You are on page 1of 12

Review Article

https://doi.org/10.1038/s41593-022-01071-z

Genetics and neurobiology of eating disorders


Cynthia M. Bulik   1,2,3 ✉, Jonathan R. I. Coleman   4,5, J. Andrew Hardaway6, Lauren Breithaupt7,8,
Hunna J. Watson1,9,10, Camron D. Bryant   11 and Gerome Breen   4,5

Eating disorders (anorexia nervosa, bulimia nervosa and binge-eating disorder) are a heterogeneous class of complex illnesses
marked by weight and appetite dysregulation coupled with distinctive behavioral and psychological features. Our understand-
ing of their genetics and neurobiology is evolving thanks to global cooperation on genome-wide association studies, neuroimag-
ing, and animal models. Until now, however, these approaches have advanced the field in parallel, with inadequate cross-talk.
This review covers overlapping advances in these key domains and encourages greater integration of hypotheses and findings
to create a more unified science of eating disorders. We highlight ongoing and future work designed to identify implicated bio-
logical pathways that will inform staging models based on biology as well as targeted prevention and tailored intervention, and
will galvanize interest in the development of pharmacologic agents that target the core biology of the illnesses, for which we
currently have few effective pharmacotherapeutics.

E
ating disorders are severe psychiatric disorders that, for many, Heritability estimates range from 0.55 to 0.62 for BN6 and 0.39 to
evolve into chronic or fluctuating conditions with serious 0.45 for BED6.
adverse outcomes1. Among adolescents and young adults, the
disability-adjusted life years for anorexia nervosa (AN) and bulimia Genome-wide association studies. Candidate gene and linkage lit-
nervosa (BN) are among the highest of all psychiatric disorders2. erature in eating disorders was rife with unreplicated small-sample
Binge-eating disorder (BED) and other specified feeding and eat- studies and was minimally informative beyond signaling the likely
ing disorders have yet to be included in estimates, but are projected complexity of their genetics6. We take as a starting point the most
to account for the majority of the global disease burden of eating recent genome-wide association study (GWAS) of AN performed by
disorders3. Although the disorders occur in pure forms, diagnostic the Eating Disorders Working Group of the Psychiatric Genomics
crossover across the course of illness is common4. For a brief defini- Consortium (PGC-ED)8. Combining existing samples9 with the
tion of the symptoms and epidemiology of the three primary eating Anorexia Nervosa Genetics Initiative10 yielded a dataset of European
disorders, see Box 1. ancestry including 16,992 AN cases and 55,525 controls, from 17
Biological effects within eating disorders range from the sub- countries, and identified 8 genome-wide significant loci, including
cellular (genetic variants and their effects on gene expression and 4 single-gene loci: CADM1, MGMT, FOXP1 and PTBP2 (refs. 8).
structure), through the cellular (signaling) and intercellular (neu- Analysis of tissue enrichment for AN-associated genes revealed a
rons and neuronal circuits), to organismal effects (eating disorders significant association with the CNS, and comparison to single-cell
and disorder-related behaviors)5. These levels of biology are inter- gene expression datasets from mice revealed significant associations
connected, but are investigated using different scientific approaches, with hippocampal pyramidal neurons and striatal medium spiny
with limited cross-talk to date. Integrating results from different neurons. Song et al.11 further showed that AN-associated genes were
approaches is vital. A hierarchically connected research approach, enriched in the prefrontal cortex (PFC).
reflecting the biological interconnectivity, will provide a more com-
plete understanding of the biology of eating disorders, providing the Genetic correlations implicate psychiatric, educational,
foundations for developing much-needed novel treatments. In this and metabolic factors in anorexia nervosa. The estimates of
review, we describe the state of the science across biological levels single-nucleotide polymorphism (SNP)-based genetic correlations
and approaches and conclude by discussing progress toward this (SNP-rg) between AN and psychiatric disorders such as obsessive–
more integrated understanding. compulsive disorder (OCD), major depressive disorder (MDD),
anxiety, and substance-related disorders8,12 correspond with clini-
Human genetics of eating disorders cal observations of comorbidity13 and twin studies14–16. Building on
Twin-based heritability. Large-scale twin studies yield herita- this work, AN and OCD risk genes pinpointed through GWASs
bility estimates ranging from 0.28 to 0.74 for AN6, with impreci- showed similar PFC expression alterations, meaning that these
sion reflecting varying definitions of illness and sample size, and disorders may have similar functional pathways11. A new positive
with more severe definitions associated with higher heritabilities7. SNP-rg with objectively measured physical activity was found in the

Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. 2Department of Nutrition, University of North Carolina at
1

Chapel Hill, Chapel Hill, NC, USA. 3Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden. 4Social, Genetic and
Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK. 5National Institute of Health
Research Maudsley Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, UK. 6Department of Psychiatry and
Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA. 7Department of Psychiatry, Harvard Medical School, Boston, MA,
USA. 8Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, MA, USA. 9School of Psychology, Curtin University,
Perth, Western Australia, Australia. 10Division of Paediatrics, School of Medicine, The University of Western Australia, Perth, Western Australia,
Australia. 11Department of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, USA.
✉e-mail: cynthia_bulik@med.unc.edu

Nature Neuroscience | VOL 25 | May 2022 | 543–554 | www.nature.com/natureneuroscience 543


Review Article NaTUrE NEUrOsciEncE

Box 1 | Overview of eating disorder phenotypes concluded that both metabolic and psychological factors may need
to be considered to fully understand AN and that doing so may
hasten the development of more effective treatments. On the basis
Anorexia nervosa. The hallmark symptom of AN is low body
of the strong associations with psychiatric traits, physical activity,
weight, accompanied by persistent restriction of energy intake
educational attainment, and metabolic traits, multivariate GWAS
and/or increased energy expenditure, intense fear of gaining
analyses (for example, genomic structural equation modeling)
weight, or persistent behavior preventing weight gain, and dis-
could offer novel opportunities to disentangle common and specific
torted body image1. Both a restricting and a binge-eating/purg-
genetic effects and help to boost power for discovery.
ing subtype exist. The lifetime prevalence is 1.4% (0.1–3.6%) for
women and 0.2% (0–0.3%) for men131. The standardized mortali-
Polygenic scores. Polygenic scores (PGS) summarize genome-wide
ty ratio of AN is >5 (ref. 132), with deaths primarily attributable to
data into a single variable of genetic liability to the phenotype—as a
illness effects and suicide133. Treatment outcome in adults is poor,
sum of the number of risk alleles present in the individual weighted
with only 30% achieving full recovery134. Family-based treatment
by the SNP effects from GWASs. Higher PGS for AN derived from
for youth is recommended; multidisciplinary interventions are
the 2019 PGC-ED GWAS were associated with increased odds
recommended for adults, although the evidence base remains
of AN, and equally in females and males8. Further, PGS of age of
particularly weak135. No medications have been identified or de-
onset of AN predicted age at onset in independent cohorts18. A rich
veloped that successfully treat the disorder or are approved by
body of literature applying PGS is emerging, with multiple stud-
the US Food and Drug Administration (FDA)135.
ies reporting that BMI and psychiatric PGS influence childhood
Bulimia nervosa. The core symptoms of BN include binge eating and adolescent disordered eating behaviors19–21. PGS for other
(eating an unusually large amount of food in a circumscribed phenotypes may also differentiate among AN, BN and BED on the
period of time accompanied by a sense of loss of control) and genomic level. UK Biobank analyses revealed strong positive asso-
inappropriate compensatory behaviors (for example, self-induced ciations between binge-type eating disorders (BN and BED) and
vomiting, abuse of laxatives, diuretics, fasting, and excessive anthropometric PGS (for example, overweight and waist circum-
exercise), with self-evaluation being strongly influenced by shape ference), suggesting that binge eating may share genomic variants
and weight1. BN can occur at all body weights, but is diagnosed with overweight and obesity. In contrast, in AN, the direction of
as AN binge-eating/purging subtype in the presence of AN. The these associations was reversed22.
lifetime prevalence is 1.9% (0.3–4.6%) for women and 0.6% We anticipate increasing evidence that PGS will predict devel-
(0.1-1.3%) for men131, and onset is typically in late adolescence opmental phenotypic manifestations of illness (that is, premorbid
or early adulthood. Cognitive-behavioral therapy is the leading behavioral traits and course of illness) and gene–environment asso-
evidence-based treatment, and fluoxetine, whose efficacy was ciations, and ultimately inform risk assessment and clinical applica-
established in placebo-controlled clinical trials in the early 1990s, tions. At present, their clinical utility is limited23,24.
is the only FDA-approved medication135. BN recovery rates are
~50% at 5–10 years of follow-up136. Cross-disorder genome-wide association studies. A cross-disorder
GWAS of eight psychiatric disorders (AN, ADHD, autism spec-
Binge-eating disorder. BED is marked by recurrent binge trum disorder, bipolar disorder, MDD, OCD, schizophrenia and
eating that causes distress, the absence of regular compensatory Tourette’s syndrome) revealed 109 pleiotropic loci (that is, signals
behaviors, and behavioral and emotional features such as eating for at least two disorders), of which 8 had signals for AN25. The top
rapidly or when not hungry, and feeling embarrassed or disgusted locus (18q21.2) was associated with all disorders, has previously
by one’s behavior1. The lifetime prevalence is 2.8% (0.6–5.8%) for been implicated in MDD and neuroticism, and includes the DCC
women and 1.0% (0.3–2.0%) for men131. Onset is typically in early gene, encoding netrin 1 receptor, which regulates axon outgrowth.
adulthood but can be at any time and in individuals spanning the A joint genomic structural equation model showed that AN loaded
normal to obese weight ranges1. Evidence-based treatments for onto a factor with OCD and Tourette’s syndrome26. These findings
BED include cognitive-behavioral therapy, second-generation suggest overlapping genetic risk across disorders and that work
antidepressants, and, since 2015, lisdexamfetamine, which is an needs to be done to separate specific from nonspecific susceptibility
FDA-approved stimulant originally prescribed for the treatment variants, and the association of specific variants with cellular gene
of attention-deficit hyperactivity disorder (ADHD)137. Preclinical, expression datasets to generate testable hypotheses for their ulti-
clinical and genetic studies (of binge eating) and neuroimaging data mate impact on brain circuit function. Other cross-disorder GWASs
converge on dysfunction in systems that regulate eating behavior on AN and OCD have yielded promising insights but require larger
and reward (that is, dopaminergic and noradrenergic systems), samples to pinpoint risk loci27.
and support the repositioning of lisdexamfetamine in BED138.
Mendelian randomization. MR studies treat genetic variants as
unconfounded proxies of an exposure of interest to evaluate the
causal effect of an exposure on a phenotypic outcome. However, MR
PGC-ED GWAS8 and may relate to the driven exercise seen in AN. analyses investigating the causal effects of AN on other traits are
Significant positive SNP-rg with educational attainment was not presently limited in power. As stated earlier, a bidirectional causal
seen for IQ (Fig. 1). association between AN and low BMI was found in the PGC-ED
Earlier results hinted at a significant negative genetic correla- GWAS8. An MR study on the ALSPAC cohort (n = 4,473) suggested
tion between AN and body mass index (BMI)9,17. The PGC-ED possible causal effects of genetically predicted childhood BMI on
GWAS revealed significant genetic correlations with metabolic, binge eating and weight and shape concern, and of binge eating
lipid, and anthropometric traits, suggesting that metabolic mecha- and overeating on BMI28. In another study, adiponectin, a fat-tissue
nisms may drive physiological resistance to healthy body weight in derived hormone, showed evidence consistent with a causal effect
AN8. The observed correlations were not confounded by the genet- on eating disinhibition but not eating restraint29. Adams et al.30
ics of BMI—a concern given that low body weight is a component found evidence consistent with a protective effect of fasting insulin
of the AN diagnosis8. Likewise, exploratory Mendelian random- level (n = 108,557) on AN (n = 72,515; odds ratio (OR) = 0.48, 95%
ization (MR) analyses in the PGC-ED GWAS indicated a bidirec- confidence interval: 0.33, 0.71), but not for fasting glucose or gly-
tional causal relationship between AN and low BMI. The authors cated hemoglobin (HbA1c) on AN. Future MR studies will help to

544 Nature Neuroscience | VOL 25 | May 2022 | 543–554 | www.nature.com/natureneuroscience


NaTUrE NEUrOsciEncE Review Article
OCD

Major depression

Anxiety disorder

HDL cholesterol

Educational attainment

Physical activity

Trait Fat-free mass

Obesity (BMI of 35–40)

Obesity (BMI of 30–35)

Type 2 diabetes

Leptin

Fasting insulin

BMI

Fat mass

–0.4 –0.3 –0.2 –0.1 0 0.1 0.2 0.3 0.4 0.5 0.6
Genetic correlation

Fig. 1 | Genetic correlations between anorexia nervosa and selected top traits. Error bar represent 95% confidence intervals. Values were retrieved from
ref. 8. HDL, high-density lipoprotein.

disentangle the nature of pleiotropy and shed light on causal expo- gene expression results currently rely on limited high-throughput
sure–outcome effects. chromatin conformation capture (Hi-C), expression quantita-
tive trait loci (eQTL) and brain cell data. Larger sample sizes are
Rare mutations and copy number variants. No Mendelian forms of required to improve statistical power and to detect more suscep-
eating disorders have been identified, and efforts to find rare genetic tibility loci. Large, systematic sequencing studies are required to
variants have not yet been successful. In an exome-chip-based assess the role of rare variants and to complement common vari-
GWAS that included 2,158 individuals with AN and 15,327 control ant GWASs. Finally, although eating disorders are likely to be
participants31, 16 independent variants were taken forward for in under-detected in males, the gender ratio is definitely tipped in the
silico and de novo replication. No findings reached genome-wide direction of females. Accruing larger samples sizes of afflicted males
significance. Scott-Van Zeeland et al.32 conducted a series of tar- is essential for identifying genetic sex differences and is a priority
geted sequencing and genotyping studies focusing on 152 candidate for future research.
genes in 1,205 individuals with AN cases and 1,948 control partici-
pants, implicating variants in the estrogen receptor-b (ESR2) and Integration. Genomic discovery in eating disorders is underway
epoxide hydrolase 2 (EPHX2) genes. A smaller exome sequencing and early discoveries highlight new directions for deepening our
study of 93 individuals with AN reported enrichment of rare vari- understanding of some of the more perplexing facets of AN (for
ants in neuropeptide/neurotrophic pathways33. Combined linkage example, extreme weight dysregulation, the frequent re-loss of
and sequencing of densely affected families with multiple individu- therapeutically restored weight, and compulsive exercise). The PGC
als with AN or BN has been reported34,35. The results of these studies is expanding genomic samples from individuals with AN, BN, and
are tentative as sufficient sample sizes to identify rare point muta- BED with efforts such as the Eating Disorders Genetics Initiative
tions have yet to be attained. (EDGI)40. Novel biological discoveries are anticipated to acceler-
The situation is similar for genome-wide analyses of copy num- ate therapeutic opportunities in drug discovery, the repositioning
ber variants (CNVs). Wang et al.36 found no evidence of CNV of current drugs, and our understanding of pleiotropy including
enrichment in people with AN compared to controls. Yilmaz opposing effects that may mitigate unintended off-target effects.
et al.37 analyzed 1,983 women with AN38 and found no occurrence The availability of biobanks with genetic and electronic health
of well-established pathogenic CNVs for neurodevelopmental record data offer opportunities to extend genetic associations to
disorders (1q21.1, 2p16.3, 3q29, 7q36.3, 13q12, 15q11.2, 15q13.3, other medical phenotypes not currently available. As with many
16p11.2(1), 16p11.2(2), 17q12 or 22q11.21). Chang et al.39 reported illnesses, global cooperation and harmonization of methods boost
the association of a 15q11.2 microduplication with AN, but cau- sample sizes and accelerate science. Clearly, collaboration with
tioned that the results require validation. Currently, there is no researchers in other fields will increase the utility of GWAS results
credible evidence that novel or known CNVs with large effect sizes and integrate them into a unified science of eating disorders. This
influence AN. Much larger sample sizes may be required before we unification can be achieved by targeting dimensional phenotypes
can exclude a role for CNVs carrying moderate effect sizes. and symptoms and generating hypotheses for testing the causal
impact of genomic variants on neural circuit function.
Limitations of human genetic studies. Several limitations in the GWASs of eating disorders have focused primarily on diagno-
field should be considered. First, to our knowledge, no GWAS of ses; however, exploring relevant dimensions or core symptoms may
BN, BED, purging disorder, atypical AN, avoidant/restrictive food enhance translation to approximate phenotypes in animal mod-
intake disorder, pica or rumination disorder have been published. els. Fine-mapping approaches are required to delve deeper into
Second, for AN, genome-wide signals currently account for a low trait-associated regions to identify specific variants or genes that
proportion of phenotypic variance (1.7%), while gene mapping and causally influence the target trait. Clearly describing the strength of

Nature Neuroscience | VOL 25 | May 2022 | 543–554 | www.nature.com/natureneuroscience 545


Review Article NaTUrE NEUrOsciEncE

a b

P P

I I

Fig. 2 | Human neuroimaging circuitry involved in eating disorders. a, Images represent the structures comprising two major dopaminergic pathways,
mesolimbic and mesocortical pathways, supported by previous work in animal models. Both pathways originate in the VTA (red). Mesolimbic pathways
project to the NAc, and is part of the complex circuit involving the amygdala (pink), hippocampus (green) and the bed nucleus of the stria terminalis
(yellow). The mesocortical pathway projects primarily to the PFC (orange) and insula (purple). b, Images represent substructures involved in the CSTC
pathway that are supported by recent genetic/GWAS studies with shared functional networks that exhibit overlapping phenotypes. The CSTC pathway is a
multisynaptic neuronal circuit that connects the cortex with the striatum and thalamus. The striatum (green) receives glutamatergic input from the cortex
and the thalamus (blue) sends out GABAergic inputs to the subthalamic nucleus (pink, purple, red).

evidence for the functional involvement of specific genes and vari- future GWASs support MGMT as a causal gene for AN, its role is
ants in eating disorders will provide strong targets for causal valida- worthy of future investigation.
tion and neurobiological exploration. The identification of conserved, eating disorder-relevant genes
is currently limited by underpowered phenotype-driven analyses in
Neurobiology of genes implicated in genome-wide association humans and rodents. Neuroscience typically focuses on a different
studies of anorexia nervosa. Although too recent to have influ- level of the biological hierarchy from statistical genetics. As such,
enced neurobiological studies directly, several genes implicated by deeper insight may be gained by examining the neuronal circuitry
the latest AN GWAS have been studied in relation to other traits and implicated directly through neuroscientific approaches and indi-
show neurobiological relevance to eating disorders. For example, rectly through GWAS-based approaches. GWAS-based approaches
FOXP1 haploinsufficiency is a rare form of intellectual disability, use information on cell-type-specific genome biology, such as epi-
related to autism spectrum disorder41. Haploinsufficiency of FOXP1 genetic markers49 or gene expression50. These methods show that
is associated not only with neurodevelopmental impairments, genes associated with AN are, on average, specifically expressed in
but also with feeding difficulties and gastrointestinal disturbance brain tissues, especially in medium spiny neurons and CA1 hippo-
in humans and in mice, supporting the role of FOXP1 in AN41. campal pyramidal neurons8. Results from these methods implicate
Conditional Foxp1-knockout studies in the mouse brain implicate the same cell types in other psychiatric disorders and are sup-
neurogenesis and neural migration, particularly the development ported by functional studies in rodents. Differential vulnerability to
and functioning of medium spiny neurons and pyramidal neurons. activity-based anorexia (ABA) in female C57BL/6J mice was asso-
Both cell types have been implicated as relevant neuronal cell types ciated with GABAergic inhibition of hippocampal CA1 pyramidal
by systems biological analyses of the AN GWAS41. cells51. Prolonged binge-like consumption of sucrose has been shown
As well as being implicated in AN, CADM1 and PTBP2 to alter the morphology of medium spiny neurons in the nucleus
have been implicated in GWASs of BMI42. PTBP2 encodes a accumbens (NAc) of rats52. However, beyond broad support for
neuron-specific RNA-binding protein that organizes axonogen- brain tissues in general, most circuits that have been implicated in
esis in the developing cortex, which is relevant across psychiat- neurobiological studies of mice and of humans are poorly supported
ric disorders43. By comparison, CADM1 has not been associated by these GWAS-based approaches, probably owing to limitations of
with psychological or behavioral traits except for BMI and AN power and insufficient data from appropriate tissues. For example,
(although the family member CADM2 has been implicated in in contrast to the neurobiological evidence supporting a role for
numerous traits related to impulsive behavior)44. CADM1 encodes DA circuitry in AN, there is no such evidence from AN GWAS8.
a synaptic cell adhesion molecule. The BMI-associated variants However, evidence for genetic variants affecting the dopamine (DA)
appear to increase the expression of CADM1 in the human hypo- system in eating disorders may emerge as the power of eating disor-
thalamus and cerebellum, and parallel experiments in mice suggest der GWASs increases. Both of these limitations are being overcome
that this increased expression contributes to weight gain, poten- through increasing GWAS sample sizes and via collaborative brain
tially through CADM1-positive innervation of POMC neurons45. mapping initiatives like the PsychENCODE project53.
CADM1 is also involved in the neural control of first estrous in
mice, which is of particular interest given amenorrhea in AN1. Human neuroimaging in eating disorders
MGMT, which encodes a DNA alkyltransferase, is the most well Neuroimaging research in eating disorders has been motivated
studied of the genes implicated in the AN GWAS, as it is involved by (1) previous work in animal models that studied the micro-
in DNA repair and protection against cancer46. The biological path- circuitry involved in homeostatic and hedonic eating path-
ways that link DNA repair dysfunction to AN are unclear, although ways and (2) genetic studies and GWASs that have identified
recent research suggests that postmitotic neurons require recurrent shared functional networks that exhibit overlapping phenotypes
DNA repair that is relevant to neuronal dysfunction47,48. Assuming (cortico-striatal-thalamo-cortical (CSTC) pathways; Fig. 2).

546 Nature Neuroscience | VOL 25 | May 2022 | 543–554 | www.nature.com/natureneuroscience


NaTUrE NEUrOsciEncE Review Article
Hedonic reward pathways—mesocorticolimbic and mesolim- Here are limited neuroimaging data to suggest that functional
bic networks. The brain reward system is well-defined and plays and structural alterations in control circuits occur early in the
a central role in the drive to eat. Mesocorticolimbic and mesolim- course of BN and may contribute to the disorder’s persistence over
bic pathways project from the ventral tegmental area (VTA) to time73,74. In patients with AN, maladaptive excessive self-control is
the cerebral cortex (frontal, cingulate, and entorhinal cortex) and commonly described although the differences in underlying neuro-
limbic structures (ventral striatum, hippocampus, and amygdala), biological correlates (that is, structures) involving cognitive control
respectively. Importantly, these systems are largely intertwined and are inconsistent in imaging studies75,76. Both individuals with AN
overlapping54, and assigning specific brain regions to one clinical and those with BN show reduced gray matter volume in the caudate
phenotype (for example, restriction) or characteristic (for example, nucleus, ACC, and insula77; however, these results may normalize in
cognitive control) is unhelpful as they act in unison. Collectively, AN and BN following successful treatment78.
mesocorticolimbic and mesolimbic pathways are responsible for
cognitive functions, reward, emotion, and motivation, which may Limitations. Deficits in cognitive control, executive functioning,
represent transdiagnostic factors underlying AN, BN, and BED55,56. reward, and affective processing are commonly reported across
Early studies focusing on understanding the neurobiology of multiple psychiatric disorders, with similar neurobiological corre-
eating disorders suggested that there were pathological alterations lates reported in the dorsolateral PFC, insula, and dorsal ACC79–81. A
in monoamine neurotransmitter systems, specifically DA and limited number of studies compare neuroimaging across psychiatric
serotonin or 5-hydroxytryptamine (5-HT)56. These findings align disorders (for example, refs. 82,83) or use meta-analytic techniques
with other works that have strongly implicated the reward system to do so (for example, refs. 79,80). Those that have been conducted
in pathological changes in hedonic eating and decision-making. have not included patients with eating disorders, despite evidence
The VTA comprises a cluster of DA-producing neurons that play of psychiatric multimorbidity (including mood, substance use, and
a key role in positive and negative reinforcement, decision making, anxiety disorders)84. Eating disorder exclusion may occur because
working memory, incentive salience, and aversion. Dopaminergic of state-specific neurobiological alterations due to malnourishment
VTA neurons innervate corticolimbic regions via the mesocorti- or dehydration stemming from key eating disorder-specific behav-
cal pathway, forming the mesocorticolimbic reward network. A iors (for example, caloric restriction, purging, and excessive exer-
subpopulation of midbrain DA neurons that project to the stria- cise). Failure to account for state-specific effects has stymied human
tum co-release glutamate and GABA onto their target neural sub- neuroimaging research in eating disorders. Eating disorder-specific
strates57. Understanding the structural and functional connectivity recommendations from experts in the fields of eating disorders and
of these reward circuits in eating disorders has been a central focus neuroimaging85 have been proposed to account for malnutrition
for the neuroimaging investigations highlighted below. effects. Importantly, novel experimental designs are essential to dis-
Functional neuroimaging studies suggest altered processing of aggregate the extent to which any observed neurobiological altera-
rewarding and aversive food stimuli in acute and recovered AN58,59. tions in AN are truly related to disease etiology or more accurately
In binge-type eating disorders (BN and BED), functional neuroim- attributed to the impact of prolonged malnutrition/starvation.
aging findings are less consistent60,61. Positron emission tomogra- Accordingly, reevaluation of previous eating disorder neuroimag-
phy (PET) data in individuals who have recovered from AN show ing findings accounting for effects of malnourishment (for example,
increased striatal dopamine receptor (D2/D3) binding, which was white matter alterations in AN86 and BN87) has led to improved
also related to striatal responses during monetary choices and understanding of state versus trait effects in the brain.
self-reported trait anxiety62. However, two PET studies focusing
on individuals with acute63 and weight-restored AN64 compared Integrating human genetics and neuroimaging. Imaging and
to healthy control participants found no difference in DA recep- genetics target distant levels of the biological hierarchy, and so inte-
tor binding. PET studies in individuals with BN have identified gration is challenging. Genetically informed models of eating disor-
decreased striatal DA transporter availability65. In individuals with ders hold the potential to generate construct-valid neurobiological
BN, increases in glutamate signaling receptors (metabotropic glu- disease models. Direct examination of the effect of genetic variation
tamate receptor subtype 5; mGlu5) were higher in the anterior cin- on brain structure and function is underway through the ENIGMA
gulate cortex (ACC), subgenual PFC, and straight gyrus compared consortium88. Further insights could be obtained by examining
with control participants66. Structural neuroimaging studies in AN intermediate levels of the biological hierarchy. Cellular-level data
and BN reveal volumetric abnormalities in the insula67. Further, can be inferred from eating disorder GWASs; this has implicated
lower white matter measures of axonal integrity (measured via hippocampal pyramidal neurons and striatal medium spiny neu-
fractional anisotropy) and increased structural white matter con- rons as the cell types that express identified genes in AN8. However,
nectivity between the insula and orbital frontal cortex suggest that this approach remains in its infancy—larger sample sizes for GWAS
altered processing of taste perception may be present across eating and for gene expression datasets in neuronal and nonneuronal cells
disorders. The insular cortex (IC) extends beyond taste function (for example, microglia and oligodendrocytes) are needed to pro-
and is a center of body awareness, integrating autonomic, cognitive vide sufficient power for the parallel implication of brain circuits
and affective processing68 of the homeostatic state of the body. Both from neuroimaging and from GWASs.
structural and functional neuroimaging studies highlight deficits in
the insula and abnormal interoceptive activity in AN69,70 and BN71. Animal models of eating disorders
As eating disorder GWASs become larger and more robust, their
Cognitive control and habitual responding—cortico- ability to interconnect with animal models will increase. Animal
striatal-thalamo-cortical pathways. The CSTC pathway is a brain models offer speed in achieving power, control over allele frequency
circuit that controls movement execution, habit formation, and and genetic background, environmental exposure, and recording
reward, all of which have been hypothesized to be relevant to eating and collection of the appropriate cell types and tissues at appro-
disorders. Hyperactivity throughout the CSTC circuits is believed priate time points to study the dynamic genetic architecture and
to underlie OCD72, which is increasingly relevant given the strong genomic adaptations across eating disorder progression and recov-
positive genetic correlation between OCD and AN8. Overlapping ery. Two popular animal models for investigating eating disorders
CSTC loops, including lateral PFC, ACC, dorsal striatum, the pre- are the ABA model and the binge-like eating (BLE) model (Box 2).
supplementary motor area, insula, and parietal regions, are involved Genes, variants, and circuit mechanisms gleaned from animal stud-
in these processes. ies can be discovered and validated first within the same species and

Nature Neuroscience | VOL 25 | May 2022 | 543–554 | www.nature.com/natureneuroscience 547


Review Article NaTUrE NEUrOsciEncE

Box 2 | Animal models for eating disorders with increased genetic diversity, additional diets (high-fat diet),
and environmental variables will increase gene identification. In
support, a recent study involving a cross between the BLE-prone
Activity-based anorexia. AN has some characteristics that can-
DBA/2J strain with the BLE-resistant C57BL/6J strain92 identi-
not be modeled in rodents (drive for thinness, body image dis-
fied multiple QTLs that influenced eating behavior and body
tortion and intense fear of weight gain). However, key behavioral
weight, including a sex-combined QTL containing the candidate
components of AN as well as traits commonly comorbid with
gene Lcorl that influences initial consumption of palatable food, a
AN can be modeled in mice (high physical activity, anxiety, de-
female-selective locus containing the candidate gene Zeb1 underly-
pression, social anxiety and obsessive-compulsive-like behav-
ing changes in body weight during BLE, and a male-selective locus
ior). Perhaps most well-known is the ABA model, which starts
for escalation in palatable food intake that contains the functional
with daily unlimited chow availability but for only 2 h per day
candidate genes Adipor2 and Plxnd1 (ref. 97).
or less followed by introduction of a running wheel, which leads
Given the diverse array of mouse crosses and populations that
to compulsive-like running, appetite suppression, voluntary re-
are now available to accelerate gene mapping, forward genetic
striction of food intake, decreased anxiety-like behavior, severe
studies of ABA and BLE in rodents provide the opportunity for
weight loss and death without intervention139. ABA induces oth-
novel gene/pathway identification. Such studies are sorely lacking.
er AN-like effects on physiology, including hypothermia, loss of
Nonetheless, Cyfip1 and Cyfip2 have homologs to study in humans.
estrus, increased hypothalamic–pituitary–adrenal axis activity,
Although neither of these genes has yet been associated with AN
anhedonia, ulcers and humoral, CNS, cardiovascular and gas-
through GWASs8, ongoing GWASs of BED and BN will be more rel-
trointestinal dysfunction140—a credible model for the behavioral
evant for assessing the association between CYFIP genes and binge
and physiological components of AN.
eating in humans.
Binge-like eating. BLE shares several features with substance
use disorders—tolerance, withdrawal, relapse, loss of control and Omic studies of activity-based anorexia. Omics analysis has been
compulsive intake. BLE has been studied in animals141, where it is applied to both brain tissue and gut microbiota to improve our
defined by increased intake of palatable food (high caloric) versus understanding of molecular adaptations associated with ABA and
chow and escalation of intake over time142. Compulsive-like BLE. Proteomic studies in ABA models have identified hypotha-
eating refers to habitual, often increased effortful intake despite lamic mitochondrial and autophagy processes98, deficits in energy
potential harm (aversive stimulus) that can relieve negative metabolism in colonic mucosa99, and an increase in ATP-producing
affect during withdrawal143. Home-cage chow restriction, stress glycolytic enzymes in gut microbiota100. These results implicate an
exposure and limited, intermittent access to palatable food can all adaptive energy source in the gut that could influence brain func-
promote BLE144. Intermittent BLE models induce the most robust tion and feeding behavior in AN. Opposing changes in energy uti-
BLE, yet do not typically induce obesity, as rodents voluntarily lization between the hypothalamus and gut mucosa during ABA
restrict chow intake in anticipation of palatable food, which in suggest that restoration of energy homeostasis between the CNS
turn, increases palatable food reinforcing efficacy145,146. BLE is a and periphery could improve treatment of AN in humans.
reasonable animal approximation of the behavioral components Animal models can be used to longitudinally measure the impact
of dysregulated eating characteristics of human binge eating. of early-life factors such as stress, diet, or genetics on eating dis-
orders. In a mouse model involving chronic, variable mild stress
in pregnant dams, prenatal stress induced transcriptomic indices
of hypothalamic–pituitary–adrenal axis and metabolic dysfunc-
genetic background and then in humans. Likewise, identified genes tion associated with obesity and protected against ABA in adoles-
from human GWAS and proposed circuitry from human neuroim- cent female mice. Prenatal stress protection was associated with
aging studies can also be tested for causality and function in animal increased DNA methylation and placental miR-340 downregula-
models, potentially on multiple genetic backgrounds (Fig. 3). tion and upregulation of miR-340 targets101. Low miR-340 and ABA
resistance were also associated with increased expression of solute
Phenotype-driven genetics of activity-based anorexia. Various carrier nutrient transporters (amino acids and glucose), and growth
mouse strains exhibit differential susceptibility to ABA. Specifically, factors that are potentially regulated by miR-340 targets. Placental
DBA/2J female mice show greater wheel running activity, greater overexpression of miR-340 recapitulated fetal and adolescent hypo-
weight loss, less food intake and more severe hypoleptinemia com- thalamic and circadian dysfunction101, supporting a role for placen-
pared to C57BL/6J mice89. C57BL/6J mice showed greater ABA than tal miR-340 in regulating nutrient availability that could influence
A/J mice and multiple chromosome substitution strains90. A genetic eating disorder susceptibility.
correlation between baseline physical activity (wheel running, loco-
motor) and ABA across inbred strains91 identified physical activity Omics of binge-like eating. Transcriptome analysis of the striatum
as a potential premorbid trait that predicts the development of ABA. found BLE-induced downregulation of myelination genes94, sup-
Despite inbred strain differences in ABA, causal genetic factors have porting reports of decreased white matter integrity with increased
yet to be found. binge eating in individuals with BN102. Gut microbiota from male rat
feces following intermittent access to an energy-rich ‘cafeteria-style’
Phenotype-driven genetics of binge-like eating. Phenotypic dif- diet showed changes in microbial flavonoid, bile acid, d-arginine,
ferences in BLE between inbred rodent strains implicate underly- d-ornithine, fatty acid, and geraniol biosynthetic pathways that cor-
ing causal genes92,93. In an intermittent, limited-access paradigm related with body weight, adipose tissue, glucose, leptin, and insu-
for BLE of sweetened palatable food, cytoplasmic FMR-interacting lin103. Although similar studies are needed in humans, these data
protein 2 (encoded by Cyfip2) was mapped and validated in a cross suggest that microbial pathways could be targeted to normalize eat-
between C57BL/6 substrains. Cyfip2+/− mice showed a decrease in ing and metabolic function in binge eating.
compulsive BLE of palatable food but not chow94. Cyfip1+/− mice To summarize, omics analyses of relevant tissues at appropri-
also showed modulation of BLE, depending on parent of origin, ate time points in animal models for eating disorders offer distinct,
genetic background, and sex95. CYFIP1 CNV is implicated in autism complementary advantages to human studies and will continue to
spectrum disorder, ADHD, psychosis96, and possibly AN, although provide unique insights into the hedonic and homeostatic adap-
laboratory validation of the CNV calling failed39. The study of BLE tations that could inform therapeutics. Furthermore, combining

548 Nature Neuroscience | VOL 25 | May 2022 | 543–554 | www.nature.com/natureneuroscience


NaTUrE NEUrOsciEncE Review Article
LOD

Proteomics

Behavioral genetics Network analysis

QTL mapping (behavior, GWAS Metabolomics


physiology, mRNA, protein) Omics

Model behaviors

ABA BLE

Neural circuits and behavior In vivo gene editing


and gene validation
Designer ligands
(PSEM, CNO, CLZ, C21)

G G
T T
T C
A A
C C

Synaptic function Optogenetics Chemogenetics

Fig. 3 | Forward and backward translation of eating disorder-relevant traits at different levels of biological hierarchy. ABA and BLE are behavioral
models for restrictive eating in AN and for binge eating in BN and BED. Within the use of animal models, there are four primary approaches that seek to
elucidate the fundamental biology of eating disorders: QTL mapping, omics, neural circuit manipulations, and in vivo gene editing. QTL mapping is used to
discover genetic loci and ultimately candidate causal genes and variants that regulate phenotypic traits at the molecular, cellular, or behavioral level. Omics
investigations are carried out at the genomic, transcriptomic (not shown), proteomic, microbiomic, or metabolomic level and can reveal novel biological
pathways that regulate feeding and/or metabolism. In vivo gene editing research can be used to establish causality for candidate genomic variants
identified from rodent QTL/GWAS or in silico studies. Neural circuit approaches are also used to establish causal molecular, cellular or circuit elements
that drive eating behavior and metabolism. They span the range of observing neural activity and synaptic function, manipulating those circuits in real time
(for example, via optogenetic, chemogenetic, or pharmacologic approaches), and layering these experiments with pathological models.

omics with phenotype-driven forward genetic studies can further causal neurobiological factors of AN. Viral overexpression of the
inform the mechanisms of gene dysfunction and the consequent D2 dopamine receptor in D2-containing mouse neurons of the NAc
genomic adaptations that drive and sustain disordered eating. core induced hyperactivity, increased food intake, and enhanced
Expanding these latter approaches would provide valuable triangu- ABA-induced wheel running and weight loss in females107. Only
lation of omics studies in humans. females showed severe weight loss during scheduled fasting alone,
even without a change in food intake or hyperactivity. More recently,
Viral overexpression and neural circuit studies of activity-based chemogenetic activation of the projection from the medial PFC to
anorexia. Contemporary circuit approaches to understand feed- the NAc shell decreased cognitive flexibility and increased ABA,
ing suppression and stimulation have been reviewed elsewhere (for whereas its inhibition prevented ABA weight loss and increased
example, refs. 104–106). Here, we distinguish our discussion from the cognitive flexibility108. Zhang and Dulawa propose that projections
physiology of homeostatic meal termination—adaptive anorexi- from the NAc shell to the lateral hypothalamus (LH) and, in turn,
genic responses—and focus on related studies involving ABA that from the LH to the VTA, could link ABA-induced mesocortico-
more holistically model AN pathology and behavioral phenotypes. limbic reward dysfunction with ABA-induced changes in metabo-
These studies provide more direct evidence as to the associative and lism109 (Fig. 4).

Nature Neuroscience | VOL 25 | May 2022 | 543–554 | www.nature.com/natureneuroscience 549


Review Article NaTUrE NEUrOsciEncE

Gs + GI
DREADD
ABA ABA Leptin
PFC Wheel
Gs running
D2R ABA DREADD
ABA
Shell
NAc VTA
Core
LH

Fig. 4 | Mesocorticolimbic reward dysfunction in activity-based anorexia. Brown receptor indicates excitation of cell type and pathway. Red receptor
indicates inhibition of pathway. Blue receptor indicates overexpression. Overexpression of D2 dopamine receptors in medium spiny neurons of NAc
core increased ABA phenotypes and when combined with scheduled fasting alone (no wheel running) was sufficient to induce weight loss and glucose
intolerance in females without affecting food intake107. Chemogenetic activation of the mPFC→NAc shell pathway decreased cognitive flexibility and
increased ABA; inhibition had the opposite effect108. Chemogenetic activation of VTA neurons decreased ABA and increased survival110. Leptin injections
into the VTA decreased wheel running112. Brown, dashed synapses from the NAc shell to the LH and from the LH to the VTA indicate a pathway proposed by
Zhang and Dulawa109 that could mediate mesocorticolimbic reward modulation of energy expenditure and metabolism in ABA. Additional work is necessary
to delineate the circuits, neurotransmitters, and hormones that link ABA reward dysfunction with increased energy expenditure. DREADD, designer receptor
exclusively activated by designer drugs; Gi, G-inhibitory DREADDS; Gs, G-stimulatory DREADDS; D2R, D2 dopamine receptor overexpression.

The NAc receives strong input from VTA DA-producing neu- consumption by transient phasic activation of VTA DA neurons.
rons, and specific chemogenetic activation of NAc-projecting VTA Conversely, persistent activation of VTA DA neurons using chemo-
DA neurons increases food intake, food anticipatory activity, and genetics or via the 5-HT2C receptor agonist lorcaserin resulted in
survival in the ABA model110. These results implicate female-specific a reduction in binge eating116. Of note, direct stimulation of VTA
metabolic dysfunction induced by NAc D2 overexpression107 and DA neurons does not stimulate eating per se117. These data suggest
suggest that NAc D2 receptor density and signaling could affect that the observed contribution of VTA DA neurons to BLE is com-
risk for AN and predispose individuals toward excessive exercise, plex and sensitive to the timescales of experimental manipulations.
potentially as a means to alleviate underlying reward deficiency— These findings also underscore the critical need to understand how
although further studies are needed to support this hypothesis. VTA DA neuronal activity is altered across multiple binge-eating
Combining these results with genetic studies implicating striatal episodes. Finally, the incorporation of pathological BLE models that
neurons that express either D1 or D2 and human imaging studies, pair food consumption with aversive consequences (for example,
we propose that striatal circuits are a biological risk hub that war- footshock) will be useful in distinguishing between patterns of VTA
rant further investigation. neuronal activity that accompany adaptive hedonic food consump-
Moreover, animal, clinical and genetic findings implicate leptin tion versus repetitive compulsive-like BLE that is insensitive to neg-
in the risk and maintenance of AN. Hyperactivity, eating restraint ative reinforcement.
and earlier weight loss after inpatient refeeding are correlated with Studies of BLE have also revealed important changes in neural
lower leptin levels in patients with AN111, implicating that a loss of circuits receiving robust DA inputs and how DA receptors modu-
leptin signaling in VTA underlies ABA hyperactivity. In rats with late neural activity in these sites. The VTA and substantia nigra
ABA, leptin treatment reduced hyperactivity via the VTA region112, pars compacta DA neurons project to the ventral (NAc) and dor-
implicating that a loss of leptin signaling in VTA underlies ABA sal striatum. Released DA then shapes ongoing neural activity in
hyperactivity. GWAS-based genetic correlations suggest an overlap medium spiny neurons, primarily via modulation of downstream
in the biological regulation of AN, leptin, and physical activity8. A PKA-dependent signaling cascades. In the NAc, a multiday course
case report suggested that leptin treatment may reduce hyperactiv- of binge eating increases delta band oscillations of local field
ity and eating disorder-related cognitions in AN113. Although it has potentials and single-unit activity in anticipation of palatable food
not been studied in AN, human neuroimaging studies in healthy intake118. Palatable food consumption was specifically disrupted
individuals demonstrate that leptin regulates mesolimbic DA sys- via targeted stimulation during anticipatory periods, a phenom-
tems under stress114. Accordingly, leptin is an important hormone in enon that may require activation of D2 receptors119. Interestingly,
linking genetics with alterations in the DA reward system. delta band oscillations in the ventral striatum were observed in
humans118. Anticipatory activity, or ramps, are also visible in the
Optogenetic and chemogenetic analysis of binge-like eating. dorsal striatum at the level of single neurons or bulk calcium tran-
Optogenetic and chemogenetic approaches seek to identify and sients during food approach120. These ramps rapidly terminate dur-
manipulate specific cell types and fibers to test proposed circuitry ing food consumption and their functional requirement to binge
in feeding115 (see reviews, for example, ref. 104; Fig. 5). Here, we focus eating is unknown.
on studies using BLE paradigms directly or indirectly associated Modulation of the dorsal and ventral striatum also occurs from
with the mesolimbic reward pathway. PFC and IC inputs. The IC integrates taste, interoception, and moti-
Several studies have found that activation of inputs to VTA DA vation to regulate feeding, and food-predictive cues reliably acti-
neurons from the LH, the bed nucleus of the stria terminalis, or vate IC neurons121,122. Chemogenetic activation of the anterior IC
the dorsal raphe nucleus produces positive valence and appetitive as a whole decreases palatable food intake and cue reactivity in a
behavior, and can increase compulsive-like eating or consump- rat model of binge eating123, and optogenetic activation of the right
tion of liquid rewards104. Broadly, these studies support a physi- anterior IC in mice similarly reduces food intake124. Conversely, in a
ological role for these brain circuits in shaping motivation and food model of compulsive binge eating in rats, pathway-specific inhibition

550 Nature Neuroscience | VOL 25 | May 2022 | 543–554 | www.nature.com/natureneuroscience


NaTUrE NEUrOsciEncE Review Article
Feeding
BLE
BLE IC
PFCVIP

PFC
IC DR
BLE

vm vm Other BLE BNST


PFC PFC NAc VTA DRPET-1
sites
LH
BNSTGABA
Feeding
Reward

D1R D2R Reward


DA GABA

ChR2 473-nm light Feeding


NAc
Arch3.0 VTA GLU
Gq DREADD (hM3D) 532-nm light
Delta band oscillations LHGABA Feeding
during BLE development

Fig. 5 | Mesolimbic-centered neural circuits that modulate binge-like eating. Behavioral neuroscientists have used circuit-level techniques such as
chemogenetics and optogenetics to study the VTA to NAc dopaminergic circuit in BLE, normal feeding, and reward-like behavior. Inputs to the VTA from
the dorsal raphe (DR), the bed nucleus of the stria terminalis (BNST), and the LH modulate reward and food consumption as shown by pathway-specific
optogenetics. Within the VTA, chemogenetic activation of VTA DA neuron reduces BLE, and direct optogenetic activation has no impact on feeding.
Within the NAc, pathway-specific optogenetic inhibition of the inputs from the IC reduces BLE. Similarly, chemogenetic activation of the NAc-projecting
input cells from the ventromedial prefrontal cortex (vmPFC) or VIP-expressing neurons in the prelimbic and infralimbic PFC also reduces BLE. ChR2,
channelrhodopsin2; D1R, dopamine D1 receptor; D2R, dopamine D2 receptor; GLU, glutamate; PET-1, PC12 ETS domain-containing transcription factor.

of the IC to NAc decreased appetitive behavior to receive palat- advance our understanding of the central neuronal adaptations
able food intermittently paired with foot shock125. These studies as well as peripheral cell-type-specific adaptations that drive mal-
indicate a complex role of the insula in reinforcement and compul- adaptive feeding. These assessments should incorporate hypotheses
sive BLE. In the PFC, activation of neurons projecting to the NAc informed by emergent GWASs and human imaging data and can, in
shell reduced food intake in a binge-eating model in rats predis- turn, contextualize and inform studies in humans.
posed to high impulsivity, a trait observed in a previous study126. Genetic, neuroimaging and animal model research studies in
Optogenetic activation of inhibitory vasoactive intestinal polypep- eating disorders have largely represented independent disciplines.
tide (VIP)-expressing interneurons of infralimbic and prelimbic These sciences of eating disorders have now matured adequately that
medial prefrontal cortex (mPFC) in male mice decreased BLE of cross-communication is both possible and essential to strengthen
a high-caloric diet127. These findings implicate reduced function causal inferences and translate observations into biological under-
of the PFC→NAc circuit in impulsivity and BLE. Further study of standing and novel therapeutics.
these circuits across the acquisition of BLE behavior (first episode
versus last episode) is necessary to investigate the dynamics and Conclusions and future directions. As human GWAS and neu-
link to pathology more precisely. Nonetheless, these findings dem- roimaging studies grow and diversify and novel animal models
onstrate that BLE can be inhibited in real time through neural cir- are developed, efforts to bridge disciplines must expand. Ongoing
cuit manipulations, demonstrating that ongoing activity of specific GWASs of BED and BN will give context to findings from mouse
cortical and limbic circuits is required for BLE. BLE models. Gene-driven approaches in neurobiology and behav-
ior should increasingly incorporate evidence for association of tar-
Animal model limitations. One of the primary limitations in the get genes from human GWASs. Interdisciplinary efforts will benefit
use of animal models is their inability to capture complex psycho- studies investigating the main effects of genes and variants, as well
logical constructs that are important to the etiology of eating dis- as gene–gene and gene–environment interactions. Exquisite control
orders, such as a drive for thinness, body dysmorphia, or intense over environmental factors in animal models will allow rigorous
fear of weight gain in the case of AN and a loss of control for BLE. testing of putative interactions from genome-wide observational
However, emerging technologies in markerless pose estimation and epidemiology in eating disorders. Finally, eating disorder research
deep learning provide basic scientists the opportunity to extract must be proactive in embracing other disciplines. For example, the
novel behavioral phenotypes that may be associated with these utility and diversity of induced pluripotent stem cell models has
constructs128. The other major limitation of animal models is that, increased rapidly, and the application of these models to eating dis-
to date, animal models have not yet been used to test the causal order research could yield valuable new insights129. Our hope is to
nature of a sum of polygenic risk-associated common variants from accelerate target identification by applying robust statistical genetic
an eating disorder group. This is an important technical and con- and pathway analysis methods, large-scale brain and neurodevel-
ceptual factor to consider as we move closer to testing causality of opmental systems biology approaches, and innovative chemoin-
disease-associated variants. formatics130. To do this, eating disorder researchers should seek to
communicate across disciplines, including establishing meetings
Integration. Circuit approaches in animal models provide increas- dedicated to translational eating disorders research. Ultimately, the
ing specificity with regard to cell types and their connections under- goal is to translate genetic and neuroscience findings directly into
lying ABA and BLE. Convergent evidence across studies suggests the clinic to enable biologically informed tailored treatment selec-
that longitudinal assessment of neuronal function in mesolimbic tion and delivery and to eliminate morbidity and mortality from
and nigrostriatal circuits, combined with single-cell omics, will these debilitating illnesses.

Nature Neuroscience | VOL 25 | May 2022 | 543–554 | www.nature.com/natureneuroscience 551


Review Article NaTUrE NEUrOsciEncE

Received: 3 May 2020; Accepted: 1 April 2022; 29. Tyrrell, J. et al. Genetic evidence for causal relationships between maternal
Published online: 6 May 2022 obesity-related traits and birth weight. JAMA 315, 1129–1140 (2016).
30. Adams, D. M., Reay, W. R., Geaghan, M. P. & Cairns, M. J. Investigating the
effect of glycaemic traits on the risk of psychiatric illness using Mendelian
References randomisation. Neuropsychopharmacology 46, 1093–1102 (2020).
1. American Psychiatric Association. Diagnostic and Statistical Manual of
31. Huckins, L. M. et al. Investigation of common, low-frequency and rare
Mental Disorders (5th edn) (American Psychiatric Publishing, 2013).
genome-wide variation in anorexia nervosa. Mol. Psychiatry 23, 1169–1180
2. Erskine, H. E., Whiteford, H. A. & Pike, K. M. The global burden of eating
(2018).
disorders. Curr. Opin. Psychiatry 29, 346–353 (2016).
32. Scott-Van Zeeland, A. A. et al. Evidence for the role of EPHX2 gene
3. Santomauro, D. F. et al. The hidden burden of eating disorders: an
variants in anorexia nervosa. Mol. Psychiatry 19, 724–732 (2013).
extension of estimates from the Global Burden of Disease Study 2019.
33. Lutter, M. et al. Novel and ultra-rare damaging variants in neuropeptide
Lancet Psychiatry 8, 320–328 (2021).
signaling are associated with disordered eating behaviors. PLoS ONE 12,
4. Schaumberg, K. et al. Patterns of diagnostic flux in eating disorders: a
e0181556 (2017).
longitudinal population study in Sweden. Psychol. Med. 49,
34. Cui, H. et al. Eating disorder predisposition is associated with ESRRA and
432–450 (2019).
HDAC4 mutations. J. Clin. Invest. 123, 4706–4713 (2013).
5. Southern, J. et al. Multi-scale computational modelling in biology and
35. Lombardi, L. et al. Anorexia nervosa is associated with Neuronatin variants.
physiology. Prog. Biophys. Mol. Biol. 96, 60–89 (2008).
Psychiatr. Genet. 29, 103–110 (2019).
6. Yilmaz, Z., Hardaway, J. & Bulik, C. Genetics and epigenetics of eating
36. Wang, K. et al. A genome-wide association study on common SNPs and
disorders. Adv. Genomics Genet. 5, 131–150 (2015).
rare CNVs in anorexia nervosa. Mol. Psychiatry 16, 949–959 (2011).
7. Dellava, J. E., Thornton, L. M., Lichtenstein, P., Pedersen, N. L. & Bulik, C.
37. Yilmaz, Z. et al. Exploration of large, rare CNVs associated with psychiatric
M. Impact of broadening definitions of anorexia nervosa on sample
and neurodevelopmental disorders in individuals with anorexia nervosa.
characteristics. J. Psychiatr. Res. 45, 691–698 (2011).
Psychiatr. Genet 27, 152–158 (2017).
8. Watson, H. et al. Genome-wide association study identifies eight risk loci
38. Boraska, V. et al. A genome-wide association study of anorexia nervosa.
and implicates metabo-psychiatric origins for anorexia nervosa. Nat. Genet.
Mol. Psychiatry 19, 1085–1094 (2014).
51, 1207–1214 (2019).
39. Chang, X. et al. Microduplications at the 15q11.2 BP1–BP2 locus are enriched
9. Duncan, L. et al. Significant locus and metabolic genetic correlations
revealed in genome-wide association study of anorexia nervosa. Am. J. in patients with anorexia nervosa. J. Psychiatr. Res. 113, 34–38 (2019).
Psychiatry 174, 850–858 (2017). 40. Bulik, C. M. et al. The Eating Disorders Genetics Initiative (EDGI): study
10. Thornton, L. et al. The Anorexia Nervosa Genetics Initiative: overview and protocol. BMC Psychiatry 21, 234 (2021).
methods. Contemp. Clin. Trials 74, 61–69 (2018). 41. Co, M., Anderson, A. G. & Konopka, G. FOXP transcription factors in
11. Song, W., Wang, W., Yu, S. & Lin, G. N. Dissection of the genetic vertebrate brain development, function, and disorders. Wiley Interdiscip.
association between anorexia nervosa and obsessive–compulsive disorder at Rev. Dev. Biol. 9, e375 (2020).
the network and cellular levels. Genes 12, 491 (2021). 42. Yengo, L. et al. Meta-analysis of genome-wide association studies for height
12. Munn-Chernoff, M. A. et al. Shared genetic risk between eating disorder- and body mass index in ∼700,000 individuals of European ancestry. Hum.
and substance-use-related phenotypes: evidence from genome-wide Mol. Genet. 27, 3641–3649 (2018).
association studies. Addict. Biol. 26, e12880 (2021). 43. Zhang, M. et al. Axonogenesis is coordinated by neuron-specific alternative
13. Kaye, W. et al. Comorbidity of anxiety disorders with anorexia and bulimia splicing programming and splicing regulator PTBP2. Neuron 101, 690–706
nervosa. Am. J. Psychiatry 161, 2215–2221 (2004). (2019).
14. Cederlöf, M. et al. Etiological overlap between obsessive–compulsive 44. Arends, R. M. et al. Associations between the CADM2 gene, substance use,
disorder and anorexia nervosa: a longitudinal cohort, family and twin study. risky sexual behavior and self-control: a phenome-wide association study.
World Psychiatry 14, 333–338 (2015). Addict. Biol. 26, e13015 (2021).
15. Thornton, L. M., Welch, E., Munn-Chernoff, M. A., Lichtenstein, P. & Bulik, 45. Rathjen, T. et al. Regulation of body weight and energy homeostasis by
C. M. Anorexia nervosa, major depression, and suicide attempts: shared neuronal cell adhesion molecule 1. Nat. Neurosci. 20, 1096–1103 (2017).
genetic factors. Suicide Life Threat. Behav. 46, 525–534 (2016). 46. Gerson, S. L. MGMT: its role in cancer aetiology and cancer therapeutics.
16. Dellava, J., Kendler, K. & Neale, M. Generalized anxiety disorder and Nat. Rev. Cancer 4, 296–307 (2004).
anorexia nervosa: evidence of shared genetic variation. Depress. Anxiety 28, 47. Reid, D. A. et al. Incorporation of a nucleoside analog maps genome repair
728–733 (2011). sites in postmitotic human neurons. Science 372, 91–94 (2021).
17. Bulik-Sullivan, B. et al. An atlas of genetic correlations across human 48. Wu, W. et al. Neuronal enhancers are hotspots for DNA single-strand break
diseases and traits. Nat. Genet. 47, 1236–1241 (2015). repair. Nature 593, 440–444 (2021).
18. Watson, H. J. et al. Common genetic variation and age at onset of anorexia 49. Finucane, H. et al. Partitioning heritability by functional annotation using
nervosa. Biol. Psychiatry https://doi.org/10.1016/j.bpsgos.2021.09.001 (2021). genome-wide association summary statistics. Nat. Genet. 47, 1228–1235
19. Solmi, F., Mascarell, M. C., Zammit, S., Kirkbride, J. B. & Lewis, G. (2015).
Polygenic risk for schizophrenia, disordered eating behaviours and body 50. Skene, N. G. et al. Genetic identification of brain cell types underlying
mass index in adolescents. Br. J. Psychiatry 215, 428–433 (2019). schizophrenia. Nat. Genet. 50, 825–833 (2018).
20. Yilmaz, Z., Gottfredson, N., Zerwas, S., Bulik, C. & Micali, N. 51. Chowdhury, T. G. et al. Voluntary wheel running exercise evoked by
Developmental premorbid body mass index trajectories of adolescents with food-restriction stress exacerbates weight loss of adolescent female rats but
eating disorders in a longitudinal population cohort. J. Am. Acad. Child also promotes resilience by enhancing GABAergic inhibition of pyramidal
Adolesc. Psychiatry 58, 191–199 (2019). neurons in the dorsal hippocampus. Cereb. Cortex 29, 4035–4049 (2019).
21. Abdulkadir, M. et al. Polygenic score for body mass index is associated 52. Klenowski, P. M. et al. Prolonged consumption of sucrose in a binge-like
with disordered eating in a general population cohort. J. Clin. Med. 9, manner, alters the morphology of medium spiny neurons in the nucleus
1187 (2020). accumbens shell. Front. Behav. Neurosci. 10, 54 (2016).
22. Hübel, C. et al. One size does not fit all. Genomics differentiates among 53. Wang, D. et al. Comprehensive functional genomic resource and integrative
anorexia nervosa, bulimia nervosa, and binge-eating disorder. Int. J. Eat. model for the human brain. Science 362, eaat8464 (2018).
Disord. 54, 785–793 (2021). 54. Rossi, M. A. & Stuber, G. D. Overlapping brain circuits for homeostatic and
23. Murray, G. K. et al. Could polygenic risk scores be useful in psychiatry?: a hedonic feeding. Cell Metab. 27, 42–56 (2018).
review. JAMA Psychiatry 78, 210–219 (2020). 55. Frank, G. K., Shott, M. E. & DeGuzman, M. C. Recent advances in
24. Wray, N. et al. Research review: polygenic methods and their application to understanding anorexia nervosa. F1000Res. 8, 504 (2019).
psychiatric traits. J. Child Psychol. Psychiatry 55, 1068–1087 (2014). 56. Kaye, W. H., Wagner, A., Fudge, J. L. & Paulus, M. Neurocircuity of eating
25. Lee, P. H. et al. Genomic relationships, novel loci, and pleiotropic disorders. Curr. Top. Behav. Neurosci. 6, 37–57 (2010).
mechanisms across eight psychiatric disorders. Cell 179, 1469–1482 (2019). 57. Lammel, S., Lim, B. K. & Malenka, R. C. Reward and aversion in a
26. Lee, J. E., Namkoong, K. & Jung, Y.-C. Impaired prefrontal cognitive heterogeneous midbrain dopamine system. Neuropharmacol 76, 351–359
control over interference by food images in binge-eating disorder and (2014).
bulimia nervosa. Neurosci. Lett. 651, 95–101 (2017). 58. Cowdrey, F. A., Park, R. J., Harmer, C. J. & McCabe, C. Increased neural
27. Yilmaz, Z. et al. Examination of the shared genetic basis of anorexia processing of rewarding and aversive food stimuli in recovered anorexia
nervosa and obsessive–compulsive disorder. Mol. Psychiatry 25, 2036–2046 nervosa. Biol. Psychiatry 70, 736–743 (2011).
(2020). 59. Kaye, W. H. et al. Neural insensitivity to the effects of hunger in women
28. Reed, Z. E., Micali, N., Bulik, C. M., Davey Smith, G. & Wade, K. H. remitted from anorexia nervosa. Am. J. Psychiatry 177, 601–610 (2020).
Assessing the causal role of adiposity on disordered eating in childhood, 60. Bohon, C. & Stice, E. Reward abnormalities among women with full and
adolescence, and adulthood: a Mendelian randomization analysis. Am. J. subthreshold bulimia nervosa: a functional magnetic resonance imaging
Clin. Nutr. 106, 764–772 (2017). study. Int. J. Eat. Disord. 44, 585–595 (2011).

552 Nature Neuroscience | VOL 25 | May 2022 | 543–554 | www.nature.com/natureneuroscience


NaTUrE NEUrOsciEncE Review Article
61. Simon, J. J. et al. Neural signature of food reward processing in bulimic-type 89. Gelegen, C. et al. Difference in susceptibility to activity-based anorexia in
eating disorders. Soc. Cogn. Affect. Neurosci. 11, 1393–1401 (2016). two inbred strains of mice. Eur. Neuropsychopharmacol. 17, 199–205 (2007).
62. Bailer, U. F. et al. Exaggerated 5-HT1A but normal 5-HT2A receptor activity 90. Gelegen, C. et al. Chromosomal mapping of excessive physical activity in
in individuals ill with anorexia nervosa. Biol. Psychiatry 61, 1090–1099 mice in response to a restricted feeding schedule. Eur.
(2007). Neuropsychopharmacol. 20, 317–326 (2010).
63. Broft, A. et al. Striatal dopamine type 2 receptor availability in anorexia 91. Pjetri, E. et al. Identifying predictors of activity based anorexia susceptibility
nervosa. Psychiatry Res. 233, 380–387 (2015). in diverse genetic rodent populations. PLoS ONE 7, e50453 (2012).
64. Bailer, U. F. et al. Amphetamine induced dopamine release increases anxiety 92. Babbs, R. K. et al. Genetic differences in the behavioral organization of
in individuals recovered from anorexia nervosa. Int. J. Eat. Disord. 45, binge eating, conditioned food reward, and compulsive-like eating in
263–271 (2012). C57BL/6J and DBA/2J strains. Physiol. Behav. 197, 51–66 (2018).
65. Broft, A. et al. Striatal dopamine in bulimia nervosa: a PET imaging study. 93. Newmyer, B. A., Whindleton, C. M., Srinivasa, N., Jones, M. K. & Scott, M.
Int. J. Eat. Disord. 45, 648–656 (2012). M. Genetic variation affects binge feeding behavior in female inbred mouse
66. Mihov, Y. et al. Metabotropic glutamate receptor 5 in bulimia nervosa. Sci. strains. Sci. Rep. 9, 1–10 (2019).
Rep. 10, 1–10 (2020). 94. Kirkpatrick, S. L. et al. Cytoplasmic FMR1-interacting protein 2 is a major
67. Frank, G. K., Shott, M. E., Hagman, J. O. & Mittal, V. A. Alterations in genetic factor underlying binge eating. Biol. Psychiatry 81, 757–769 (2017).
brain structures related to taste reward circuitry in ill and recovered 95. Babbs, R. K. et al. Cyfip1 haploinsufficiency increases compulsive-like
anorexia nervosa and in bulimia nervosa. Am. J. Psychiatry 170, 1152–1160 behavior and modulates palatable food intake in mice: dependence on
(2013). Cyfip2 genetic background, parent of origin and sex. G3 9, 3009–3022
68. Craig, A. D. & Craig, A. How do you feel—now? The anterior insula and (2019).
human awareness. Nat. Rev. Neurosci. 10, 59–70 (2009). 96. Farrell, M. et al. Treatment-resistant psychotic symptoms and the 15q11.2
69. Kerr, K. L. et al. Altered insula activity during visceral interoception in BP1–BP2 (Burnside–Butler) deletion syndrome: case report and review of
weight-restored patients with anorexia nervosa. Neuropsychopharmacology the literature. Transl. Psychiatry 10, 1–8 (2020).
41, 521–528 (2016). 97. Yao, E. J. et al. Systems genetic analysis of binge-like eating in a
70. Zucker, N. L. et al. The clinical significance of posterior insular volume in C57BL/6J × DBA/2J-F2 cross. Genes Brain Behav. e12751 (2021).
adolescent anorexia nervosa. Psychosom. Med. 79, 1025–1035 (2017). 98. Nobis, S. et al. Alterations of proteome, mitochondrial dynamic and
71. Berner, L. A. et al. Altered anticipation and processing of aversive autophagy in the hypothalamus during activity-based anorexia. Sci. Rep. 8,
interoceptive experience among women remitted from bulimia nervosa. 1–15 (2018).
Neuropsychopharmacology 44, 1265–1273 (2019). 99. Nobis, S. et al. Colonic mucosal proteome signature reveals reduced energy
72. Ting, J. T. & Feng, G. Neurobiology of obsessive–compulsive disorder: metabolism and protein synthesis but activated autophagy during
insights into neural circuitry dysfunction through mouse genetics. Curr. anorexia‐induced malnutrition in mice. Proteomics 18, 1700395 (2018).
Opin. Neurobiol. 21, 842–848 (2011). 100. Breton, J. et al. Proteome modifications of gut microbiota in mice with
73. Marsh, R. et al. An fMRI study of self-regulatory control and conflict activity-based anorexia and starvation: role in ATP production. Nutrition
resolution in adolescents with bulimia nervosa. Am. J. Psychiatry 168, 67, 110557 (2019).
1210–1220 (2011). 101. Schroeder, M. et al. Placental miR-340 mediates vulnerability to activity
74. Skunde, M. et al. Neural signature of behavioural inhibition in women with based anorexia in mice. Nat. Commun. 9, 1–15 (2018).
bulimia nervosa. J. Psychiatry Neurosci. 41, E69–E78 (2016). 102. He, X., Stefan, M., Terranova, K., Steinglass, J. & Marsh, R. Altered white
75. Wierenga, C. et al. Altered BOLD response during inhibitory and error matter microstructure in adolescents and adults with bulimia nervosa.
processing in adolescents with anorexia nervosa. PLoS ONE 9, e92017 Neuropsychopharmacology 41, 1841–1848 (2015).
(2014). 103. Kaakoush, N. O. et al. Alternating or continuous exposure to cafeteria diet
76. Oberndorfer, T. A., Kaye, W. H., Simmons, A. N., Strigo, I. A. & Matthews, leads to similar shifts in gut microbiota compared to chow diet. Mol. Nutr.
S. C. Demand‐specific alteration of medial prefrontal cortex response Food Res. 61, 1500815 (2017).
during an inhibition task in recovered anorexic women. Int. J. Eat. Disord. 104. Sweeney, P. & Yang, Y. Neural circuit mechanisms underlying emotional
44, 1–8 (2011). regulation of homeostatic feeding. Trends Endocrinol. Metab. 28, 437–448
77. Finch, J. E., Palumbo, I. M., Tobin, K. E. & Latzman, R. D. Structural brain (2017).
correlates of eating pathology symptom dimensions: a systematic review. 105. Webber, E. S., Bonci, A. & Krashes, M. J. The elegance of energy balance:
Psychiatry Res. Neuroimaging 317, 111379 (2021). insight from circuit‐level manipulations. Synapse 69, 461–474 (2015).
78. Geisler, D. et al. Altered white matter connectivity in young acutely 106. Andermann, M. L. & Lowell, B. B. Toward a wiring diagram understanding
underweight patients with anorexia nervosa. J. Am. Acad. Child Adolesc. of appetite control. Neuron 95, 757–778 (2017).
Psychiatry 61, 331–340 (2022). 107. Welch, A. C. et al. Dopamine D2 receptor overexpression in the nucleus
79. Goodkind, M. et al. Identification of a common neurobiological substrate accumbens core induces robust weight loss during scheduled fasting
for mental illness. JAMA Psychiatry 72, 305–315 (2015). selectively in female mice. Mol. Psychiatry 26, 3765–3777 (2019).
80. McTeague, L. M. et al. Identification of common neural circuit disruptions 108. Milton, L. K. et al. Suppression of corticostriatal circuit activity improves
in emotional processing across psychiatric disorders. Am. J. Psychiatry 177, cognitive flexibility and prevents body weight loss in activity-based anorexia
411–421 (2020). in rats. Biol. Psychiatry 90, 819–828 (2021).
81. Shanmugan, S. et al. Common and dissociable mechanisms of executive 109. Zhang, J. & Dulawa, S. C. The utility of animal models for studying the
system dysfunction across psychiatric disorders in youth. Am. J. Psychiatry metabo-psychiatric origins of anorexia nervosa. Front. Psychiatry 12, 711181
173, 517–526 (2016). (2021).
82. Wang, S. et al. Neurobiological commonalities and distinctions among 3 110. Foldi, C. J., Milton, L. K. & Oldfield, B. J. The role of mesolimbic reward
major psychiatric disorders: a graph theoretical analysis of the structural neurocircuitry in prevention and rescue of the activity-based anorexia
connectome. J. Psychiatry Neurosci. 45, 15–22 (2020). phenotype in rats. Neuropsychopharmacology 42, 2292–2300 (2017).
83. Xia, M. et al. Shared and distinct functional architectures of brain networks 111. Hebebrand, J., Muller, T., Holtkamp, K. & Herpertz-Dahlmann, B. The role
across psychiatric disorders. Schizophr Bull. 45, 450–463 (2019). of leptin in anorexia nervosa: clinical implications. Mol. Psychiatry 12,
84. Hudson, J. I., Hiripi, E., Pope, H. G. Jr. & Kessler, R. C. The prevalence and 23–35 (2007).
correlates of eating disorders in the National Comorbidity Survey 112. Verhagen, L. A., Luijendijk, M. C. & Adan, R. A. Leptin reduces
Replication. Biol. Psychiatry 61, 348–358 (2007). hyperactivity in an animal model for anorexia nervosa via the ventral
85. Frank, G. K., Favaro, A., Marsh, R., Ehrlich, S. & Lawson, E. A. Toward tegmental area. Eur. Neuropsychopharmacol. 21, 274–281 (2011).
valid and reliable brain imaging results in eating disorders. Int. J. Eat. 113. Antel, J. et al. Rapid amelioration of anorexia nervosa in a male adolescent
Disord. 51, 250–261 (2018). during metreleptin treatment including recovery from hypogonadotropic
86. Kaufmann, L.-K. et al. Fornix under water? Ventricular enlargement biases hypogonadism. Eur. Child Adolesc. Psychiatry https://doi.org/10.1007/
forniceal diffusion magnetic resonance imaging indices in anorexia nervosa. s00787-021-01778-7 (2021).
Biol. Psychiatry Cogn. Neurosci. Neuroimaging 2, 430–437 (2017). 114. Burghardt, P. R. et al. Leptin regulates dopamine responses to sustained
87. Gaudio, S., Carducci, F., Piervincenzi, C., Olivo, G. & Schiöth, H. B. Altered stress in humans. J. Neurosci. 32, 15369–15376 (2012).
thalamo–cortical and occipital–parietal–temporal–frontal white matter 115. Wu, Q., Han, Y. & Tong, Q. in Neural Regulation of Metabolism (eds. Wu,
connections in patients with anorexia and bulimia nervosa: a systematic Q. & Zheng, R.) 211–233 (Springer, 2018).
review of diffusion tensor imaging studies. J. Psychiatry Neurosci. 44, 116. Xu, P. et al. Activation of serotonin 2C receptors in dopamine neurons
324–339 (2019). inhibits binge-like eating in mice. Biol. Psychiatry 81, 737–747 (2017).
88. Thompson, P. M. et al. ENIGMA and global neuroscience: a decade of 117. Marino, R. A. M. et al. Control of food approach and eating by a
large-scale studies of the brain in health and disease across more than 40 GABAergic projection from lateral hypothalamus to dorsal pons. Proc. Natl
countries. Transl. Psychiatry 10, 100 (2020). Acad. Sci. USA 117, 8611–8615 (2020).

Nature Neuroscience | VOL 25 | May 2022 | 543–554 | www.nature.com/natureneuroscience 553


Review Article NaTUrE NEUrOsciEncE
118. Wu, H. et al. Closing the loop on impulsivity via nucleus accumbens 140. Schalla, M. A. & Stengel, A. Activity based anorexia as an animal model for
delta-band activity in mice and man. Proc. Natl Acad. Sci. USA 115, anorexia nervosa—a systematic review. Front Nutr. 6, 69 (2019).
192–197 (2018). 141. Novelle, M. G. & Diéguez, C. Food addiction and binge eating: lessons
119. Halpern, C. H. et al. Amelioration of binge eating by nucleus accumbens learned from animal models. Nutrients 10, 71 (2018).
shell deep brain stimulation in mice involves D2 receptor modulation. J. 142. Corwin, R. L. & Babbs, R. K. Rodent models of binge eating: are they
Neurosci. 33, 7122–7129 (2013). models of addiction? ILAR J. 53, 23–34 (2012).
120. London, T. D. et al. Coordinated ramping of dorsal striatal pathways 143. Moore, C. F., Sabino, V., Koob, G. F. & Cottone, P. Pathological overeating:
preceding food approach and consumption. J. Neurosci. 38, 3547–3558 (2018). emerging evidence for a compulsivity construct. Neuropsychopharmacology
121. Livneh, Y. et al. Homeostatic circuits selectively gate food cue responses in 42, 1375–1389 (2017).
insular cortex. Nature 546, 611–616 (2017). 144. Rospond, B., Szpigiel, J., Sadakierska-Chudy, A. & Filip, M. Binge eating in
122. Kusumoto-Yoshida, I., Liu, H., Chen, B. T., Fontanini, A. & Bonci, A. preclinical models. Pharmacol. Rep. 67, 504–512 (2015).
Central role for the insular cortex in mediating conditioned responses to 145. Cottone, P., Sabino, V., Steardo, L. & Zorrilla, E. P. Intermittent access to
anticipatory cues. Proc. Natl Acad. Sci. USA 112, 1190–1195 (2015). preferred food reduces the reinforcing efficacy of chow in rats. Am. J.
123. Price, A. E., Stutz, S. J., Hommel, J. D., Anastasio, N. C. & Cunningham, K. Physiol. Regul. Integr. Comp. Physiol. 295, R1066–R1076 (2008).
A. Anterior insula activity regulates the associated behaviors of high fat food 146. Hardaway, J. A. et al. Nociceptin receptor antagonist SB 612111 decreases
binge intake and cue reactivity in male rats. Appetite 133, 231–239 (2019). high-fat diet binge eating. Behav. Brain Res. 307, 25–34 (2016).
124. Wu, Y. et al. The anterior insular cortex unilaterally controls feeding in
response to aversive visceral stimuli in mice. Nat. Commun. 11, 640 (2020). Acknowledgements
125. Spierling, S. et al. Insula to ventral striatal projections mediate compulsive C.M.B. is supported by the National Institute of Mental Health (R01MH120170,
eating produced by intermittent access to palatable food. R01MH124871, R01MH119084, R01MH118278 and R01MH124871), a Brain and
Neuropsychopharmacology 45, 579–588 (2020). Behavior Research Foundation Distinguished Investigator grant, the Swedish Research
126. Anastasio, N. C. et al. Convergent neural connectivity in motor impulsivity Council (Vetenskapsrådet, award no. 538-2013-8864), and the Lundbeck Foundation
and high-fat food binge-like eating in male Sprague-Dawley rats. (grant no. R276-2018-4581). J.R.I.C. and G.B. acknowledge that the paper represents
Neuropsychopharmacology 44, 1752–1761 (2019). independent research part funded by the National Institute for Health Research (NIHR)
127. Newmyer, B. A. et al. VIPergic neurons of the infralimbic and prelimbic Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation
cortices control palatable food intake through separate cognitive pathways. Trust and King’s College London. The views expressed are those of the authors and
JCI Insight 5, e126283 (2019). not necessarily those of the NHS, the NIHR or the Department of Health and Social
128. Mathis, A. et al. DeepLabCut: markerless pose estimation of user-defined Care. J.A.H. is supported by K01DK115902. C.D.B. is supported by U01DA050243
body parts with deep learning. Nat. Neurosci. 21, 1281–1289 (2018). and U01DA055299. L.B. is supported by T32MH112485, a Harvard Medical School
129. Maussion, G., Demirova, I., Gorwood, P. & Ramoz, N. Induced pluripotent Livingston Fellowship and the International OCD Foundation. G.B. is also supported
stem cells: new tools for investigating molecular mechanisms in anorexia by the UK Medical Research Council (MR/V012878/1, MR/V03605X/1 and MR/
nervosa. Front. Nutr. 6, 118 (2019). R024804/1) and Charlotte’s Helix.
130. Breen, G. et al. Translating genome-wide association findings into new
therapeutics for psychiatry. Nat. Neurosci. 19, 1392–1396 (2016).
131. Galmiche, M., Déchelotte, P., Lambert, G. & Tavolacci, M. P. Prevalence of Author contributions
eating disorders over the 2000–2018 period: a systematic literature review. C.M.B., J.R.I.C., J.A.H., L.B., C.D.B., H.W. and G.B. all substantially contributed to the
Am. J. Clin. Nutr. 109, 1402–1413 (2019). writing of the manuscript, approved the submitted version, are personally accountable
132. Papadopoulos, F., Ekbom, A., Brandt, L. & Ekselius, L. Excess mortality, for their own contribution, and ensure that questions related to the accuracy or integrity
causes of death and prognostic factors in anorexia nervosa. Br. J. Psychiatry of any part of the work, even ones in which the author was not personally involved, are
194, 10–17 (2009). appropriately investigated, resolved, and the resolution documented in the literature.
133. Yao, S. et al. Familial liability for eating disorders and suicide attempts:
evidence from a population registry in Sweden. JAMA Psychiatry 73, Competing interests
284–291 (2016). C.M.B. reports the following competing interests: Shire (grant recipient, Scientific
134. Fichter, M. M., Quadflieg, N., Crosby, R. D. & Koch, S. Long-term outcome Advisory Board member); Idorsia (consultant); Pearson (author, royalty recipient);
of anorexia nervosa: results from a large clinical longitudinal study. Int J. Equip Health (Clinical Advisory Board Member). G.B. reports the following competing
Eat. Disord. 50, 1018–1030 (2017). interests: Otsuka Pharma (advisory board); Illumina (grant recipient, conference
135. Berkman, N. et al. Management of eating disorders. Evid. Rep. Technol. sponsorship). The other authors declare no competing interests.
Assess. 135, 1–166 (2006).
136. Keski-Rahkonen, A. et al. Epidemiology and course of anorexia nervosa in
the community. Am. J. Psychiatry 164, 1259–1265 (2007). Additional information
137. Brownley, K. et al. Binge-eating disorder in adults: a systematic review and Correspondence should be addressed to Cynthia M. Bulik.
meta-analysis. Ann. Intern. Med. 165, 409–420 (2016). Peer review information Nature Neuroscience thanks Roger Adan, Sandra
138. Fornaro, M. et al. Lisdexamfetamine in the treatment of moderate-to-severe Sanchez-Roige, and the other, anonymous, reviewer(s) for their contribution to the peer
binge eating disorder in adults: systematic review and exploratory review of this work.
meta-analysis of publicly available placebo-controlled, randomized clinical Reprints and permissions information is available at www.nature.com/reprints.
trials. Neuropsychiatr. Dis. Treat. 12, 1827–1836 (2016).
139. Hall, J. F., Smith, K., Schnitzer, S. B. & Hanford, P. V. Elevation of activity Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
level in the rat following transition from ad libitum to restricted feeding. J. published maps and institutional affiliations.
Comp. Physiol. Psychol. 46, 429–433 (1953). © Springer Nature America, Inc. 2022

554 Nature Neuroscience | VOL 25 | May 2022 | 543–554 | www.nature.com/natureneuroscience

You might also like