+ All Categories
Home > Documents > Anorexia nervosa From single SNP studies, through ......van de rector magnificus, prof.dr. G.J. van...

Anorexia nervosa From single SNP studies, through ......van de rector magnificus, prof.dr. G.J. van...

Date post: 27-Jan-2021
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
281
Anorexia nervosa From single SNP studies, through biomarkers, to genome-wide association Marek K. Brandys
Transcript
  • Anorexia nervosa From single SNP studies, through biomarkers, to genome-wide association

    Marek K. Brandys

  • ISBN: 978-94-6182-640-4

    Printed by: Offpage, Amsterdam

    Layout: Marek K. Brandys

    Cover design: Marek K. Brandys, based on Effect of Butterfly by Anastasiya

    Markovich (Picture Labberté K.J.) via Wikimedia Commons

    © Marek K. Brandys

  • Anorexia nervosa

    From single SNP studies, through biomarkers, to genome-wide association

    Anorexia nervosa

    Van SNP studies via biomarkers naar genoomwijde associatie

    (met een samenvatting in het Nederlands)

    Anorexia nervosa

    Od badań polimorfizmów pojednynczego nukleotydu, przez biomarkery, po

    badania asocjacyjne całego genomu

    (ze streszczeniem w języku polskim)

    Proefschrift

    ter verkrijging van de graad van doctor aan de Universiteit Utrecht op gezag

    van de rector magnificus, prof.dr. G.J. van der Zwaan, ingevolge het besluit

    van het college voor promoties in het openbaar te verdedigen op

    dinsdag 19 januari 2016 des ochtends te 10.30 uur

    door

    Marek Kajetan Brandys

    geboren op 27 november 1983 te Kraków, Polska (Polen)

  • Promotoren: Prof. dr. R.A.H. Adan

    Prof. dr. A. van Elburg

    Copromotoren: Dr. M.J.H. Kas

    Dr. C. de Kovel

    This thesis was partly accomplished with financial support from the Marie

    Curie Research Training Network INTACT (Individually tailored stepped care

    for women with eating disorders; reference number: MRTN-CT-2006-

    035988)

  • Table of contents CHAPTER 1 ................................................................................................................................... 6

    Introduction Scope and outline of the thesis

    CHAPTER 2 ................................................................................................................................. 35

    Are recently identified genetic variants regulating BMI in the general population associated with anorexia nervosa?

    CHAPTER 3 ................................................................................................................................. 46

    Association study of POMC variants with body composition measures and nutrient choice CHAPTER 4 ................................................................................................................................. 69

    Anorexia nervosa and the Val158Met polymorphism of the COMT gene: meta-analysis and new data

    CHAPTER 5 ................................................................................................................................. 90

    A meta-analysis of circulating BDNF concentrations in anorexia nervosa CHAPTER 6 ............................................................................................................................... 129

    The Val66Met polymorphism of the BDNF gene in anorexia nervosa: new data and a meta-analysis

    CHAPTER 7 ............................................................................................................................... 164

    No evidence for involvement of CNVs associated with neurodevelopmental disorders in anorexia nervosa

    APPENDIX ................................................................................................................................ 193

    A genome-wide association study of anorexia nervosa CHAPTER 8 Discussion and conclusions................................................................................... 226

    Overview of genetic research in anorexia nervosa: the past, the present and the future Concluding remarks

    ADDENDUM ............................................................................................................................ 264

    English summary Nederlandse samenvatting Streszczenie w języku polskim Curriculum Vitae List of publications Acknowledgements

  • Chapter 1

    6

    Chapter 1

    Introduction

    The main focus of the present thesis is to describe the scientific undertaking

    of exploring the genetic underpinnings and biomarkers of anorexia nervosa

    (AN). We begin by introducing the history, intricate phenotypic

    manifestations, as well as the clinical and epidemiological characteristics of

    this intriguing disease.

    According to DSM-5 AN belongs to the category of feeding and

    eating disorders (ED), under the code 307.1 (F50.01) for AN restricting type

    and (F50.02) for the binge-eating/purging type. Other classes in this category

    include bulimia nervosa (BN; 307.51 (F50.2)), binge eating disorder (BED;

    307.51 (F50.8)), other specified feeding or eating disorder (OSFED; 307.59

    (F50.8)) and unspecified feeding or eating disorder (307.50 (F50.9)).

    Diagnostic criteria of AN, according to DSM-5, are listed in a later section.

    History

    It was a British royal physician, Sir William Gull, who in 1873 established the

    term ‘anorexia’ (derived from Greek ‘an-’, meaning negation, and ‘orexis’,

    signifying appetite) 1. The first medical descriptions of cases with AN are

    dated earlier than that, and ascribed to Richard Morton, also a British

    physician. Looking even further back, there exist historical accounts of

    people who appear to have suffered from this disorder. In the ancient

    Hellenistic culture fasting and self-starvation were seen as expressions of

    religious zealousness. While only a few reports are available from the

    medieval ages, a larger number of descriptions of the possible cases of AN

    comes from the times of the Renaissance. Religious ascetics would forge

    their way to sanity via starvation, self-mutilation and social isolation 2. A

  • Chapter 1

    7

    number of historical figures are suspected to have suffered from AN, such as

    Saint Catherine of Siena, Mary, Queen of Scotts or Elisabeth, Empress of

    Austria (source: http://divainternational.ch/spip.php?article97). In the

    modern times, a general interest in AN surged after the death of a famous

    musician, Karen Carpenter (4 February 1983).

    Somatic health risks

    The most striking feature of the patients suffering from AN is their low body-

    weight (85% or less than the weight expected). This symptom is

    accompanied by a refusal to consume sufficient amount of calories – an

    amount necessary to prevent further emaciation and restoration of the body

    weight to the normative levels. This continuous undernourishment damages

    body systems and, in extreme cases, leads to death. Serious medical

    complications associated with malnutrition in AN include:

    � Reduced heart rate and low blood pressure, entailing increased risk

    of heart failure

    � Amenorrhea in post-pubertal females (lack of menstruation)

    � Osteoporosis (decreased bone density)

    � Loss of muscles

    � Dehydration, possibly leading to kidney failure

    � Fainting, fatigue, general weakness

    � Hair loss, changes of complexion, growth of lanugo – a thin hair layer

    covering the body

    Furthermore, health risks associated with the purging behaviors present in

    the purging subtype of AN include:

    � Electrolyte imbalance (caused by dehydration, loss of potassium,

    chloride and sodium), possibly leading to a heart failure

    � Inflammation and possible rupture of esophagus (as a result of

    vomiting)

    � Tooth decay

    � Constipation and chronic irregular bowel movements, coming from

    abuse of laxatives (source:

  • Chapter 1

    8

    http://www.nationaleatingdisorders.org/nedaDir/files/documents/h

    andouts/HlthCons.pdf)

    Criteria and symptoms

    These serious somatic complications are paralleled by the devastation which

    the disease incurs to the psyche. Suicide is the most frequent cause of death

    in EDs 3. There is a 57-fold increase in risk of death from suicide among

    patients with AN, compared to the age-matched cohort 4. Individuals with AN

    are characterized by the immense fear of weight (fat) gain and disturbed

    body image. The criteria for diagnosis of AN established in the 5-th edition of

    The Diagnostic and Statistical Manual of Mental Disorders (DSM-5) are

    presented below:

    A. Restriction of energy intake relative to requirements leading to a

    significantly low body weight in the context of age, sex, developmental

    trajectory, and physical health.

    B. Intense fear of gaining weight or becoming fat, even though underweight.

    C. Disturbance in the way in which one's body weight or shape is

    experienced, undue influence of body weight or shape on self-evaluation, or

    denial of the seriousness of the current low body weight.

    Symptoms which might suggest AN (warning signs) include (after

    http://www.allianceforeatingdisorders.com/):

    • Significant weight loss

    • Distorted body image

    • Intense fear/anxiety about gaining weight

    • Preoccupation with weight, calories, food, etc.

    • Feelings of guilt after eating

    • Denial of low weight

    • High levels of anxiety and/or depression

  • Chapter 1

    9

    • Low self-esteem

    • Self-injury

    • Withdrawal from friends and activities

    • Excuses for not eating/denial of hunger

    • Food rituals

    • Intense, dramatic mood swings

    • Pale appearance/yellowish skin-tone

    • Thin, dull, and dry hair, skin, and nails

    • Cold intolerance/hypothermia

    • Fatigue/fainting

    • Abuse of laxatives, diet pills, or diuretics

    • Excessive and compulsive exercise

    Subtypes and diagnostic cross-over

    There are two subtypes within the category of AN: the restricting (restrictive)

    type (ANR) and the bingeing-purging type (ANBP). The main difference is that

    the individuals with the latter one experience periods of binge eating

    (consumption of excessive amounts of food coupled with a subjective feeling

    of loss of control over eating) followed by purging behaviors, which are

    means to compensate for the calories consumed. Purging can take forms of

    self- induced vomiting or/and use of laxatives, diuretics or diet pills. Patients

    exhibiting the restricting type do not purge, but maintain low body weight

    solely by reduced food intake and increased energy expenditure via

    exercising and hyperactivity.

    In the clinical reality, the observed phenotypes are often not clear-

    cut. This is exemplified by the fact that EDNOS used to be the most

    frequently assigned diagnosis among EDs (from 40 to 60% of all intakes in ED

    units, 5 and about 75% of cases of EDs detected among adolescent female

    population 6), according to DSM-IV criteria. Furthermore, although the

    category of EDs is relatively stable, moving across the diagnoses within this

    category is quite common 7. Cases with AN often turn into BN or EDNOS,

    whereas cases with BN evolve into EDNOS and, much less frequently, into AN

  • Chapter 1

    10

    8. Moving between AN subtypes is also frequent – over 7 years, nearly 50% of

    women diagnosed with AN crossed over from one subtype into another and

    34% evolved from AN into BN (with a high chance of relapse into AN) 9.

    Agras et al. observed that 80% of patients with EDNOS diagnosis had

    a lifetime history of AN, BN or BED. Additional 10% developed AN, BN or BED

    during a 4-year follow-up. In this study, EDNOS was a way station between a

    fully-blown ED and recovery 10.

    In the most recent, 5-th edition of the DSM the criteria for diagnosis

    of AN became more inclusive. The amenorrhea criterion has been removed

    and criterion A became more general. These changes resulted in a better

    classification of patients with AN and reduction of the vague EDNOS

    category. A study of 309 patients with ED found that almost all of the 60% of

    patients with EDNOS according to DSM-IV were re-assigned to the specific

    diagnoses within the DSM-5 framework 11.

    Empirical classifications

    The outward symptoms ground the division of AN into the ANR and ANBP

    subtypes. There is a growing body of literature, however, indicating that this

    classification has limited usefulness for aethiological research and treatment

    improvement 12. Most of the patients with ANR are going to develop binge-

    purge behavior at some point, suggesting that these subtypes may in fact

    represent alternate stages of the same condition, rather than the subtypes 13. Finally, the differences in treatment utilization, relapse and mortality rates

    are very slight if any 13,14. The observations of the limited usefulness of the

    clinically-derived subphenotypes drove a number of studies which applied

    formal statistical procedures to estabilish the experimental classification of

    eating disorders. Traditional approaches, such as the cluster analysis or the

    taxometric analysis are currently being replaced by the latent class or latent

    profile analyses (LPA) 15. In short, LPA employs a maximum likelihood

    estimation to assign participants to mutually exclusive (unobserved) latent

    classes. Classes are inferred by the pattern of inter-correlations between

    indicators (the variables used to infer the classes, e.g. personality traits). It

  • Chapter 1

    11

    uses general probability model which allows for inequality of variances in

    groups and enables determination of the optimal number of classes via

    formal statistical procedures 15. These analyses were performed in a number

    of studies on eating disorders (see review 16), and although the particular

    results will depend on the selected indicators and parameters’ set-up 17, the

    picture which emerged from these studies is quite consistent 18. In general,

    the empirical approaches to classification result in the division of the

    patients with eating disorders into three distinct classes 16,19:

    1. the over-regulated and over-controlled class, characterized by constraint

    and inhibition,

    2. the under-regulated and disinhibited subtype, with impulsivity and

    dysregulated emotional functioning,

    3. low psychopathology group, characterized by normative levels of

    personality functioning and perfectionism.

    The relevance of these classes has been confirmed in several studies.

    For instance, Wildes et al. 18 have shown that the empirically derived

    classification of patients with AN proves useful clinically. The subtypes

    differed in terms of multiple baseline characteristics, initial response to

    treatment, readmission rates and outcome at discharge (the undercontrolled

    patients had worse outcome than the overcontrolled (OR=3.56, p=.01), who,

    in turn, were worse than the low psychopathology class (OR=11.23, p

  • Chapter 1

    12

    on a strict definition of AN (according to DSM-IV), and they increase

    substantially when some of the criteria are relaxed 25. On the whole,

    although EDs are rare in the general population, they are quite common in

    young females 26. The incidence of AN has been increasing in the past

    century until the 1970s and remained stable henceforth 27.

    AN occurs more often in women (men are affected in only 5% to 10%

    of cases) 28.

    AN is notorious for having the highest standardized mortality ratio

    among psychiatric illnesses (mortality rate being 5 to 10 times higher than in

    a reference population 29,30. Stratification of patients according to body mass

    index (height in cm divided by weight in kg squared; BMI) or age of onset

    shows that SMR is highest in a group of lower BMI and a group of onset later

    than 17 years of age 31. 20% of individuals with AN who died had committed

    suicide 32.

    Risk factors

    Although the list of putative risk factors for AN is long 33, the studies

    examining them are most frequently of a cross-sectional design and hindered

    by the low frequency of AN in the general population. The focus on the

    psychosocial factors, rather than on the biological ones, results from the fact

    that the former are better established in the field and are more easily

    measurable. A last decade has observed a surge in the number of studies of

    genetic risk factors. These, however, will be discussed in the other sections.

    A study with a longitudinal design is preferable when it comes to

    establishing or verifying putative risk factors. In a prospective study on a

    birth cohort, Nicholls et al. (2009) tested 22 childhood risk factors proposed

    in the literature and found that only six were independently associated with

    development of AN at older age. As expected, female sex turned out to be

    the most potent risk factor (OR=22.1), followed by history of undereating

    (OR 2.7), infant feeding problems (OR=2.6), and maternal depressive

    symptoms (OR 1.8). Conversely, higher self-esteem and higher maternal BMI

    were found to be protective (OR=0.3 and 0.91, respectively)34. Other studies

  • Chapter 1

    13

    add childhood sleeping problems, excessive physical exercise, anxious

    parenting and perfectionism to this list 35. Another longitudinal study of risk

    factors in EDs investigated 88 putative factors in a high risk group and found

    that 7 were independently associated with a chance of developing an ED

    (critical comments about eating from teacher/coach/siblings and a history of

    depression had strongest effects on ED risk) 33. Distinguishing a causative risk

    factor from proxies remains a problematic issue in all studies.

    There is some evidence supporting the effect of the season of birth

    on the risk of developing AN, although the effect sizes are small 36. An excess

    of patients with AN was found among those born in spring (March to June;

    OR=1.15) 37. The mechanisms underlying the association are not clear.

    Interestingly, in utero exposure to virus infections (higher incidence of

    chickenpox and rubella infections) was also related to AN risk (OR=1.6 and

    1.5, respectively) 38.

    Some studies suggested that in utero exposure to male or female

    steroids may alter the risk of disordered eating in the future 39, but others

    could not replicate this finding 40.

    Comorbidity

    87% of patients with EDs 31 had some kind of lifetime psychiatric

    comorbidity, such as (in order of frequency) depressive disorders, anxiety

    disorders, suicide attempts, substance abuse disorders, personality disorders

    (predominantly the borderline personality disorder), obsessive compulsive

    disorders and others. Suicide attempts were more frequent among ANBP

    (34%) than in ANR (20%). In about 20% of patients with AN, developmental

    disorders (autistic spectrum disorder, attention deficit-hyperactivity

    disorder) are also observed 41. From the range of somatic disorders, which

    can be comorbid with AN, diabetes mellitus, thyroid disorders and renal

    calculus are seen most often. Psychiatric and somatic comorbidities are

    negatively associated with the outcome of AN 30.

  • Chapter 1

    14

    Treatment and outcome

    AN is a disease of a serious social significance. It often runs a chronic course

    and mainly affects young people 42. Its treatment is expensive 43. Therefore,

    AN generates substantial direct and indirect costs (for example, the cost of

    an inpatient treatment for AN in Germany was estimated to be 4647 EUR per

    case 44).

    Studies of efficacy and effectiveness of treatment for AN offer only a

    moderate or low level of evidence 28. Psychotherapeutical approaches which

    were studied in the context of AN include cognitive-behavioral therapy (CBT),

    interpersonal therapy, dialectical behavior therapy, psychodynamic therapy,

    family therapy, adolescent-focused therapy and several others. CBT is the

    most often recommended modality of treatment (with specific, disease-

    tailored approaches preferred over non-specific approaches), although the

    evidence for superiority of any particular approach is far from being

    conclusive. The main treatment goals include normalization of body weight

    and eating behaviors and alleviation of psychological problems related to EDs 28. Both outpatient and inpatient settings are used. In cases of extreme

    emaciation and resistance to treatment, a forced treatment may be used.

    However, it should be avoided whenever possible.

    There is little evidence for justification of pharmacotherapy use in

    AN. Initially promising findings with regards to Olanzapine 45,46 were 47 or

    were not confirmed in more recent studies 48,49. All these studies were based

    on small samples and their results are not conclusive. Antidepressive

    medications have no effect on the course of AN, but they might be used to

    treat co-morbid depression 28,50. Nonetheless, pharmacological treatment of

    patients with AN is performed by means of antidepressants (tricyclic and

    selective serotonin reuptake inhibitors), antipsychotics (typical and atypical),

    Lithium, naltrexone, antihistamines, clonidine, human growth hormone or

    cannabis 51.

    About 57% of patients whose original diagnosis was AN were fully-

    recovered at a follow-up measurement (the mean duration of the follow-up

    of 4.8 years) in the Netherlands 52. Another conclusion of this study is that

  • Chapter 1

    15

    early detection is associated with a more positive outcome. A study in

    Germany found that at the 12-year follow-up measurement 27.5% of the

    patients initially diagnosed with AN had a good outcome, 25.3% an

    intermediate outcome, 39.6% had a poor outcome, and 7 (7.7%) were

    deceased 53. Factors associated most strongly with an unfavourable outcome

    were sexual problems, impulsivity, long duration of inpatient treatment, and

    long duration of an eating disorder. A review by Steinhausen (2002) adds

    vomiting, bulimia, and purgative abuse, chronicity of illness, and obsessive-

    compulsive personality symptoms to the list of unfavourable prognostic

    features, and notes that other psychiatric disorders at follow-up

    measurements are very common 42.

    One of the reasons why therapy of AN is particularly challenging and

    treatment drop-out rates are high (30-50% 54) is the fact that at least some of

    the AN symptoms are ego-syntonic. This means that they are in harmony

    with patients' goals and desires, hence, it is difficult to illicit motivation for

    treatment.

    Some of the aspects of AN which might be experienced as rewarding

    by patients include:

    • physiological sensations associated with starvation (e.g. stress

    response leading to endogenous opioid secretion)

    • gratification from exerting control over body weight and bodily

    drives (appetite, hunger)

    • positive feedback from the society or societal groups of reference

    (e.g. pro-ana groups)

    • positive feedback from the internalized social mirror (satisfaction of

    own standards)

    • hyperactivity might be rewarding (hypothetically, an evolutionary

    conserved reaction to food scarcity which is supposed to promote

    foraging)

    • excessive exercising might be rewarding in several ways

    • initially rewarding stimuli might lead to adaptation and possible

    withdrawal effects

  • Chapter 1

    16

    Cultural context

    The modern societies no longer struggle with scarcity of food and the times

    of famine fade away in the memory of the Western countries. Food has

    become easily obtainable, both in terms of financial resources and time.

    Being one of the greatest achievements of the Western civilization, this

    availability appears to entail increased rates of obesity and, presumably, EDs.

    There is a stark contrast between ubiquitous food advertisements, which are

    encouraging overindulging (especially in food that is of low nutritional

    quality), and the societal pressure to be thin, exercised implicitly or explicitly

    by our culture. The ambiguous attitude towards food and bodies permeates

    the modern societies. Of note, some believe that the efforts focused on

    combating obesity may unintentionally lead to an increase in incidence of

    EDs. Solid evidence for or against this conviction is lacking. It should be kept

    in mind that in spite of these conflicting societal pressures and common

    dissatisfaction with own body (body dissatisfaction in both men and women

    in western societies is so common that it is considered to be normative 55),

    only a small fraction of individuals develop an eating disorder.

    It is a matter of a debate to what extent culture determines EDs.

    Historical studies and studies on Western and non-Western populations

    report occurrences of AN without body image concerns or fear of gaining

    weight (non-fat-phobic AN) 56,57. This means that the sociocultural pressures

    are neither necessary, nor sufficient for the development of AN 58. Keel &

    Klump (2003) in their systematic review of the historical and epidemiological

    data as well as the data coming from studies on non-Western cultures

    concluded that BN is a more culturally bound condition than AN 59. On the

    other hand, the incidence of AN was much lower in the Netherlands Antilles

    than in the Netherlands, but it was found to be similarly common among

    Netherlands Antilleans living in the Netherlands as among native Dutch 60.

    The Western idealization of thinness (pressure to be thin) appears to be a

    risk factor for the development of AN (possibly in interaction with migration-

    related stress and increased drive to conform in order to counteract

    alienation) and dieting is a possible triggering factor for the onset of an ED.

  • Chapter 1

    17

    Selected candidate molecules for association with AN

    The alterations of biological functioning in patients with AN are quite

    dramatic. The difficulty in investigating those lies in determining the

    difference between premorbid effects (predisposing factors) and effects

    elicited by starvation and hyperactivity (biomarkers). Thorough discussion of

    those alterations is beyond the scope of the present thesis (see for example 61,62), but three molecules which are plausible candidates to play a role in AN

    will be briefly introduced.

    Brain-derived neurotrophic factor (BDNF) is the most ubiquitous

    member of the family of neurotrophins. It plays a role in neurodevelopment 63, neural plasticity, connectivity and synaptogenesis 64. It also has been

    implicated in the regulation of body weight and eating behavior in humans 65

    and animals 66. Genome-wide association studies (GWASs) found the BDNF

    gene locus to be strongly associated with body mass index 67,68. Furthermore,

    mice with reduced expression of BDNF display increased locomotor activity

    and aberrant eating behavior leading to obesity 69,70. A hyperphagic

    phenotype has also been observed in mice with reduced hypothalamic

    expression of the TrkB – high affinity BDNF receptor 71. Similarly,

    hyperphagia, obesity and hyperactivity are present in humans who have a

    functional loss of one copy of the BDNF gene 72. BDNF operates downstream

    of the melanocortin pathway to regulate energy balance 71. Finally, there is

    evidence showing BDNF's involvement in reward and stress functioning 73.

    Catechol-O-methyl transferase (COMT) is an enzyme which degrades

    catecholamines, such as dopamine and noradrenaline 74. It has been

    implicated in the pathogenesis of several mental disorders 75. One allele of a

    functional variant on the COMT gene (rs4680) has been associated with a

    less stable product and, therefore, lower enzymatic activity 76, which in turn

    has been hypothesized to lead to higher dopamine availability 77. Rs4680 was

    studied in mental disorders such as schizophrenia 78, autism 79, depression 80

    and eating disorders 81,82.

    Pro-opiomelanocortin (POMC) is a precursor peptide in the

    melanocortin system (the melanocortin system is involved in body weight

  • Chapter 1

    18

    regulation via effects on appetite and energy expenditure)61. POMC can be

    cleaved into several important peptides, such as α, β, and γ-MSH and β-

    Endorphin. Among many other functions, it plays a role in regulation of

    feeding behaviour 83.

    BDNF, COMT and POMC and their genetic loci are viable candidates

    to study in the context of body-weight related phenotypes, especially AN.

    The next five sections are based on:

    Brandys MK, de Kovel CG, Kas MJ, van Elburg AA, Adan RA. Overview of

    genetic research in anorexia nervosa: The past, the present and the future.

    Int J Eat Disord 2015. 84

    Rationale for gene-association studies

    Several lines of evidence suggest that there is a substantial genetic

    component in the aetiology of AN. AN has been observed across many

    cultures 59. Strong familiar aggregation of AN has been documented (relative

    risk of 11.3 in first-degree relatives of cases with AN, as compared to the

    general population 85,86), and the heritability (h2) has been estimated in

    several twin studies and one adoption study of disordered eating symptoms 87. These estimates range from 0.56 (95% CI, 0.00-0.87) 88 to 0.74 (95% CI:

    0.35-0.95) 89, depending on the studied population, definition of AN and

    applied methodology.

    The evidence coming from several lines of research demonstrates

    that the genetic factors are pivotal in the aetiology of AN. No monogenic

    forms of AN have been found and the data suggest that the genetic

    underpinning of AN is multifactorial (i.e. multiple genetic variants with small

    effects, rather than one or a few potent variants, working in concert with

    environmental factors) 90. Two main types of studies have been employed in

    a search for those genetic factors.

    The linkage approach, which investigates co-segregation of the

    genetic regions with the disease status in large families, has been successful

    in detecting rare and very potent genetic variants involved in the aetiology of

  • Chapter 1

    19

    single-gene disorders (Mendelian), e.g. cystic fibrosis or Huntington’s disease 91,92. However, its usefulness in unravelling common variants of small effects

    in complex, polygenic diseases or traits remains very limited.

    The second category is a population-based genetic-association study,

    which investigates whether frequencies of certain genotypes or alleles are

    different between cases and controls (significant difference implies

    association) or if they are correlated with a quantitative trait. This approach

    focuses on variants with small or medium effects, in a multifactorial model.

    Within this category, candidate-gene studies (CGSs) look into the single-

    nucleotide polymorphisms (SNPs) in biologically plausible genes, whereas

    GWASs test the common SNPs distributed throughout the whole genome.

    Candidate gene approach

    The candidate-gene approach in AN, much like in other psychiatric disorders,

    turned out to be a primarily futile effort. The scarcity of successful

    replications can be explained by several reasons, such as genetic differences

    between the discovery population and the populations in the replication

    attempts, or by the errors and biases leading to false positive results.

    Retrospectively, given the complexity and redundancy of biological

    pathways, and in light of what is now known about the genetic architecture

    of psychiatric diseases, the hypotheses about which genes could potentially

    harbor causative mutations had small chances to be proven right. Out of the

    hundreds of associations indicated by CGSs in the biomedical research only a

    few were replicated in GWASs 93. This ratio is even less favourable in the field

    of psychiatry. One study found a lack of enrichment of the association signal

    in a large genome-wide dataset of cases with schizophrenia and controls

    after the analysis of 732 autosomal genes indicated in 1374 CGSs

    (investigation of signal enrichment involves collective testing of a selected

    group of variants in an independent dataset; it has much greater power,

    compared to testing of individual variants) 94.

  • Chapter 1

    20

    Candidate gene studies in anorexia nervosa

    Comprehensive reviews of CGSs in AN are available elsewhere 95,96. Although

    the selection of candidate genes for studies of AN was based on interesting

    hypotheses 97, and more than 200 gene-association studies were performed

    in the context of EDs, up to date none of the initially promising findings have

    been convincingly replicated in the subsequent candidate or genome-wide

    studies. Meta-analyses, which summarized and weighted the evidence from

    multiple studies, were also disillusioning 98-101. Also the relatively recent CGS

    which used the modern standards of design, quality control and statistical

    significance was negative 102. Still, there are a few findings which await

    replication attempts, such as rs1473473 of TPH2 103, the 5-HTTLPR

    polymorphism on SLC6A4 104, rs7180942 in NTRK3 105 and Ala67Thr variant in

    AGRP 106 (these polymorphisms were not tested in two recent GWASs of AN,

    because they were not present on the genotyping arrays used in those

    studies).

    In parallel to the growing disillusionment about the candidate-gene

    method, a new approach towards investigation of genetic associations

    emerged. GWAS technology is relatively recent (first GWAS dates back to

    2005 107), but it already has had significant impact on the landscape of

    biomedical research and resulted in progression of the aetiological

    knowledge about diseases and traits 108.

    Genome-wide association approach

    GWAS is a hypothesis-free approach. It uses microarray platforms to

    examine the genotypic data from a large number of SNPs (from hundreds of

    thousands up to millions), which cover most of the human common SNP

    variation (a SNP is considered common if the frequency of its minor allele is

    larger than 1%). This coverage is increased via imputation - a procedure

    which uses statistical algorithms to infer the genotypes of the ungenotyped

    SNPs by employing the reference data coming from e.g. HapMap or 1000

    Genomes Project populations. Genome-wide data also allows for

  • Chapter 1

    21

    investigation of copy number variants (CNVs; deleted or duplicated stretches

    of the genome).

    Below is a list of the main goals of GWASs:

    • Furthering the understanding of the biological mechanisms of the

    disease, by finding the genes and pathways involved in the aetiology.

    This is the foremost goal of GWASs.

    • Learning about the genetic architecture. This includes the expected

    range of effect sizes, allelic frequencies of the associated variants,

    underlying genetic models (additive, dominant, recessive,

    overdominant, multiplicative) and the possibility of gene x

    environment and gene x gene interactions.

    • Understanding of the genetic overlap between diseases and traits.

    This has a potential of enhancing the nosological system and

    treatment.

    • Genetic screening to identify populations at risk (risk prediction) or

    individual genotyping of a patient to inform diagnosis and treatment

    (personalized medicine). As exciting as these prospects are, they are

    distant goals, and in view of a highly polygenic nature of psychiatric

    diseases, they are unlikely to be achievable in the near future 109.

    What needs to be remembered when interpreting a GWAS is that its

    results inform about association but do not determine causality, and that a

    statistical strength of association at a given locus should not be confused

    with its biological relevance (the most significant finding in GWASs might not

    be the most informative).

    Scope and outline of the thesis

    The overarching theme of this thesis is the effort to shed light on the genetic

    background of AN. That undertaking predominantly involves searching for

  • Chapter 1

    22

    the genetic associations via the candidate-gene and genome-wide studies.

    Thus, we aimed to find the genetic variants which change the risk of

    developing AN, and to increase the understanding of the mechanisms of the

    disease. Beyond investigating the genetic associations, in one chapter we

    also take interest in studies examining the serum levels of the BDNF

    neurotrophin in patients with AN. BDNF is a product of the BDNF gene which

    was also investigated in this thesis.

    We begin by studying a possible genetic relation of AN and obesity

    by testing several genetic variants associated with the latter in a sample of

    patients with AN110. This work adds to the discussion about the genetic

    nature of AN and its hypothesized relation to the opposite extreme of the

    weight spectrum - obesity (chapter 2). The next chapter (3) presents the only

    study which does not directly involve patients with AN. In that publication

    we describe the search for association of variants from the POMC locus with

    detailed measures of body composition and nutrient choice in the general

    population111. POMC molecule, due to its effects on the appetite regulation,

    was a plausible candidate for having a role in AN diathesis.

    We also explore the alterations of biological functioning in AN, in the

    context of possible candidates for genetic associations. Chapter 4 is a meta-

    analysis of several studies which compared the serum BDNF levels in patients

    with AN and healthy controls112. A discussion of whether the observed

    effects are state or trait dependent is included. Thereafter, in chapter 5, we

    apply the meta-analytical methodology to a promising candidate for genetic

    association with AN, the Val66Met polymorphism on the BDNF gene101.

    Meta-analyses combine evidence from multiple studies on a single subject,

    which (together with our own, novel data) allows us to draw stronger

    conclusions than any of these studies alone.

    The further investigations (chapter 6) consider a genetic

    polymorphism long thought to play a role in several mental disorders,

    including ED. Val158Met polymorphism of the COMT gene was tested in the

  • Chapter 1

    23

    new data and the results were merged in a meta-analytical framework with

    the results of previous studies on this subject100.

    Chapter 7 presents the work which used the genome-wide data of

    patients with AN and controls to test for association of AN with selected

    structural variants (rather than SNPs). The nature of the available data was

    not sufficient to render the results fully credible; hence this study remains a

    preliminary investigation.

    The appendix describes a large collaborative study which uses

    thousands of DNA samples from all over the world and analyses them in a

    hypothesis-free, genome-wide approach113. This remains the largest genetic

    study in AN up to date.

    The final publication included in this dissertation (chapter 8) is an

    opinion paper which reviews and discusses the past approaches to the

    studies of gene-association in AN, shows how they evolved over the years (a

    process well reflected in the present thesis), and tries to outline the

    directions for future research.

    References

    1. Gull WW. Anorexia nervosa (apepsia hysterica, anorexia hysterica). 1868.

    Obes Res 1997;5:498-502.

    2. Pearce JMS. Richard morton: Origins of anorexia nervosa. Eur Neurol

    2004;52:191-192.

    3. Sullivan P. Mortality in anorexia nervosa. Am J Psychiatry 1995;152:1073-

    1074.

    4. Keel PK, Dorer DJ, Eddy KT, Franko D, Charatan DL, Herzog DB. Predictors

    of mortality in eating disorders. Arch Gen Psychiatry 2003;60:179-183.

    5. Thomas JJ, Vartanian LR, Brownell KD. The relationship between eating

    disorder not otherwise specified (EDNOS) and officially recognized eating

    disorders: Meta-analysis and implications for DSM. Psychol Bull

    2009;135:407-433.

    6. Machado PPP, Machado BC, Gonçalves S, Hoek HW. The prevalence of

    eating disorders not otherwise specified. Int J Eat Disord 2007;40:212-217.

  • Chapter 1

    24

    7. Milos G, Spindler A, Schnyder U, Fairburn CG. Instability of eating disorder

    diagnoses: Prospective study. The British Journal of Psychiatry 2005;187:573-

    578.

    8. Tozzi F, Thornton LM, Klump KL, Fichter MM, Halmi KA, Kaplan AS, et al.

    Symptom fluctuation in eating disorders: Correlates of diagnostic crossover.

    Am J Psychiatry 2005;162:732-740.

    9. Eddy KT, Dorer DJ, Franko DL, Tahilani K, Thompson-Brenner H, Herzog DB.

    Diagnostic crossover in anorexia nervosa and bulimia nervosa: Implications

    for DSM-V. Am J Psychiatry 2008;165:245-250.

    10. Agras WS, Crow S, Mitchell JE, Halmi KA, Bryson S. A 4-year prospective

    study of eating disorder NOS compared with full eating disorder syndromes.

    Int J Eat Disord 2009;42:565-570.

    11. Fisher MM, Gonzalez M, Malizio J. Eating disorders in adolescents: How

    does the DSM-5 change the diagnosis? Journal of Adolescent Health

    2014;54:S9.

    12. Peat C, Mitchell JE, Hoek H, Wonderlich S. Validity and utility of subtyping

    anorexia nervosa. Int J Eat Disord 2009;9999:NA.

    13. Eddy KT, Keel PK, Dorer DJ, Delinsky SS, Franko DL, Herzog DB.

    Longitudinal comparison of anorexia nervosa subtypes. Int J Eat Disord

    2002;31:191-201.

    14. Keel PK, Dorer DJ, Eddy KT, Delinsky SS, Franko DL, Blais MA, et al.

    Predictors of treatment utilization among women with anorexia and bulimia

    nervosa. Am J Psychiatry 2002;159:140-142.

    15. Magidson J, Vermunt JK. Latent class models for clustering: A comparison

    with K-means. Canadian Journal of Marketing Research 2002;20:36-36-43.

    16. Wonderlich SA, Joiner TE, Keel PK, Williamson DA, Crosby RD. Eating

    disorder diagnoses: Empirical approaches to classification. Am Psychol

    2007;62:167-180.

    17. Swanson SA, Lindenberg K, Bauer S, Crosby RD. A monte carlo

    investigation of factors influencing latent class analysis: An application to

    eating disorder research. Int J Eat Disord 2011:n/a-n/a.

  • Chapter 1

    25

    18. Wildes JE, Marcus MD, Crosby RD, Ringham RM, Dapelo MM, Gaskill JA,

    et al. The clinical utility of personality subtypes in patients with anorexia

    nervosa. J Consult Clin Psychol 2011;79:665-674.

    19. Steiger H, Richardson J, Schmitz N, Israel M, Bruce KR, Gauvin L. Trait-

    defined eating-disorder subtypes and history of childhood abuse. Int J Eat

    Disord 2010;43:428-432.

    20. Miller MW, Kaloupek DG, Dillon AL, Keane TM. Externalizing and

    internalizing subtypes of combat-related PTSD: A replication and extension

    using the PSY-5 scales. J Abnorm Psychol 2004;113:636-645.

    21. Hudson JI, Hiripi E, Pope Jr. HG, Kessler RC. The prevalence and correlates

    of eating disorders in the national comorbidity survey replication. Biological

    Psychiatry 2007;61:348-358.

    22. Keski-Rahkonen A, Hoek HW, Susser ES, Linna MS, Sihvola E, Raevuori A,

    et al. Epidemiology and course of anorexia nervosa in the community. Am J

    Psychiatry 2007;164:1259-1265.

    23. Lucas A, Beard C, O'Fallon W, Kurland L. 50-year trends in the incidence

    of anorexia nervosa in rochester, minn.: A population-based study. Am J

    Psychiatry 1991;148:917-922.

    24. Hoek HW, van Hoeken D. Review of the prevalence and incidence of

    eating disorders. International Journal of Eating Disorders 2003;34:383-396.

    25. Isomaa R, Isomaa A, Marttunen M, Kaltiala-Heino R, Björkqvist K. The

    prevalence, incidence and development of eating disorders in finnish

    adolescents - a two-step 3-year follow-up study. European Eating Disorders

    Review 2009;17:199-207.

    26. Smink FR, van Hoeken D, Hoek HW. Epidemiology of eating disorders:

    Incidence, prevalence and mortality rates. Curr Psychiatry Rep 2012;14:406-

    414.

    27. Currin L, Schmidt U, Treasure J, Jick H. Time trends in eating disorder

    incidence. The British Journal of Psychiatry 2005;186:132-135.

    28. Herpertz S, Hagenah U, Vocks S, von Wietersheim J, Cuntz U, Zeeck A.

    The diagnosis and treatment of eating disorders. Dtsch Arztebl Int

    2011;108:678-685.

  • Chapter 1

    26

    29. Birmingham CL, Su J, Hlynsky JA, Goldner EM, Gao M. The mortality rate

    from anorexia nervosa. Int J Eat Disord 2005;38:143-146.

    30. Papadopoulos FC, Ekbom A, Brandt L, Ekselius L. Excess mortality, causes

    of death and prognostic factors in anorexia nervosa. The British Journal of

    Psychiatry 2009;194:10-17.

    31. Rosling AM, Sparén P, Norring C, von Knorring A. Mortality of eating

    disorders: A follow-up study of treatment in a specialist unit 1974?2000. Int J

    Eat Disord 2011;44:304-310.

    32. Arcelus J, Mitchell AJ, Wales J, Nielsen S. Mortality rates in patients with

    anorexia nervosa and other eating disorders: A meta-analysis of 36 studies.

    Arch Gen Psychiatry 2011;68:724-731.

    33. Jacobi C, Fittig E, Bryson SW, Wilfley D, Kraemer HC, Taylor CB. Who is

    really at risk? identifying risk factors for subthreshold and full syndrome

    eating disorders in a high-risk sample. Psychol Med 2011;41:1939.

    34. Nicholls DE, Viner RM. Childhood risk factors for lifetime anorexia

    nervosa by age 30 years in a national birth cohort. J Am Acad Child Adolesc

    Psychiatry 2009;48:791-799.

    35. Pike KM, Hilbert A, Wilfley DE, Fairburn CG, Dohm FA, Walsh BT, et al.

    Toward an understanding of risk factors for anorexia nervosa: A case-control

    study. Psychol Med 2008;38:1443-1453.

    36. Winje E, Willoughby K, Lask B. Season of birth bias in eating disorders--

    fact or fiction? Int J Eat Disord 2008;41:479-490.

    37. Disanto G, Handel AE, Para AE, Ramagopalan SV, Handunnetthi L. Season

    of birth and anorexia nervosa. The British Journal of Psychiatry 2011.

    38. Favaro A, Tenconi E, Ceschin L, Zanetti T, Bosello R, Santonastaso P. In

    utero exposure to virus infections and the risk of developing anorexia

    nervosa. Psychol Med 2011:1-7.

    39. Culbert KM, Breedlove SM, Burt SA, Klump KL. Prenatal hormone

    exposure and risk for eating disorders: A comparison of opposite-sex and

    same-sex twins. Arch Gen Psychiatry 2008;65:329-336.

  • Chapter 1

    27

    40. Baker JH, Lichtenstein P, Kendler KS. Intrauterine testosterone exposure

    and risk for disordered eating. The British Journal of Psychiatry

    2009;194:375-376.

    41. Wentz E, Lacey JH, Waller G, Rastam M, Turk J, Gillberg C. Childhood

    onset neuropsychiatric disorders in adult eating disorder patients. A pilot

    study. Eur Child Adolesc Psychiatry 2005;14:431-437.

    42. Steinhausen H. The outcome of anorexia nervosa in the 20th century. Am

    J Psychiatry 2002;159:1284-1293.

    43. Striegel-Moore RH, Leslie D, Petrill SA, Garvin V, Rosenheck RA. One-year

    use and cost of inpatient and outpatient services among female and male

    patients with an eating disorder: Evidence from a national database of health

    insurance claims. Int J Eat Disord 2000;27:381-389.

    44. Haas L, Stargardt T, Schreyoegg J, Schlösser R, Danzer G, Klapp BF.

    Inpatient costs and predictors of costs in the psychosomatic treatment of

    anorexia nervosa. Int J Eat Disord 2011:n/a-n/a.

    45. Brambilla F, Garcia CS, Fassino S, Daga GA, Favaro A, Santonastaso P, et

    al. Olanzapine therapy in anorexia nervosa: Psychobiological effects. Int Clin

    Psychopharmacol 2007;22:197-204.

    46. Leggero C, Masi G, Brunori E, Calderoni S, Carissimo R, Maestro S, et al.

    Low-dose olanzapine monotherapy in girls with anorexia nervosa, restricting

    subtype: Focus on hyperactivity. J Child Adolesc Psychopharmacol

    2010;20:127-133.

    47. Attia E, Kaplan AS, Walsh BT, Gershkovich M, Yilmaz Z, Musante D, et al.

    Olanzapine versus placebo for out-patients with anorexia nervosa. Psychol

    Med 2011:1-6.

    48. Kafantaris V, Leigh E, Hertz S, Berest A, Schebendach J, Sterling WM, et al.

    A placebo-controlled pilot study of adjunctive olanzapine for adolescents

    with anorexia nervosa. J Child Adolesc Psychopharmacol 2011;21:207-212.

    49. Norris ML, Spettigue W, Buchholz A, Henderson KA, Gomez R, Maras D,

    et al. Olanzapine use for the adjunctive treatment of adolescents with

    anorexia nervosa. J Child Adolesc Psychopharmacol 2011.

  • Chapter 1

    28

    50. Suarez-Pinilla P, Pena-Perez C, Arbaizar-Barrenechea B, Crespo-Facorro B,

    Del Barrio JA, Treasure J, et al. Inpatient treatment for anorexia nervosa: A

    systematic review of randomized controlled trials. J Psychiatr Pract

    2015;21:49-59.

    51. Aigner M, Treasure J, Kaye W, Kasper S. World federation of societies of

    biological psychiatry (WFSBP) guidelines for the pharmacological treatment

    of eating disorders. World Journal of Biological Psychiatry 2011;12:400-443.

    52. van Son GE, van Hoeken D, van Furth EF, Donker GA, Hoek HW. Course

    and outcome of eating disorders in a primary care-based cohort. Int J Eat

    Disord 2009;9999:NA.

    53. Fichter MM, Quadflieg N, Hedlund S. Twelve-year course and outcome

    predictors of anorexia nervosa. Int J Eat Disord 2006;39:87-100.

    54. Guarda AS. Treatment of anorexia nervosa: Insights and obstacles.

    Physiology & Behavior, 2008;94:113-120.

    55. Tantleff-Dunn S, Barnes RD, Larose JG. It's not just a "woman thing:" The

    current state of normative discontent. Eat Disord 2011;19:392-402.

    56. Santonastaso P, Bosello R, Schiavone P, Tenconi E, Degortes D, Favaro A.

    Typical and atypical restrictive anorexia nervosa: Weight history, body image,

    psychiatric symptoms, and response to outpatient treatment. Int J Eat Disord

    2009;42:464-470.

    57. Becker AE, Thomas JJ, Pike KM. Should non-fat-phobic anorexia nervosa

    be included in DSM-V? Int J Eat Disord 2009;42:620-635.

    58. Schmidt U. Epidemiology and aetiology of eating disorders. Psychiatry,

    2005;4:5-9.

    59. Keel PK, Klump KL. Are eating disorders culture-bound syndromes?

    implications for conceptualizing their etiology. Psychol Bull 2003;129:747-

    769.

    60. van Hoeken D, Veling W, Smink FRE, Hoek HW. The incidence of anorexia

    nervosa in netherlands antilles immigrants in the netherlands. Eur Eat Disord

    Rev 2010;18:399-403.

    61. Miller K. Endocrine dysregulation in anorexia nervosa update. The Journal

    of Clinical Endocrinology & Metabolism 2011;96:2939-2949.

  • Chapter 1

    29

    62. Adan RAH, Hillebrand JJG, Danner UN, Cano SC, Kas MJH, Verhagen LAW.

    Neurobiology driving hyperactivity in activity-based anorexia. Curr Top Behav

    Neurosci 2011;6:229-250.

    63. Hyman C, Hofer M, Barde Y, Juhasz M, Yancopoulos GD, Squinto SP, et al.

    BDNF is a neurotrophic factor for dopaminergic neurons of the substantia

    nigra. Nature 1991;350:230-232.

    64. Greenberg ME, Xu B, Lu B, Hempstead BL. New insights in the biology of

    BDNF synthesis and release: Implications in CNS function. J Neurosci

    2009;29:12764-12767.

    65. Lommatzsch M, Zingler D, Schuhbaeck K, Schloetcke K, Zingler C, Schuff-

    Werner P, et al. The impact of age, weight and gender on BDNF levels in

    human platelets and plasma. Neurobiol Aging 2005;26:115-123.

    66. King BM. The rise, fall, and resurrection of the ventromedial

    hypothalamus in the regulation of feeding behavior and body weight. Physiol

    Behav 2006;87:221-244.

    67. Willer CJ, Speliotes EK, Loos RJ, Li S, Lindgren CM, Heid IM, et al. Six new

    loci associated with body mass index highlight a neuronal influence on body

    weight regulation. Nat Genet 2009;41:25-34.

    68. Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR, et al. Genetic

    studies of body mass index yield new insights for obesity biology. Nature

    2015;518:197-206.

    69. Kernie SG, Liebl DJ, Parada LF. BDNF regulates eating behavior and

    locomotor activity in mice. EMBO J 2000;19:1290-1300.

    70. Rios M, Fan G, Fekete C, Kelly J, Bates B, Kuehn R, et al. Conditional

    deletion of brain-derived neurotrophic factor in the postnatal brain leads to

    obesity and hyperactivity. Mol Endocrinol 2001;15:1748-1757.

    71. Xu B, Goulding EH, Zang K, Cepoi D, Cone RD, Jones KR, et al. Brain-

    derived neurotrophic factor regulates energy balance downstream of

    melanocortin-4 receptor. Nat Neurosci 2003;6:736-742.

    72. Gray J, Yeo GSH, Cox JJ, Morton J, Adlam AR, Keogh JM, et al.

    Hyperphagia, severe obesity, impaired cognitive function, and hyperactivity

  • Chapter 1

    30

    associated with functional loss of one copy of the brain-derived neurotrophic

    factor (BDNF) gene. Diabetes 2006;55:3366-3371.

    73. Narita M, Aoki K, Takagi M, Yajima Y, Suzuki T. Implication of brain-

    derived neurotrophic factor in the release of dopamine and dopamine-

    related behaviors induced by methamphetamine. Neuroscience

    2003;119:767-775.

    74. Chen J, Lipska BK, Halim N, Ma QD, Matsumoto M, Melhem S, et al.

    Functional analysis of genetic variation in catechol-O-methyltransferase

    (COMT): Effects on mRNA, protein, and enzyme activity in postmortem

    human brain. Am J Hum Genet 2004;75:807-821.

    75. Mier D, Kirsch P, Meyer-Lindenberg A. Neural substrates of pleiotropic

    action of genetic variation in COMT: A meta-analysis. Mol Psychiatry

    2010;15:918-927.

    76. Lachman HM, Papolos DF, Saito T, Yu YM, Szumlanski CL, Weinshilboum

    RM. Human catechol-O-methyltransferase pharmacogenetics: Description of

    a functional polymorphism and its potential application to neuropsychiatric

    disorders. Pharmacogenetics 1996;6:243-250.

    77. Shield AJ, Thomae BA, Eckloff BW, Wieben ED, Weinshilboum RM.

    Human catechol O-methyltransferase genetic variation: Gene resequencing

    and functional characterization of variant allozymes. Mol Psychiatry

    2004;9:151-160.

    78. Costas J, Sanjuán J, Ramos-Ríos R, Paz E, Agra S, Ivorra JL, et al.

    Heterozygosity at catechol-O-methyltransferase Val158Met and

    schizophrenia: New data and meta-analysis. J Psychiatr Res 2011;45:7-14.

    79. Stergiakouli E, Thapar A. Fitting the pieces together: Current research on

    the genetic basis of attention-deficit/hyperactivity disorder (ADHD).

    Neuropsychiatr Dis Treat 2010;6:551-560.

    80. Åberg E, Fandiño-Losada A, Sjöholm LK, Forsell Y, Lavebratt C. The

    functional Val158Met polymorphism in catechol-O-methyltransferase

    (COMT) is associated with depression and motivation in men from a swedish

    population-based study. J Affect Disord 2011;129:158-166.

  • Chapter 1

    31

    81. Mikolajczyk E, Smiarowska M, Grzywacz A, Samochowiec J. Association of

    eating disorders with catechol- O -methyltransferase gene functional

    polymorphism. Neuropsychobiology 2006;54:82-86.

    82. Frieling H, Romer KD, Wilhelm J, Hillemacher T, Kornhuber J, de Zwaan

    M, et al. Association of catecholamine-O-methyltransferase and 5-HTTLPR

    genotype with eating disorder-related behavior and attitudes in females with

    eating disorders. Psychiatr Genet 2006;16:205-208.

    83. Millington G. The role of proopiomelanocortin (POMC) neurones in

    feeding behaviour. Nutr Metab (Lond) 2007;4:18.

    84. Brandys MK, de Kovel CG, Kas MJ, van Elburg AA, Adan RA. Overview of

    genetic research in anorexia nervosa: The past, the present and the future.

    Int J Eat Disord 2015.

    85. Lilenfeld LR, Kaye WH, Greeno CG, Merikangas KR, Plotnicov K, Pollice C,

    et al. A controlled family study of anorexia nervosa and bulimia nervosa:

    Psychiatric disorders in first-degree relatives and effects of proband

    comorbidity. Arch Gen Psychiatry 1998;55:603-610.

    86. Strober M, Freeman R, Lampert C, Diamond J, Kaye W. Controlled family

    study of anorexia nervosa and bulimia nervosa: Evidence of shared liability

    and transmission of partial syndromes. Am J Psychiatry 2000;157:393-401.

    87. Klump KL, Suisman JL, Burt SA, McGue M, Iacono WG. Genetic and

    environmental influences on disordered eating: An adoption study. J Abnorm

    Psychol 2009;118:797.

    88. Bulik CM, Sullivan PF, Tozzi F, Furberg H, Lichtenstein P, Pedersen NL.

    Prevalence, heritability, and prospective risk factors for anorexia nervosa.

    Arch Gen Psychiatry 2006;63:305-312.

    89. Klump KL, Miller K, Keel P, McGue M, Iacono W. Genetic and

    environmental influences on anorexia nervosa syndromes in a

    population–based twin sample. Psychol Med 2001;31:737-740.

    90. Steinhausen H, Jakobsen H, Helenius D, Munk-Jorgensen P, Strober M. A

    nationwide study of the family aggregation and risk factors in anorexia

    nervosa over three generations. Int J Eat Disord 2014;48:1-8.

  • Chapter 1

    32

    91. Knowlton RG, Cohen-Haguenauer O, Van Cong N, Frezal J, Brown VA,

    Barker D, et al. A polymorphic DNA marker linked to cystic fibrosis is located

    on chromosome 7. Nature 1985;318:380-382.

    92. Gusella JF, Wexler NS, Conneally PM, Naylor SL, Anderson MA, Tanzi RE,

    et al. A polymorphic DNA marker genetically linked to huntington's disease.

    Nature 1983;306:234-238.

    93. Siontis KC, Patsopoulos NA, Ioannidis JP. Replication of past candidate

    loci for common diseases and phenotypes in 100 genome-wide association

    studies. European Journal of Human Genetics 2010;18:832-837.

    94. Collins AL, Kim Y, Sklar P, International Schizophrenia Consortium,

    O'Donovan MC, Sullivan PF. Hypothesis-driven candidate genes for

    schizophrenia compared to genome-wide association results. Psychol Med

    2012;42:607-616.

    95. Clarke TK, Weiss ARD, Berrettini WH. The genetics of anorexia nervosa.

    Clin Pharmacol Ther 2011;91:181-188.

    96. Rask-Andersen M, Olszewski PK, Levine AS, Schioth HB. Molecular

    mechanisms underlying anorexia nervosa: Focus on human gene association

    studies and systems controlling food intake. Brain Res Rev 2010;62:147-164.

    97. Hinney A, Volckmar A. Genetics of eating disorders. Curr Psychiatry Rep

    2013;15:1-9.

    98. Slof-Op't Landt MC, Furth EF, Meulenbelt I, Bartels M, Hottenga JJ,

    Slagboom PE, et al. Association study of the estrogen receptor I gene (ESR1)

    in anorexia nervosa and eating disorders: No replication found. Int J Eat

    Disord 2013;47:211-214.

    99. Zhang C, Chen J, Jia X, Yu S, Jiang W, Zhang R, et al. Estrogen receptor 1

    gene rs2295193 polymorphism and anorexia nervosa: New data and

    meta†analysis. Asia⠀ Pacific Psychiatry 2013;5:331-335.

    100. Brandys MK, Slof-Op't Landt MC, van Elburg AA, Ophoff R, Verduijn W,

    Meulenbelt I, et al. Anorexia nervosa and the Val158Met polymorphism of

    the COMT gene: Meta-analysis and new data. Psychiatr Genet 2012;22:130-

    136.

  • Chapter 1

    33

    101. Brandys MK, Kas MJ, van Elburg AA, Ophoff R, Slof-Op't Landt MC,

    Middeldorp CM, et al. The Val66Met polymorphism of the BDNF gene in

    anorexia nervosa: New data and a meta-analysis. The World Journal of

    Biological Psychiatry 2013;14:441-451.

    102. Pinheiro AP, Bulik CM, Thornton LM, Sullivan PF, Root TL, Bloss CS, et al.

    Association study of 182 candidate genes in anorexia nervosa. Am J Med

    Genet B Neuropsychiatr Genet 2010;153B:1070-1080.

    103. Slof-O't Landt M, Meulenbelt I, Bartels M, Suchiman E, Middeldorp C,

    Houwing†Duistermaat J, et al. Association study in eating disorders:

    TPH2 associates with anorexia nervosa and self-â induced vomiting. Genes,

    Brain and Behavior 2011;10:236-243.

    104. Calati R, De Ronchi D, Bellini M, Serretti A. The 5-HTTLPR polymorphism

    and eating disorders: A meta- analysis. Int J Eat Disord 2011;44:191-199.

    105. Mercader JM, Saus E, Aguera Z, Bayes M, Boni C, Carreras A, et al.

    Association of NTRK3 and its interaction with NGF suggest an altered cross-

    regulation of the neurotrophin signaling pathway in eating disorders. Hum

    Mol Genet 2008;17:1234-1244.

    106. Vink T, Hinney A, Van Elburg A, van Goozen SH, Sandkuijl L, Sinke R, et

    al. Association between an agouti-related protein gene polymorphism and

    anorexia nervosa. Mol Psychiatry 2001;6:325-328.

    107. Klein RJ, Zeiss C, Chew EY, Tsai JY, Sackler RS, Haynes C, et al.

    Complement factor H polymorphism in age-related macular degeneration.

    Science 2005;308:385-389.

    108. Kim Y, Zerwas S, Trace SE, Sullivan PF. Schizophrenia genetics: Where

    next? Schizophr Bull 2011;37:456-463.

    109. Wray NR, Yang J, Hayes BJ, Price AL, Goddard ME, Visscher PM. Pitfalls

    of predicting complex traits from SNPs. Nature Reviews Genetics

    2013;14:507-515.

    110. Brandys MK, van Elburg AA, Loos RJF, Bauer F, Hendriks J, van der

    Schouw YT, et al. Are recently identified genetic variants regulating BMI in

    the general population associated with anorexia nervosa? American Journal

    of Medical Genetics Part B: Neuropsychiatric Genetics 2010;153B:695-699.

  • Chapter 1

    34

    111. Ternouth A, Brandys MK, van der Schouw YT, Hendriks J, Jansson J,

    Collier D, et al. Association study of POMC variants with body composition

    measures and nutrient choice. Eur J Pharmacol 2011.

    112. Brandys MK, Kas MJH, van Elburg AA, Campbell IC, Adan RAH. A meta-

    analysis of circulating BDNF concentrations in anorexia nervosa. World J Biol

    Psychiatry 2011.

    113. Boraska V, Franklin C, Floyd J, Thornton L, Huckins L, Southam L, et al. A

    genome-wide association study of anorexia nervosa. Mol Psychiatry

    2014;19:1085-1094.

  • Chapter 2

    35

    Chapter 2

    Are recently identified genetic variants regulating BMI in the

    general population associated with anorexia nervosa?

    Marek K. Brandys

    Annemarie A. van Elburg

    Ruth J.F. Loos

    Florianne Bauer

    Judith Hendriks

    Yvonne T. van der Schouw

    Roger A.H. Adan

    American Journal of Medical Genetics Part B: Neuropsychiatric Genetics

    2010; 153B(2): 695-699.

  • Chapter 2

    36

    Abstract

    The influence of body mass index (BMI) on susceptibility to anorexia nervosa

    (AN) is not clear. Recently published genome-wide association (GWA) studies

    of the general population identified several variants influencing BMI. We

    genotyped these variants in an AN sample to test for association and to

    investigate a combined effect of BMI-increasing alleles (as determined in the

    original GWA studies) on the risk of developing the disease. Individual single

    nucleotide polymorphisms (SNPs) were tested for association with AN in a

    sample of 267 AN patients and 1636 population controls. A logistic

    regression for the combined effect of BMI-increasing alleles included 225

    cases and 1351 controls. We found no significant association between

    individual SNPs and AN. The analysis of a combined effect of BMI-increasing

    alleles showed absence of association with the investigated condition. The

    percentages of BMI-increasing alleles were equal between cases and

    controls. This study found no evidence that genetic variants regulating BMI in

    the general population are significantly associated with susceptibility to AN.

  • Chapter 2

    37

    Introduction

    Two extensive population-based genome-wide association studies (GWA) of

    BMI and obesity have been published recently 1,2. Both studies revealed

    associations of new loci and confirmed already known roles of FTO and

    MC4R 3,4. All together, these loci harbor 10 genes, most of them

    predominantly expressed in the central nervous system (Table I).

    Although little is known about the function of the genes linked to the

    newly identified genetic variants for body mass index (BMI) and common

    obesity, preliminary evidence suggests that they might affect BMI via

    involvement in the neuronal regulation of food intake 1,2. Additionally, it has

    been proposed that eating disorders and obesity may be considered on the

    same continuum of psychopathology (as opposed to discrete models; 5). We,

    therefore, hypothesized that variants affecting BMI and obesity may

    potentially alter the risk of developing an eating disorder such as anorexia

    nervosa (AN).

    It is under debate whether high or low BMI before the onset of the

    disease has an impact upon susceptibility to AN 6,7. AN patients with lower

    premorbid BMI (self-reported) tend to present with lower BMI at first

    referral for the treatment 8. A recent study found a correlation between

    premorbid BMI and BMI at discharge from the treatment and at follow-up in

    AN 6,8. Patients with lower BMI before the onset of the disease and at

    admission had poorer general indices of functioning 6. Elevated BMI could

    either protect against the disease – since AN is a disorder of low body weight

    – or increase the risk, via a tendency to a general eating pathology such as

    e.g. restrained eating, excessive dieting or a persistent desire to lose weight.

    In the present study we aimed to test whether the genetic variants

    increasing BMI in the general population escalate the susceptibility to AN or

    diminish it (or have no effect upon it).

  • Chapte

    r 2

    38

    TA

    BL

    E I

    . C

    ha

    ract

    eris

    tics

    of

    inv

    esti

    ga

    ted

    SN

    Ps

    an

    d a

    na

    lysi

    s o

    f a

    sso

    cia

    tio

    n w

    ith

    AN

    (a

    llel

    ic t

    est,

    1d

    f)

    SN

    P

    Nea

    rby

    gene

    BM

    I

    incr.

    alle

    le

    Eff

    ect

    all

    ele

    freq

    . re

    port

    ed

    in r

    efe

    rred

    GW

    AS

    1,2

    Eff

    ect

    alle

    le

    freq

    . in

    case

    s

    Eff

    ect

    all

    ele

    freq

    . in

    contr

    ols

    HW

    E i

    n

    cont.

    Eff

    ect

    size

    s det

    ecta

    ble

    at

    80%

    pow

    er

    OR

    fo

    r

    the

    asso

    c.

    (95%

    CI)

    P-v

    alu

    e, a

    lleli

    c te

    st

    OR

    fo

    r hete

    ro-

    zygo

    tes

    OR

    fo

    r ho

    mo

    -

    zygo

    tes

    rs1121980

    *

    FT

    O

    A

    41%

    41.3

    %

    42.1

    %

    .10

    1.3

    1.7

    .9

    6

    (.80-1

    .16)

    .73

    rs17700633

    M

    C4R

    A

    32%

    29.5

    %

    30.1

    %

    .99

    1.3

    1.7

    .9

    7

    (.79-1

    .18)

    .78

    rs17782313

    M

    C4R

    C

    21%

    26.0

    %

    26.1

    %

    1.0

    0

    1.4

    1.7

    .9

    9

    (.80-1

    .22)

    .96

    rs6548238

    T

    ME

    M1

    8

    C

    84%

    83.3

    %

    83.7

    %

    .86

    1.5

    2.3

    1.0

    3

    (.80-1

    .32)

    .81

    rs10938397

    G

    NP

    DA

    2

    G

    45%

    42.2

    %

    42.0

    %

    .98

    1.3

    1.7

    1.0

    0

    (.83-1

    .21)

    .94

    rs7498665

    S

    H2

    B1

    G

    41%

    38.5

    %

    37.8

    %

    .86

    1.3

    1.7

    1.0

    2

    (.84-1

    .23)

    .79

    rs368794

    *

    KC

    TD

    15

    T

    68%

    68.5

    %

    67.5

    %

    1.0

    0

    1.3

    1.8

    .9

    5

    (.78-1

    .17)

    .67

    rs10838738

    M

    TC

    H2

    G

    34%

    34.0

    %

    32.5

    %

    .45

    1.3

    1.7

    1.0

    6

    (.87-1

    .29)

    .51

    rs2568958

    *

    NE

    GR

    1

    A

    62%

    60.2

    %

    57.9

    %

    .30

    1.3

    1.8

    .9

    0

    (.75-1

    .09)

    .32

    rs1488830

    *

    BD

    NF

    T

    79%

    75.9

    %

    78.0

    %

    .69

    1.5

    2.1

    1.1

    3

    (.91-1

    .40)

    .26

    rs925946

    B

    DN

    F

    T

    30%

    28.1

    %

    29.2

    %

    .57

    1.3

    1.8

    .9

    5

    (.77-1

    .16)

    .62

    rs7647305

    E

    TV

    5

    C

    80%

    79.9

    %

    80.1

    %

    .93

    1.5

    2.2

    1.0

    1

    (.80-1

    .27)

    .93

    SN

    P,

    Sin

    gle

    Nucle

    oti

    de

    Po

    lym

    orp

    his

    m;

    BM

    I, b

    ody m

    ass

    index;

    Fre

    q.,

    fre

    quency;

    Co

    nt.

    , co

    ntr

    ols

    ; O

    R,

    odds

    rati

    o;

    CI,

    confi

    dence

    inte

    rvals

    ; H

    WE

    , H

    ard

    y-W

    ein

    ber

    g e

    quil

    ibri

    um

    ; χ

    2

    test

    wit

    h 1

    df

    for

    HW

    E;

    assu

    mp

    tio

    ns

    for

    pow

    er c

    alcula

    tio

    n:

    all

    eli

    c te

    st (

    1d

    f), α

    =.0

    5,

    pre

    vale

    nce

    =.0

    2.

    * S

    NP

    s in

    LD

    wit

    h S

    NP

    s id

    enti

    fied i

    n G

    WA

    stu

    die

    s o

    f B

    MI

    (pro

    xie

    s)

  • Chapter 2

    39

    Methods and materials

    A total of 13 BMI-associated single nucleotide polymorphisms (SNP), selected

    from the recent GWA studies of BMI 1-4, were genotyped in 267 AN patients

    and 1636 control individuals. Nine SNPs were the same as those identified in

    the GWA studies and four SNPs were in perfect or high linkage disequilibrium

    (LD; r2 > 0.84) with the SNPs of interest (Table I).

    The patients’ group consisted of female AN cases with ascertained

    Dutch descent (patients are asked whether all of their grandparents were of

    Dutch origin). There were 182 AN restrictive type and 99 AN purging type

    cases. Subjects were recruited for the study after referral to Eating Disorders

    treatment center (in- and outpatients, at various stages of the disease).

    Diagnosis was established by experienced clinicians according to DSM-IV

    criteria, with use of a semi-structured interview (Eating Disorder

    Examination; 9). Cases in which AN was not the primary diagnosis or with

    physical illnesses such as diabetes mellitus were excluded.

    The control group consisted of a random sample of Dutch female

    participants in the Utrecht contribution to the European Prospective

    Investigation into Cancer and Nutrition, also known as Prospect-EPIC 10. Lack

    of selection criteria is balanced by a relatively large size of this random

    population sample.

    Fourteen (5%) out of 281 cases and twenty (1%) out of 1656 controls

    were excluded because of more than two missing genotypes. In the

    remaining 267 cases and 1636 controls the mean age (SD) was 22.4 (4.3)

    years and 49.0 (6.0) years, and the mean BMI was 16.4 (2.1) kg/m2 and 25.9

    (4.0) kg/m2, respectively.

    Genotyping call rate among successfully genotyped individuals was

    98.4 %. Apart from SNP rs2844479, which was excluded from further analysis

    because of significant difference in missing calls between cases and controls,

    all SNPs passed the quality control requirements (more than 95% successful

    calls per SNP, Hardy-Weinberg Equilibrium test (χ2; 1 degree of freedom (df))

  • Chapter 2

    40

    p > .01, minor allele frequency > .05, difference in missing calls between

    groups at p>.01). To further ensure quality, blind duplicates were included on

    plates (100% concordance of duplicates, excluding missed calls). Genotyping

    was performed on a commercial platform (KBiosciences; Hertsfordshire,

    U.K.). Statistical analyses were conducted with PLINK 11, UNPHASED 12 and

    SPSS 15.0 (SPSS, Chicago, Illinois).

    In the first step of analysis we performed case-control tests for

    individual SNPs using a standard allelic test with 1 df. In the next step, after

    having ascertained which alleles from the SNPs identified in GWA studies 1-4

    were carrying the risk for higher BMI, we combined the information from 12

    SNPs by counting the number of BMI-increasing alleles present in each

    subject. Only subjects with 12 complete genotypes were included, i.e. 225

    cases and 1351 controls. This score was entered into a logistic regression

    model with case-control status as an outcome.

    Figure 1. Distribution of BMI-increasing alleles in cases and controls.

    Results

    The analysis of individual SNPs showed an absence of association between

    any of the studied markers and AN (allelic test, 1 df). Assuming a power of

    80%, an α-level of .05 and a disease prevalence of 2.2 % 13, we would be able

    to detect an association with odds ratio of at least 1.3 or 0.77 for a

    heterozygote and 1.7 or 0.59 for a risk homozygote for most of the SNPs

    0

    2

    4

    6

    8

    10

    12

    14

    16

    18

    20

    =18

    Number of BMI increasing alleles

    Perc

    enta

    ge o

    f in

    div

    iduals

    _

    cases

    controls

  • Chapter 2

    41

    individually, except for rs6548238, rs1488830, rs7647305 for which the

    effect sizes would have to be larger (Table I).

    Performing the same analysis solely on the ANR subset yielded

    nearly identical results, but with diminished power. For this reason both

    subsets are taken together in the study.

    To make sure that obese individuals in the control group were not relevantly

    influencing results we conducted a separate single SNP analysis with obese

    controls (BMI>30) excluded. Results were not materially different (data not

    shown).

    To test whether variants increasing BMI in the general population

    play a role in AN, we entered the combined number of effect alleles (i.e.

    BMI-increasing alleles from GWAS 1-4) into a logistic regression model.

    Frequencies of cases and controls per number of effect alleles are shown in

    Fig. 1 and the results of the logistic regression analysis are presented in Table

    II.

    The logistic regression analysis, assuming an α-level of .05, 80%

    power and a two-sided hypothesis, would be able to detect a change from

    the baseline probability (prevalence of the disease) of .02 to .05 with an

    increase of one SD (SD=2.47) in a number of effect alleles.

    TABLE II. Logistic regression: the number of effect alleles is not associated with

    probability of being a case

    Independent variable Β-coefficient df p-

    value OR

    95% CI

    Lower Upper

    Number of effect alleles .00 1 .84 1.00 .95 1.06

    Β-coefficient represents a change in probability of being a case with each additional risk-

    allele; OR, represents an increase in the odds of being a case with each additional risk-allele.

    The analysis included only individuals with 12 complete genotypes; n cases = 225; n controls

    = 1351. Mean (SD) number of effect alleles in cases = 12.20(2.32) and controls =

    12.17(2.50). CI, confidence intervals.

    The number of BMI-increasing alleles was not associated with the risk of AN

    (p = 0.84; Table II).

  • Chapter 2

    42

    Accordingly, mean numbers of effect alleles were similar between groups

    (p=.48 in a t-test) with a mean (SD) of 12.3 (2.4) alleles in cases and 12.2 (2.5)

    in controls (with α=.05 and at 80% power the test would detect difference in

    means of at least .18).

    FIG 2. A plot showing OR for being a case along increasing number of

    effect alleles. The vertical bars represent 95% CIs.

    Discussion

    In the present study 12 SNPs found to be associated with BMI in the general

    population 1-4 were successfully genotyped in AN cases (n=267) and

    population controls (n=1636). We found no evidence for association with the

    risk of AN. Furthermore, by calculating the combined score of effect alleles

    (i.e. BMI-increasing alleles from GWA studies of BMI 1-4) we tested whether

    genetic variants increasing BMI in the general population play a role in AN.

    Logistic regression model revealed absence of association between the

    number of effect alleles and the risk of AN.

    These results contribute to the discussion about a supposed

    continuum of eating disorders, normal weight range and obesity 14,15. In our

    study, on the level of genetic aetiology, AN appeared to be a discrete entity

    rather than a part of this continuum.

    0

    0.25

    0.5

    0.75

    1

    1.25

    1.5

    1.75

    2

    2.25

    =16

    Number of effect alleles

    Odds r

    atio _

  • Chapter 2

    43

    A limitation of the current study is its relatively small sample size and

    thus limited power to identify small effect sizes. Our sample has sufficient

    power (80%) to identify effect sizes of at least 1.3 OR at a 5% α-level, which is

    substantially larger than the effect sizes (OR 1.07 – 1.67) reported for obesity

    or extreme childhood obesity in the original GWA studies 1-4. With the same

    assumptions, a combined analysis of BMI-increasing alleles could detect a

    change in risk of the disease from baseline of .02 to .05, with an increase of

    one SD (2.47) in a number of effect alleles. To reduce phenotypic

    heterogeneity, we focused solely on AN because this subtype is distinct from

    the other types of eating disorders 15,16. Our main conclusions are based on

    the analysis of the combined score of effect alleles which, along with the fact

    that mean numbers of effect alleles between cases and controls were

    remarkably similar (power in the t-test sufficient to detect a difference of at

    least .18), shows that the investigated SNPs had no significant impact on

    susceptibility to AN. In this study no support was found for the hypothesis

    that the common genetic variants influencing BMI in the general population

    are substantial risk factors of AN, suggesting that effects of those variants

    may be overridden by other genetic factors of susceptibility to the disease.

    However, we cannot exclude that some association might be found with a

    considerable increase in sample size and refinement of phenotypes.

    In conclusion, this study found no evidence that SNPs which were

    previously proven to be robustly associated with BMI in the general

    population protect against or contribute to the risk of AN.

    Acknowledgements

    We are thankful to The GIANT (Genetic Investigation of ANtropometric

    Traits) Consortium for sharing the data on SNPs associated with BMI.

    This work was supported by funding from the Marie Curie Research Training

    Network INTACT (Individually tailored stepped care for women with eating

    disorders; reference number: MRTN-CT-2006-035988).

  • Chapter 2

    44

    We thank Bobby Koeleman, Behrooz Alizadeh and Caroline de Kovel for

    helpful comments.

    Financial disclosures

    The authors reported no potential conflicts of interests.

    References

    1. Willer CJ, Speliotes EK, Loos RJ, Li S, Lindgren CM, Heid IM, et al. Six new

    loci associated with body mass index highlight a neuronal influence on body

    weight regulation. Nat Genet 2009;41:25-34.

    2. Thorleifsson G, Walters GB, Gudbjartsson DF, Steinthorsdottir V, Sulem P,

    Helgadottir A, et al. Genome-wide association yields new sequence variants

    at seven loci that associate with measures of obesity. Nat Genet 2009;41:18-

    24.

    3. Frayling TM, Timpson NJ, Weedon MN, Zeggini E, Freathy RM, Lindgren

    CM, et al. A common variant in the FTO gene is associated with body mass

    index and predisposes to childhood and adult obesity. Science 2007;316:889-

    894.

    4. Loos RJF, Lindgren CM, Li S, Wheeler E, Zhao JH, Prokopenko I, et al.

    Common variants near MC4R are associated with fat mass, weight and risk of

    obesity. Nat Genet 2008;40:768-775.

    5. Stice E, Killen JD, Hayward C, Taylor CB. Support for the continuity

    hypothesis of bulimic pathology, J Consult Clin Psychol 1998;66:784-790.

    6. Steinhausen H, Grigoroiu-Serbanescu M, Boyadjieva S, Neumärker K,

    Metzke CW. The relevance of body weight in the medium-term to long-term

    course of adolescent anorexia nervosa. findings from a multisite study. Int J

    Eat Disord 2008;9999:NA.

  • Chapter 2

    45

    7. Hebebrand J, Remschmidt H. Anorexia nervosa viewed as an extreme

    weight condition: Genetic implications. Hum Genet 1995;95:1-11.

    8. Coners H, Remschmidt H, Hebebrand J. The relationship between

    premorbid body weight, weight loss, and weight at referral in adolescent

    patients with anorexia nervosa. Int J Eat Disord 1999;26:171-178.

    9. Cooper Z, Fairburn C. The eating disorder examination: A semi-structured

    interview for the assessment of the specific psychopathology of eating

    disorders. Int J Eat Disord 1987;6:1-8.

    10. Boker LK, van Noord PA, van der Schouw YT, Koot NV, Bueno de

    Mesquita HB, Riboli E, et al. Prospect-EPIC utrecht: Study design and

    characteristics of the cohort population. european prospective investigation

    into cancer and nutrition. Eur J Epidemiol 2001;17:1047-1053.

    11. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MAR, Bender D, et

    al. PLINK: A tool set for whole-genome association and population-based

    linkage analyses. The American Journal of Human Genetics 2007;81:559-575.

    12. Dudbridge F. Likelihood-based association analysis for nuclear families

    and unrelated subjects with missing genotype data. Hum Hered 2008;66:87-

    98.

    13. Keski-Rahkonen A, Hoek HW, Susser ES, Linna MS, Sihvola E, Raevuori A,

    et al. Epidemiology and course of anorexia nervosa in the community. Am J

    Psychiatry 2007;164:1259-1265.

    14. Collier DA, Treasure JL. The aetiology of eating disorders. The British

    Journal of Psychiatry 2004;185:363-365.

    15. Gleaves DH, Brown JD, Warren CS. The continuity/discontinuity models of

    eating disorders: A review of the literature and implications for assessment,

    treatment, and prevention. Behav Modif 2004;28:739-762.

    16. Keel PK, Fichter M, Quadflieg N, Bulik CM, Baxter MG, Thornton L, et al.

    Application of a latent class analysis to empirically define eating disorder

    phenotypes. Arch Gen Psychiatry 2004;61:192-200.

  • Chapter 3

    46

    Chapter 3

    Association study of POMC variants with body composition

    measures and nutrient choice

    Andrew Ternouth*

    Marek K. Brandys*

    Yvonne T. van der Schouw

    Judith Hendriks

    John-Olov Jansson

    David Collier

    Roger A. Adan

    *These authors contributed equally

    European Journal of Pharmacology 2011; 660(1): 220-5.

  • Chapter 3

    47

    Abstract

    Genome linkage scans and candidate gene studies have implicated the pro-

    opiomelanocortin (POMC) locus in traits related to food intake, metabolic

    function, and body mass index. Here we investigate single nucleotide

    polymorphisms at the POMC locus in order to evaluate the influence of its

    genetic variance on body fat distribution and diet in a sample of middle-aged

    men from the Netherlands. 366 Dutch males from the Hamlet cohort were

    asked detailed questions about food choice, nutrient intake and exercise.

    Furthermore, their weight and body fat composition were measured. Each

    cohort member was genotyped for a set of single nucleotide polymorphisms

    (SNPs) at the POMC locus. Regression analysis, adjusted for several

    covariates, was used to test for association between genetic variants and the

    phenotypes measured. POMC variation was associated with waist:hip ratio,

    visceral fat and abdominal fat (rs6713532, P=0.020, 0.019, 0.021,

    respectively), and nutrient choice (rs1042571, P=0.034), but in light of

    limited power and multiple testing these results should be taken with

    caution. POMC is a strong candidate for involvement in appetite regulation

    as supported by animal, physiological, and genetic studies and variation at

    the POMC locus may affect an individual’s energy intake which in turn leads

    to variation in body composition and


Recommended