though there was no in- be thin. Across all 3 EDs, the prevalence is higher in fe-
crease in the Caribbean Black population [33]. Similarly, males than in males. In females, puberty is a risk period
the prevalence of AN is very similar in Western and non- for the development of AN and BN which is at least par-
Western countries when weight concerns were removed tially mediated by the effects of estrogen. Girls that reach
as a diagnostic criteria [34, 145]. puberty earlier than their peers or are more advanced de-
BN is also less prevalent in non-Western countries velopmentally are at the highest risk. The relationship be-
compared to Western countries, and there are not any tween puberty status in males and EDs is less clear [148].
studies in non-Western countries reporting the presence The peak incidence of AN is between 10 and 19 years,
of BN without exposure to Western ideals [34]. This sug- which coincidences with puberty. The peak incidence of
gests that the culturally derived concept of being thin may BN is less clear, but has been estimated to be between 10
be an important driver of the disease. Supporting this the- and 20 years by some studies and up to 25–29 years in
ory, a study of the Latino-American population found that others. There are too few studies to adequately assess the
recent immigrants have the lowest rates of BN while those relationship between puberty status or age with BED.
who have resided in the United States for 70% or more of
their lives have the highest rates [28]. Similarly, a study of Personality, Environmental, and Neurobiological
Fijian adolescent and adult females found that accultura- Factors
tion to Western ideals was associated with increased body Environmental factors such as family interaction, ad-
shape concerns [146] and watching Westernized televi- verse life events, sexual abuse, and athletic competition

Genes, Evolution, and Eating Disorders Neuroendocrinology 2018;106:292–306 301


DOI: 10.1159/000484525
are all associated with EDs and have been shown to be as- the inability to regulate body weight despite the drive for
sociated with an increase in the desire to be thin. The re- thinness in BN. Finally, stepping beyond the diagnostic
lationship between personality characteristics and the de- criteria of full-phenotype EDs, the important ED-related
sire to be thin have not been established. However, it is phenotypes (e.g., highest and lowest illness-related BMIs,
conceivable that traits such as perfectionism and obses- anxiety, anhedonia, obsessionality, impulsivity, etc.)
sive personality characteristics may be important drivers could be explored in a cross-disorder manner in order to
of idealizing a thin body type. Neurobiological differenc- maximize sample size and explore the possibility of com-
es relating to cognitive self-regulatory control, motiva- mon genetic etiology for these overlapping phenotypes
tion, reward processing, and hunger regulation between across psychiatric diagnoses [72, 150].
people may also drive the desire to be thin. For example,
serotonin is thought to contribute to the development of
anxiety and perfectionism. Certain personality pheno- Conclusions
types such as depression and anxiety are common to all
ED [16, 21, 41–43], while other characteristics are more In this review, we have provided a summary of the lit-
frequently found in specific EDs. For example, low nov- erature on the epidemiological and genetic origins of
elty seeking and decreased pleasure are common in the EDs. Using this evidence, we have developed a novel the-
restrictive subtype of AN, impulsivity and novelty seek- oretical framework which provides a potential explana-
ing are common to BN, and BED is associated with per- tion for the overlap of different types of EDs. This frame-
fectionism [39, 40]. These underlying phenotypes have work provides a simplified view of the interaction of the
genetic contributions. Different combinations of these potential genetics of EDs and the environment, specifi-
phenotypes and underlying genetic predispositions may cally the pressure to be thin. We hope that this framework
also contribute to overlapping characteristics between the will stimulate research looking at the evolutionary roots
ED types. of EDs and ED-predisposing genes. With recent techno-
logical advances (high-throughput genotyping and se-
quencing) the ability to detect genetic associations is im-
Perspectives proving. Understanding the genes underlying EDs may
shed light on new and more effective methods of treating
Even if great progress in genetics and epigenetics have these disorders.
unveiled the pathophysiological architecture of many
complex human diseases, most of the genes predisposing
to EDs still remain to be discovered. The genetic hetero- Disclosure Statement
geneity in different ethnic groups invites collaborations
None of the authors report having a conflict of interest. This
in order to initiate multiethnic international recruitment
research was conducted in the absence of any commercial or finan-
and come up with novel methodologies to exhaustively cial relationships that could be viewed as possible conflicts.
study genetic associations. Methodological developments
such as hypothesis-driven GWAS and methylation stud-
ies relaxing Bonferroni, case-control case design studies, Funding Sources
Bayesian approaches [149], and deep phenotype-based
cohorts [72, 150] may boost the identification of novel Funding was provided by Canada Research Chair. The funder
ED-predisposing genes. Moreover, the real effect of genes had no role in study design, data collection and analysis, decision
to publish, or preparation of the manuscript.
and their interplay with environmental risk factors is not
completely understood yet. Taking advantage of G × E
interactions can lead to original experimental designs,
Author Contributions
such as genome-wide environmental interaction studies
[151], variance prioritization [152], and studying EDs in A.J.M. and D.M. were both responsible for developing the key
specific comorbid psychopathology contexts (such as ob- concepts to be assessed by the review, drafting and revising the
sessionality in AN and impulsivity in BN) [72, 150]. manuscript, providing approval for journal submission and ensur-
Perfectionism and obsessionality are factors in the dis- ing the accuracy and integrity of the work. M.P. and J.C. provided
a critical review of the manuscript. All authors agreed with the final
ordered eating behavior and maintenance of low weight content of the review.
in AN, and impulsivity could be among the reasons for

302 Neuroendocrinology 2018;106:292–306 Mayhew/Pigeyre/Couturier/Meyre


DOI: 10.1159/000484525
References
  1 Hudson JI, Hiripi E, Pope H, Kessler RC: The 17 Marques L, Alegria M, Becker A, Chen C, 31 Hoek H, van Harten PN, Hermans KME,
prevalence and correlates of eating disorders Fang A, Chosak A, et al: Comparative preva- Katzman MA, Matroos GE, Susser ES: The In-
in the National Comorbidity Survey replica- lence, correlates of impairment, and service cidence of Anorexia Nervosa on Curaçao. Am
tion. Biol Psychiatry 2008;61:348–358. utilization for eating disorders across U.S. J Psychiatry 2005;162:748–752.
 2 Striegel-Moore RH: Risk factors for eating ethnic groups: implications for reducing eth- 32 Striegel-Moore RH, Dohm FA, Kraemer HC,
disorders. Ann NY Acad Sci 2007; 817: 98– nic disparities in health care access for eating Taylor CB, Daniels S, Crawford PB, et al: Eat-
109. disorders. Int J Eat Disord 2011;44:412–420. ing disorders in white and black women. Am
  3 Mazzeo SE, Mitchell KS, Bulik CM, Reich- 18 Heaner MK, Walsh BT: A history of the iden- J Psychiatry 2003;160:1326–1331.
born- T, Kendler KS, Neale MC: Assessing the tification of the characteristics eating distur- 33 Taylor JY, Caldwell CH, Baser RE, Matuski N,
heritability of anorexia nervosa symptoms us- bances of bulimia nervosa, binge eating disor- Faison N, Jackson JS: Classification and cor-
ing a marginal maximal likelihood approach. ders and anorexia nervosa. Appetite 2013;65: relates of eating disorders among blacks: find-
Psychol Med 2009;39:463–473. 185–188. ings from the National Survey of American
  4 Slof-Op ’t Landt MCT, van Furth EF, Meulen- 19 Kessler RC, Berglund PA, Chiu WT, Deitz Life. J Health Care Poor Underserved 2013;
belt I, Slagboom PE, Bartels M, Boomsma DI, AC, Hudson JI, Shahly V, et al: The preva- 24:289–310.
et al: Eating disorders: from twin studies to lence and correlates of binge eating disorder 34 Keel PK, Klump KL: Are eating disorders cul-
candidate genes and beyond. Twin Res Hum in the World Health Organization World ture-bound syndromes? Implications for
Genet 2005;8:467–482. Mental Health Surveys. Biol Psychiatry 2013; conceptualizing their etiology. Psychol Bull
 5 Thornton LM, Mazzeo SE, Bulik CM: The 73:904–914. 2003;129:747–769.
heritability of eating disorders: methods and 20 Franko DL, Keshaviah A, Eddy KT, Krishna 35 Nicdao EG, Hong S, Takeuchi DT: Prevalence
current findings. Curr Top Behav Neurosci M, Davis MC, Keel PK, et al: A longitudinal and correlates of eating disorders among
2011;6:141–156. investigation of mortality in anorexia nervosa Asian Americans: results from the National
  6 Hinney A, Volckmar AL: Genetics of eating and bulimia nervosa. Am J Psychiatry 2013; Latino and Asian American Study. Int J Eat
disorders. Curr Psychiatry Rep 2013;15. 170:917–925. Disord 2007;40:S22–S26.
  7 American Psychiatric Association: Diagnos- 21 Meczekalski B, Podfigurna-Stopa A, Katulski 36 Smith DE, Marcus MD, Lewis CE, Fitzgibbon
tic and Statistical Manual of Mental Disor- K: Long-term consequences of anorexia ner- M, Schreiner P: Prevalence of binge eating
ders, ed 5. Arlington, American Psychiatric vosa. Maturitas 2013;75:215–220. disorder, obesity, and depression in a biracial
Association, 2013. 22 Keski-Rahkonen A, Hoek HW, Linna MS, cohort of young adults. Ann Behav Med 1998;
  8 Grilo CM: Why no cognitive body image fea- Raevuori A, Sihvola E, Bulik CM, et al: Inci- 20:227–232.
ture such as overvaluation of shape/weight in dence and outcomes of bulimia nervosa: a na- 37 Hay PJ, Mond J, Buttner P, Darby A: Eating
the binge eating disorder diagnosis? Int J Eat tionwide population-based study. Psychol disorder behaviors are increasing: findings
Disord 2013;46:208–211. Med 2009;39:823–831. from two sequential community surveys in
  9 Fornari VM, Braun DL, Sunday SR, Sandberg 23 Smink FRE, van Hoeken D, Hoek HW: Epi- South Australia. PLoS One 2008;3:e1541.
DE, Matthews M, Chen I, et al: Seasonal pat- demiology of eating disorders: incidence, 38 Keel PK, Heatherton TF, Dorer DJ, Joiner TE,
terns in eating disorder subgroups. Compr prevalence and mortality rates. Curr Psychia- Zalta AK: Point prevalence of bulimia nervo-
Psychiatry 1994;35:450–456. try Rep 2012;14:406–414. sa in 1982, 1992, and 2002. Psychol Med 2006;
10 Favaro A, Ferrara S, Santonastaso P: The 24 Kinzl JF, Traweger C, Trefalt E, Mangweth B, 36:119–127.
spectrum of eating disorders in young wom- Biebl W: Binge eating disorder in females: a 39 Cassin SE, von Ranson KM: Personality and
en: a prevalence study in a general population population-based investigation. Int J Eat Dis- eating disorders: a decade in review. Clin Psy-
sample. Psychosom Med 2003;65:701–708. ord 1999;25:287–292. chol Rev 2005;25:895–916.
11 Hoek HW, van Hoeken D: Review of the prev- 25 Spitzer R., Devlin M, Walsh TB, Hasin DS, 40 Kaye W: Neurobiology of anorexia and buli-
alence and incidence of eating disorders. Int J Wing R, Marcus M, et al: Binge eating disor- mia nervosa. Physiol Behav 2008;94:121–135.
Eat Disord 2003;34:383–396. der: a multisite field trial of the diagnostic cri- 41 Jaite C, Hoffmann F, Glaeske G, Bachmann
12 Currin L, Schmidt U, Treasure J, Jick H: Time teria. Int J Eat Disord 1992;11:191–204. CJ: Prevalence, comorbidities and outpatient
trends in eating disorder incidence. Br J Psy- 26 Spitzer R, Yanovski SZ, Wadden T, Wing R, treatment of anorexia and bulimia nervosa in
chiatry 2005;186:132–135. Marcus MD, Stunkard A, et al: Binge eating German children and adolescents. Eat Weight
13 Hoek HW: Incidence, prevalence and mortal- disorder: its further validation in a multi-site Disord 2013;18:157–165.
ity of anorexia nervosa and other eating dis- study. Int J Eat Disord 1993;13:137–154. 42 Reichborn-Kjennerud T, Bulik CM, Sullivan
orders. Curr Opin Psychiatry 2006; 19: 389– 27 Hay P: The epidemiology of eating disorder PF, Tambs K, Harris JR: Psychiatric and med-
394. behaviors: an Australian community-based ical symptoms in binge eating in the absence
14 van Son GE, van Hoeken D, Bartelds AI, van survey. Int J Eat Disord 1998;23:371–382. of compensatory behaviors. Obes Res 2004;
Furth EF, Hoek HW: Time trends in the inci- 28 Alegria M, Woo M, Cao Z, Torres M, Meng 12:1445–1454.
dence of eating disorders: a primary care X, Striegel-Moore R: Prevalence and corre- 43 Keski-Rahkonen A, Raevuori A, Bulik CM,
study in the Netherlands. Int J Eat Disord lates of eating disorders in Latinos in the Hoek HW, Sihvola E, Kaprio J, et al: Depres-
2006;39:565–569. United States. Int J Eat Disord 2007; 40:S15– sion and drive for thinness are associated with
15 Wade TD, Bergin JL, Tiggemann M, Bulik S21. persistent bulimia nervosa in the community.
CM, Fairburn CG: Prevalence and long-term 29 Grucza RA, Przybeck TR, Cloninger CR: Eur Eat Disord Rev 2013;21:121–129.
course of lifetime eating disorders in an adult Prevalence and correlates of binge eating dis- 44 Mazzeo S, Bulik C: Environmental and genet-
Australian twin cohort. Aust N Z J Psychiatry order in a community sample. Compr Psychi- ic risk factors for eating disorders: what the
2006;40:121–128. atry 2007;48:124–131. clinician needs to know. Child Adolesc Psy-
16 Preti A, de Girolamo G, Vilagut G, Alonso J, 30 Klump KL, Miller KB, Keel PK, McGue M, chiatr Clin North 2009;18:1–15.
de Graaf R, Bruffaerts R, et al: The epidemiol- Iacono WG: Genetic and environmental in- 45 Klump KL, Wonderlich S, Lehoux P, Lilenfeld
ogy of eating disorders in six European coun- fluences on anorexia nervosa syndromes in a LRR, Bulik CM: Does environment matter? A
tries: results of the ESEMeD-WMH project. J population-based twin sample. Psychol Med review of nonshared environment and eating
Psychiatr Res 2009;43:1125–1132. 2001;31:737–740. disorders. Int J Eat Disord 2002;31:118–135.

Genes, Evolution, and Eating Disorders Neuroendocrinology 2018;106:292–306 303


DOI: 10.1159/000484525
46 Kardum I, Gra A, Hudek-Kneževi J: Evolu- 62 Munn-Chernoff M, Grant JD, Agrawal A, Ko- 76 Gervasini G, Gamero-Villarroel C: Discuss-
tionary explanations of eating disorders. Psy- ren R, Glowinski AL, Bucholz KK, et al: Are ing the putative role of obesity-associated
chol Top 2008;17:247–263. there common familial influences for major genes in the etiopathogenesis of eating dis­
47 O’Shaughnessy R, Dallos R: Attachment re- depressive disorder and an overeating-binge orders. Pharmacogenomics 2015; 16: 1287–
search and eating disorders: a review of the eating dimension in both European-Ameri- 1305.
literature. Clin Child Psychol Psychiatry can and African-American female twins? Int 77 Hinney A, Kesselmeier M, Jall S, Volckmar
2009;14:559–574. J Eat Disord 2015;48:375–382. A-L, Föcker M, Antel J, et al: Evidence for
48 Holtom-Viesel A, Allan S: A systematic re- 63 Munn-Chernoff M, Grant JD, Agrawal A, three genetic loci involved in both anorexia
view of the literature on family functioning Sartor CE, Werner KB, Bucholz KK, et al: Ge- nervosa risk and variation of body mass in-
across all eating disorder diagnoses in com- netic overlap between alcohol use disorder dex. Mol Psychiatry 2017;22:192–201.
parison to control families. Clin Psychol Rev and bulimic behaviors in European American 78 Bulik-Sullivan B, Finucane HK, Anttila V,
2014;34:29–43. and African American women. Drug Alcohol Gusev A, Day FR, et al: An atlas of genetic cor-
49 Fairburn CG, Harrison PJ: Eating disorders. Depend 2016;153:335–340. relations across human diseases and traits.
Lancet 2003;361:407–416. 64 Lander E, Kruglyak L: Genetic dissection of Nat Genet 2015;47:1236–1241.
50 Kaye WH, Wierenga CE, Bailer UF, Simmons complex traits: guidelines for interpreting 79 Wang K, Zhang H, Bloss CS, Duvvuri V, Kaye
AN, Bischoff-Grethe A: Nothing tastes as and reporting linkage results. Nat Genet 1995; W, Schork NJ, et al: A genome-wide associa-
good as skinny feels: the neurobiology of an- 11:241–247. tion study on common SNPs and rare CNVs
orexia nervosa. Trends Neurosci 2013; 36: 65 Grice DE, Halmi KA, Fichter MM, Strober M, in anorexia nervosa. Mol Psychiatry 2011;16:
110–120. Woodside DB, Treasure JT, et al: Evidence for 949–959.
51 Wierenga CE, Ely A, Bischoff-Grethe A, Bail- a susceptibility gene for anorexia nervosa on 80 Winham SJ, Cuellar-Barboza AB, McElroy
er UF, Simmons AN, Kaye WH: Are extremes chromosome 1. Am J Hum Genet 2002; 70: SL, Oliveros A, Crow S, Solby CL, et al: Bipo-
of consumption in eating disorders related to 787–792. lar disorder with comorbid binge eating his-
an altered balance between reward and inhi- 66 Devlin B, Bacanu S-A, Klump KL, Bulik CM, tory: a genome-wide association study impli-
bition? Front Behav Neurosci 2014;8:410. Fichter MM, Halmi KA, et al: Linkage analysis cates APOB. J Affect Disord 2014; 165: 151–
52 Jacobi C, Hayward C, de Zwaan M, Kraemer of anorexia nervosa incorporating behavioral 158.
HC, Agras WS: Coming to terms with risk fac- covariates. Hum Mol Genet 2002;11:689–696. 81 Boraska V, Davis OSP, Cherkas LF, Helder
tors for eating disorders: application of risk 67 Bulik CM, Devlin B, Bacanu S-A, Thornton L, SG, Harris J, Krug I, et al: Genome-wide as-
terminology and suggestions for a general Klump KL, Fichter MM, et al: Significant link- sociation analysis of eating disorder-related
taxonomy. Psychol Bull 2004;130:19–65. age on chromosome 10p in families with bu- symptoms, behaviors, and personality traits.
53 Kaye WH, Wierenga CE, Bailer UF, Simmons limia nervosa. Am J Hum Genet 2003;72:200– Am J Med Genet B Neuropsychiatr Genet
AN, Bischoff-Grethe A: Nothing tastes as 207. 2012;159B:803–811.
good as skinny feels: the neurobiology of an- 68 Hager J, Dina C, Francke S, Dubois S, Houari 82 Boraska V, Franklin CS, Floyd JAB, Thornton
orexia nervosa. Trends Neurosci 2013; 36: M, Vatin V, et al: A genome-wide scan for hu- LM, Huckins LM, Southam L, et al: A ge-
110–120. man obesity genes reveals a major susceptibil- nome-wide association study of anorexia ner-
54 O’Hara CB, Campbell IC, Schmidt U: A re- ity locus on chromosome 10. Nat Genet 1998; vosa. Mol Psychiatry 2014;19:1085–1094.
ward-centred model of anorexia nervosa: a 20:304–308. 83 Li D, Chang X, Connolly JJ, Tian L, Liu Y,
focussed narrative review of the neurological 69 Hinney A, Ziegler A, Oeffner F, Wedewardt Bhoj EJ, et al: A genome-wide association
and psychophysiological literature. Neurosci C, Vogel M, Qulftange H, et al: Independent study of anorexia nervosa suggests a risk locus
Biobehav Rev 2015;52:131–152. confirmation of a major locus for obesity on implicated in dysregulated leptin signaling.
55 O’Hara CB, Keyes A, Renwick B, Leyton M, chromosome 10. J Clin Endocrinol Metab Sci Rep 2017;7:3847.
Campbell IC, Schmidt U: The effects of acute 2000;85:2962–2965. 84 Duncan L, Yilmaz Z, Gaspar H, Walters R,
dopamine precursor depletion on the rein- 70 Groves CJ, Zeggini E, Walker M, Hitman GA, Goldstein J, Anttila V, et al: Significant locus
forcing value of exercise in anorexia nervosa. Levy JC, O’Rahilly S, et al: Significant linkage and metabolic genetic correlations revealed in
PLoS One 2016;11:e0145894. of BMI to chromosome 10p in the U.K. popu- genome-wide association study of anorexia
56 Teare MD, Barrett J: Genetic epidemiology lation and evaluation of GAD2 as a positional nervosa. Am J Psychiatry 2017;174:850–858.
2: genetic linkage studies. Lancet 2005; 366: candidate. Diabetes 2006;55:1884–1889. 85 Wade TD, Gordon S, Medland S, Bulik CM,
1036–1044. 71 Li A, Meyre D: Challenges in reproducibility Heath AC, Montgomery GW, et al: Genetic
57 Zhu M, Zhao S: Candidate gene identification of genetic association studies: lessons learned variants associated with disordered eating. Int
approach: progress and challenges. Int J Biol from the obesity field. Int J Obes 2013; 559– J Eat Disord 2013;46:594–608.
Sci 2007;3:420–427. 567. 86 Cui H, Moore J, Ashimi SS, Mason BL, Draw-
58 Bush WS, Moore JH: Chapter 11: Genome- 72 Yilmaz Z, Hardaway JA, Bulik CM: Genetics bridge JN, Han S, et al: Eating disorder pre­
wide association studies. PLoS Comput Biol and epigenetics of eating disorders. Adv Ge- disposition is associated with ESRRA and
2012;8:e1002822. nomics Genet 2015;5:131. HDAC4 mutations. J Clin Invest 2013; 123:
59 Booij L, Casey KF, Antunes JM, Szyf M, Joo- 73 Monteleone P, Maj M: Dysfunctions of leptin, 4706–4713.
ber R, Israël M, et al: DNA methylation in ghrelin, BDNF and endocannabinoids in eat- 87 Darcy MJ, Calvin K, Cavnar K, Ouimet CC:
individuals with anorexia nervosa and in ing disorders: beyond the homeostatic con- Regional and subcellular distribution of
matched normal-eater controls: a genome- trol of food intake. Psychoneuroendocrinol- HDAC4 in mouse brain. J Comp Neurol 2010;
wide study. Int J Eat Disord 2015;48:874–882. ogy 2013;38:312–330. 518:722–740.
60 Wray NR, Visscher PM: Estimating trait her- 74 Trace SE, Baker JH, Peñas-Lledó E, Bulik CM: 88 Sando III R, Gounko N, Pieraut S, Liao L,
itability. Nat Educ 2008;1:29. The genetics of eating disorders. Annu Rev Yates III J, Maximov A: HDAC4 governs a
61 Tenesa A, Haley CS: The heritability of hu- Clin Psychol 2013;9:589–620. transcriptional program essential for snypatic
man disease: estimation, uses and abuses. Nat 75 Brandys MK, Van Elburg AA, Loos RJF, Bau- plasticity and memory. Cell 2010; 151: 821–
Rev 2013;14:139–149. er F, Hendriks J, Van Der Schouw YT, et al: 834.
Are recently identified genetic variants regu-
lating BMI in the general population associ-
ated with anorexia nervosa? Am J Med Genet
B Neuropsychiatr Genet 2010;153:695–699.

304 Neuroendocrinology 2018;106:292–306 Mayhew/Pigeyre/Couturier/Meyre


DOI: 10.1159/000484525
 89 Mattson MP: Mitochondrial regulation of 102 Ehrlich S, Weiss D, Burghardt R, Infante- 114 Neul JL, Kaufmann WE, Glaze DG, Clarke
neuronal plasticity. Neurochem Res 2007; Duarte C, Brockhaus S, Muschler MA, et al: AJ, Leonard H, Bailey MES, et al: Rett syn-
32:707–715. Promoter specific DNA methylation and drome: revised diagnostic criteria and no-
  90 Ren Y, Jiang H, Ma D, Nakaso K, Feng J: Par- gene expression of POMC in acutely under- menclature. Ann Neurol 2010;68:944–950.
kin degrades estrogen-related receptors to weight and recovered patients with anorexia 115 Klump KL, Burt A, McGue M, Iacono W:
limit the expression of monoamine oxidases. nervosa. J Psychiatr Res 2010;44:827–833. Changes in genetic and environmental in-
Hum Mol Genet 2011;20:1074–1083. 103 Frieling H, Rómer KD, Scholz S, Mittelbach fluences on disordered eating between early
  91 Liu D, Zhang Z, Gladwell W, Teng CT: Es- F, Wilhelm J, De Zwaan M, et al: Epigenetic and late adolescence: a longitudinal twin
trogen stimulates estrogen-related receptor dysregulation of dopaminergic genes in eat- study. Psychol Med 2007;45:3249–3258.
α gene expression through conserved hor- ing disorders. Int J Eat Disord 2010;43:577– 116 Silberg JL, Bulik CM: The developmental as-
mone response elements. Endocrinology 583. sociation between eating disorders symp-
2003;144:4894–4904. 104 Saffrey R, Novakovic B, Wade TD: Assessing toms and symptoms of depression and anxi-
  92 Bochukova EG, Huang N, Keogh J, Henning global and gene specific DNA methylation ety in juvenile twin girls. J Child Psychol
E, Purmann C, Blaszczyk K, et al: Large, rare in anorexia nervosa: a pilot study. Int J Eat Psychiatry 2005;46:1317–1326.
chromosomal deletions associated with se- Disord 2014;47:206–210. 117 Culbert KM, Burt A, McGue M, Iacono WG,
vere early-onset obesity. Nature 2010; 463: 105 Tremolizzo L, Conti E, Bomba M, Uccellini Klump KL: Puberty and the genetic diathesis
666–670. O, Rossi MS, Marfone M, et al: Decreased of disordered eating attitudes and behaviors.
  93 Walters RG, Jacquemont S, Valsesia A, de whole-blood global DNA methylation is re- J Abnorm Psychol 2009;118:788–796.
Smith A, Martinet D, Andersson J, et al: A lated to serum hormones in anorexia ner- 118 Östlund H, Keller E, Hurd YL: Estrogen re-
new highly penetrant form of obesity due to vosa adolescents. World J Biol Psychiatry ceptor gene expression in relation to neuro-
deletions on chromosome 16p11.2. Nature 2014;15:327–333. psychiatric disorders. Ann NY Acad Sci
2010;463:671–675. 106 Kesselmeier M, Pütter C, Volckmar A-L, 2003;1007:54–63.
  94 Doche ME, Bochukova EG, Su H, Pearce LR, Baurecht H, Grallert H, Illig T, et al: High- 119 Asarian L, Geary N: Sex differences in the
Keogh JM, Henning E, et al: Human SH2B1 throughput DNA methylation analysis in physiology of eating. Am J Physiol Regul In-
mutations are associated with maladaptive anorexia nervosa confirms TNXB hyper- tegr Comp Physiol 2013;305:R1215–R1267.
behaviors and obesity. J Clin Invest 2012; methylation. World J Biol Psychiatry, Epub 120 Wasser SK, Barash DP: Reproductive sup-
122:4732–4736. ahead of print. pression among female mammals: implica-
  95 Gill R, Chen Q, D’Angelo D, Chung WK: 107 Perroud N, Paoloni-Giacobino A, Prada P, tions for biomedicine and sexual selection
Eating in the absence of hunger but not loss Olié E, Salzmann A, Nicastro R, et al: In- theory. Q Rev Biol 1983;58:513–538.
of control behaviors are associated with creased methylation of glucocorticoid re- 121 Abed R: The sexual competition hypothesis
16p11.2 deletions. Obesity 2014; 22: 2625– ceptor gene (NR3C1) in adults with a history for eating disorders. Br J Med Psychol 1998;
2631. of childhood maltreatment: a link with the 71:525–547.
  96 Maillard AM, Hippolyte L, Rodriguez-Her- severity and type of trauma. Transl Psychia- 122 Guisinger S: Adapted to flee famine: adding
reros B, Chawner SJRA, Dremmel D, Agüera try 2011;1:e59. an evolutionary perspective on anorexia
Z, et al: 16P11.2 Locus modulates response 108 Baker JH, Maes HH, Lissner L, Aggen SH, nervosa. Psychol Rev 2003;110:745–761.
to satiety before the onset of obesity. Int J Lichtenstein P, Kendler KS: Genetic risk fac- 123 Wells JCK: The evolution of human fatness
Obes (Lond) 2015;40:870–876. tors for disordered eating in adolescent and susceptibility to obesity: an ethological
 97 Jacquemont S, Reymond A, Zufferey F, males and females. J Abnorm Psychol 2002; approach. Biol Rev 2006;81:183–205.
Harewood L, Walters RG, Kutalik Z, et al: 118:576–586. 124 Speakman JR, Levitsky DA, Allison DB, Bray
Mirror extreme BMI phenotypes associat- 109 Keski-Rahkonen A, Bulik CM, Neale BM, MS, de Castro JM, Clegg DJ, et al: Set points,
ed with gene dosage at the chromosome Rose RJ, Rissanen A, Kaprio J: Body dissat- settling points and some alternative models:
16p11.2 locus. Nature 2011;478:97–102. isfaction and drive for thinness in young theoretical options to understand how genes
  98 Strober M, Peris T, Steiger H: The plasticity adult twins. Int J Eat Disord 2005; 37: 188– and environments combine to regulate body
of development: how knowledge of epi- 199. adiposity. Dis Model Mech 2011;4:733–745.
genetics may advance understanding of eat- 110 Keski-Rahkonen A, Neale BM, Bulik CM, 125 Field AE, Sonneville KR, Micali N, Crosby
ing disorders. Int J Eat Disord 2014;47:696– Pietilainen KH, Rose RJ, Kaprio J, et al: In- RD, Swanson SA, Laird NM, et al: Prospec-
704. tentional weight loss in young adults: sex- tive association of common eating disorders
  99 Frieling H, Gozner A, Römer KD, Lenz B, specific genetic and environmental effects. and adverse outcomes. Pediatrics 2012; 130:
Bönsch D, Wilhelm J, et al: Global DNA hy- Obes Res 2005;13:745–753. e289–e295.
pomethylation and DNA hypermethylation 111 Slof-Op ’t Landt MCT, Bartels M, Van Furth 126 Kaye WH, Klump KL, Frank GK, Strober M:
of the alpha synuclein promoter in females EF, Van Beijsterveldt CEM, Meulenbelt I, Anorexia and bulimia nervosa. Annu Rev
with anorexia nervosa. Mol Psychiatry 2007; Slagboom PE, et al: Genetic influences on Med 2000;51:299–313.
12:229–230. disordered eating behaviour are largely in- 127 Neel J V: Diabetes mellitus: a “thrifty” geno-
100 Frieling H, Bleich S, Otten J, Römer KD, dependent of body mass index. Acta Psychi- type rendered detrimental by “progress.”
Kornhuber J, de Zwaan M, et al: Epigenetic atr Scand 2008;117:348–356. Am J Hum Genet 1962;14:353–362.
downregulation of atrial natriuretic peptide 112 Wolfe BE, Baker CW, Smith AT, Kelly- 128 Swinburn BA, Sacks G, Hall KD, McPherson
but not vasopressin mRNA expression in fe- Weeder S: Validity and utility of the current K, Finegood DT, Moodie ML, et al: The
males with eating disorders is related to im- definition of binge eating. Int J Eat Disord global obesity pandemic: shaped by glob-
pulsivity. Neuropsychopharmacology 2008; 2009;42:674–686. al drivers and local environments. Lancet
33:2605–2609. 113 Reichborn-Kjennerud T, Bulik CM, Kendler 2011;378:804–814.
101 Kim Y-R, Kim J-H, Kim MJ, Treasure J: Dif- KS, Røysamb E, Maes H, Tambs K, et al: 129 Mitchell NS, Catenacci VA, Wyatt HR, Hill
ferential methylation of the oxytocin recep- Gender differences in binge-eating: a popu- JO: Obesity: overview of an epidemic. Psy-
tor gene in patients with anorexia nervosa: a lation-based twin study. Acta Psychiatr chiatr Clin North Am 2011;34:717–732.
pilot study. PLoS One 2014;9:e88673. Scand 2003;108:196–202. 130 Scolnick B, Mostofsky DI: Anorexia nervo-
sa: a rogue hibernation? Med Hypotheses
2014;82:231–235.

Genes, Evolution, and Eating Disorders Neuroendocrinology 2018;106:292–306 305


DOI: 10.1159/000484525
131 Farooqi SI, Keogh JM, Yeo GSH, Lank EJ, 137 Gamero-Villarroel C, Rodriguez-Lopez R, 144 Gluck ME, Geliebter A: Racial/ethnic differ-
Cheetham T, O’Rahilly S: Clinical spectrum Jimenez M, Carrillo JA, Garcia-Herraiz A, ences in body image and eating behaviors.
of obesity and mutations in the melanocor- Albuquerque D, et al: Melanocortin-4 recep- Eat Behav 2002;3:143–151.
tin 4 receptor gene. N Engl J Med 2003;348: tor gene variants are not associated with 145 Simpson KJ: Anorexia nervosa and culture.
2373–2383. binge-eating behavior in nonobese patients J Psychiatr Ment Health Nurs 2002;9:65–71.
132 Geller F, Reichwald K, Dempfle A, Illig T, with eating disorders. Psychiatr Genet 2015; 146 Becker AE, Fay K, Gilman SE, Striegel-
Vollmert C, Herpertz S, et al: Melanocor- 25:35–38. Moore R: Facets of acculturation and their
tin-4 receptor gene variant I103 is negatively 138 Urwin RE, Bennetts BH, Wilcken B, Beu- diverse relations to body shape concern in
associated with obesity. Am J Hum Genet mont PJ V, Russell JD, Nunn KP: Investiga- Fiji. Int J Eat Disord 2007;40:42–50.
2004;74:572–581. tion of epistasis between the serotonin trans- 147 Becker AE, Burwell RA, Gilman SE, Herzog
133 Stutzmann F, Vatin V, Cauchi S, Morandi A, porter and norepinephrine transporter DB, Hamburg P: Eating behaviours and at-
Jouret B, Landt O, et al: Non-synonymous genes in anorexia nervosa. Neuropsycho- titudes following prolonged exposure to
polymorphisms in melanocortin-4 receptor pharmacology 2003;28:1351–1355. television among ethnic Fijian adolescent
protect against obesity: the two facets of a 139 Urwin RE, Nunn KP: Epistatic interaction girls. Br J Psychiatry 2002;180:509–514.
Janus obesity gene. Hum Mol Genet 2007; between the monoamine oxidase A and se- 148 Klump KL: Puberty as a critical risk period
16:1837–1844. rotonin transporter genes in anorexia ner- for eating disorders: a review of human and
134 Hebebrand J, Fichter M, Gerber G, Goerg T, vosa. Eur J Hum Genet 2005;13:370–375. animal studies. Horm Behav 2013; 64: 399–
Hermann H, Geller F, et al: Genetic predis- 140 Thaler L, Groleau P, Badawi G, Sycz L, Zer- 410.
position to obesity in bulimia nervosa: a mu- amdini N, Too A, et al: Epistatic interactions 149 Stephens M, Baliding DJ: Bayesian statistical
tation screen of the melanocortin-4 receptor implicating dopaminergic genes in bulimia methods for genetic association studies. Nat
gene. Mol Psychiatry 2002;7:647–651. nervosa (BN): relationships to eating- and Rev Genet 2009;10:681–90.
135 Branson R, Potoczna N, Kral JG, Lentes KU, personality-related psychopathology. Prog 150 Bulik CM, Kleiman SC, Yilmaz Z: Genetic
Hoehe MR, Horber FF: Binge eating as a ma- Neuropsychopharmacol Biol Psychiatry epidemiology of eating disorders. Curr Opin
jor phenotype of melanocortin 4 receptor 2012;39:120–128. Psychiatry 2016;383–388.
gene mutations. N Engl J Med 2003; 348: 141 Stice E, Shaw HE: Role of body dissatisfac- 151 Aschard H, Lutz S, Maus B, Duell EJ, Finger-
1096–1103. tion in the onset and maintenance of eating lin TE, Chatterjee N, et al: Challenges and
136 Stutzmann F, Tan K, Vatin V, Dina C, Jouret pathology: a synthesis of research findings. J opportunities in genome-wide environmen-
B, Tichet J, et al: Prevalence of melanocor- Psychosom Res 2002;53:985–993. tal interaction (GWEI) studies. Hum Genet
tin-4 receptor deficiency in Europeans and 142 Neumark-Sztainer DR, Wall MM, Haines JI, 2012;131:1591–1613.
their age-dependent penetrance in multi- Story MT, Sherwood NE, van den Berg PA: 152 Deng WQ, Paré G: A fast algorithm to opti-
generational pedigrees. Diabetes 2008; 57: Shared risk and protective factors for over- mize SNP prioritization for gene-gene and
2511–2518. weight and disordered eating in adolescents. gene-environment interactions. Genet Epi-
Am J Prev Med 2007;33:359–369. demiol 2011;35:729–738.
143 Pike KM, Dohm FA, Striegel-Moore RH,
Wilfley DE, Fairburn CG: A comparison of
black and white women with binge eating
disorder. Am J Psychiatry 2001; 158: 1455–
1460.

306 Neuroendocrinology 2018;106:292–306 Mayhew/Pigeyre/Couturier/Meyre


DOI: 10.1159/000484525

You might also like