Skip to main content
Erschienen in: European Child & Adolescent Psychiatry 3/2010

Open Access 01.03.2010 | Review

From monogenic to polygenic obesity: recent advances

verfasst von: Anke Hinney, Carla I. G. Vogel, Johannes Hebebrand

Erschienen in: European Child & Adolescent Psychiatry | Ausgabe 3/2010

Abstract

The heritability of obesity and body weight in general is high. A small number of confirmed monogenic forms of obesity—the respective mutations are sufficient by themselves to cause the condition in food abundant societies—have been identified by molecular genetic studies. The elucidation of these genes, mostly based on animal and family studies, has led to the identification of important pathways to the disorder and thus to a deeper understanding of the regulation of body weight. The identification of inborn deficiency of the mostly adipocyte-derived satiety hormone leptin in extremely obese children from consanguineous families paved the way to the first pharmacological therapy for obesity based on a molecular genetic finding. The genetic predisposition to obesity for most individuals, however, has a polygenic basis. A polygenic variant by itself has a small effect on the phenotype; only in combination with other predisposing variants does a sizeable phenotypic effect arise. Common variants in the first intron of the ‘fat mass and obesity associated’ gene (FTO) result in an elevated body mass index (BMI) equivalent to approximately +0.4 kg/m² per risk allele. The FTO variants were originally detected in a genome wide association study (GWAS) pertaining to type 2 diabetes mellitus. Large meta-analyses of GWAS have subsequently identified additional polygenic variants. Up to December 2009, polygenic variants have been confirmed in a total of 17 independent genomic regions. Further study of genetic effects on human body weight regulation should detect variants that will explain a larger proportion of the heritability. The development of new strategies for diagnosis, treatment and prevention of obesity can be anticipated.
Hinweise
An erratum to this article can be found at http://​dx.​doi.​org/​10.​1007/​s00787-010-0108-6

Introduction

Obesity is a complex disorder that is caused by several genetic and non-genetic risk factors [1]. The causes of obesity have been perceived differently over the past 100 years; both biological and psychological explanations have emerged. In the early part of the twentieth century, pituitary/hypothalamic dysfunction was assumed to lead to obesity [2]. From the 1940s through to the 1970s it was thought that psychological and psychodynamic aspects played the most prominent role [3, 4]. Prader, Labhart and Willi described the first syndromal form of obesity in 1956 [5]. From the 1970s on, pertinence of (psycho)social factors has been discussed [69]. Milestone twin and adoption studies at the end of the 1980s and during the early 1990s provided evidence that genetic factors play a considerable role in body weight regulation [1012]. Cloning of the leptin gene in 1994 [13] led to a rapid expansion of biomedical research. Large scale molecular genetic studies ensued [summarized in: 14]. The successful treatment of children with leptin deficiency [15] with recombinant leptin [1618] showed for the first time that mutations in a single gene could lead to hyperphagia and obesity. More recently support has increased for the hypothesis that obesity is a neuroendocrine disorder in which environmental risk factors and a genetic predisposition act in concert [5, 19, 20]. Recent large scale molecular genetic studies have substantiated that the genetic predisposition is in many cases due to the combined net effect of polygenic variants.
Environmental changes affecting both energy intake and expenditure are assumed to underlie the recent obesity epidemic as the gene pool of a population is unlikely to have changed substantially within the past generation [21]. This is in accordance with the ‘thrifty genotype hypothesis’ [22]. This hypothesis implies that gene variants which result in an increased energy deposition as fat have accumulated over time to maintain reproductive function as long as possible and to enhance survival during famines. Undoubtedly, gene–environment interactions will prove to play an important role in the aetiology of obesity. Such interactions could for example be conveyed via changes in DNA methylation patterns [23].
Recent discussions concern the definition of obesity or a subtype(s) of obesity as a psychiatric disorder(s). Resolution of this question depends on whether or not obesity can be viewed as an eating disorder [24]. Undoubtedly, obesity frequently has a behavioural basis; in addition, recent biomedical research has indicated that the brain and in particular the hypothalamus figure prominently in the regulation of appetite, body weight and physical activity. The respective neurotransmitter and neuropeptide pathways overlap with pathways involved in mood regulation and other psychological traits. Within the context of this special issue, a focus on the genetics of obesity is particularly warranted because body weight represents a readily quantifiable phenotype, which is thus amenable to quantitative genetics. This is in contrast to the categorical approach frequently taken for psychiatric disorders; however, due to the fact that many psychiatric disorders can also be viewed dimensionally, the recent success pertaining to the elucidation of polygenic variants involved in weight regulation serves to illustrate this approach. It may be noted that this success rests on the analysis of DNA samples of tens of thousands of individuals [24].

Twin, family and adoption studies

The strongest risk factor for childhood and adolescent obesity is parental obesity [25]. The risk is especially elevated if both parents are obese [26]. Some studies have found a stronger effect of maternal than paternal obesity [27] which may reflect pre- and postnatal environmental factors and sex-dependent genetic mechanisms. Maternal weight gain in pregnancy is positively correlated with the child’s BMI into adulthood [28].
Twin, family and adoption studies have led to the conclusion that the strong predictive value of parental BMI mainly stems from genetic rather than environmental factors [29, 30]. Twin studies, in comparison to family and adoption studies, have revealed that between 60 and 90% of the BMI variance within a population can be accounted for by genetic effects. In single comparatively small twin studies where the twins were reared apart, the heritability estimates proved to be similar to those where the twins were raised together [e.g. 12]. For example, maximum-likelihood model-fitting analyses and intrapair correlation coefficients for BMI of identical twins reared apart were approximately 0.7. This value was comparable to those for twins reared together. Non-additive genetic variance made a significant contribution to these estimates of heritability [12]. However, a substantial number of twin pairs reared apart were not separated immediately after birth, so that apart from intrauterine influences, a shared early postnatal environment could account for inflated heritability estimates [29, 31].
Age affects heritability estimates of BMI only to a minor degree. An exception is the newborn period, for which a substantially lower heritability (0.4) was calculated [32]. Intrauterine environment has a strong influence on birth weight. It is widely known that, especially for monozygotic (MZ) twins, other anthropometric measurements, like body height, also correlate less well in infancy than in childhood. A recent, large scale twin study (longitudinal sample of more than 3,500 twin pairs) indicated that the genetic influence on BMI became progressively stronger during childhood. Heritability estimates were shown to increase from 0.48 at age 4 years to 0.78 at age 11 [33]. According to one study [34], the heritability of BMI is highest during late adolescence (~0.9). However, one needs to be cautious with the interpretation of twin studies as they often underestimate shared environmental effects and tend to overestimate dominant genetic effects.
The pathophysiological and behavioural basis of obesity is complex and applies to ingestion, absorption, metabolism and energy expenditure. Both macronutrient intake [35] and activity levels [36] have been shown to be genetically determined. Restrained eating, drive for thinness and other eating behaviours show heritability estimates in the range of 0.20–0.55 [37]. It appears that even television viewing may have an, albeit small, heritable component [38].

Epigenetic considerations

Apart from variation at the DNA level, epigenetic events are also assumed to contribute to the obesity epidemic [39, 40]. Modern-day living could affect methylation patterns of specific genes which in turn increase the risk of obesity. Fraga et al. [41] observed that during the early years of life MZ twins are epigenetically indistinguishable from each other. However, with increasing age remarkable differences in their overall content and genomic distribution of 5-methylcytosine DNA and histone acetylation (with an effect on gene-expression) become apparent. Such changes induced by the environment could have an influence on individual BMI. A recent comparison of epigenetic metastability of 6,000 unique genomic regions between matched MZ and dizygotic (DZ) twins delineated epigenetic differences in both the MZ and DZ twins. As expected these differences were much more pronounced in samples of buccal cells from DZ co-twins. The epigenetic differences between MZ twins were most pronounced in buccal cells, but were also observed in gut and blood cells. Whereas DNA sequence differences could underlie the higher epigenetic discordance in DZ twins, animal studies and in silico SNP analyses favour the hypothesis that it is due to epigenomic differences in the zygotes. Molecular mechanisms of heritability may thus not be limited to DNA sequence differences [42].
In conclusion, family, twin and adoption studies have provided ample evidence for moderate to high heritability of BMI. Whereas, current molecular research is focussing on variation at the DNA level, epigenetic research will likely add a novel dimension towards the explanation of intra-individual variance of body weight.

Monogenic obesity

The elucidation of the mutations underlying diverse rodent mouse models (monogenic and one transgenic) led to the identification of most of the currently known monogenic forms of obesity in humans [43]. Family studies and in particular those on consanguineous pedigrees based on individuals with extreme obesity and in most cases with additional characteristic phenotypic features, like red hair, hyperinsulinism and infertility, proved to be highly successful in detecting mutations in the homologous human genes [43, 44]. This work proved fundamental in the elucidation of the leptinergic–melanocortinergic system. Put simply, leptin is a mostly adipocyte-derived satiety hormone that is secreted into the blood. It signals the size of the fat depot via the nucleus arcuatus in the hypothalamus. Increased leptin levels are registered by the leptin receptor. The signal is than forwarded to the melanocortin 4 receptor (MC4R) in higher order neurons. The endogenous agonist alpha-melanocyte stimulating hormone (α-MSH; product of the pro-opiomelanocortin gene; POMC) induces a satiating effect via the MC4R. The brain derived neurotrophic factor (BDNF) is involved in weight regulation downstream of the MC4R. Mutations in single genes of the leptinergic–melanocortinergic pathway have been shown to have a monogenic effect on the development of obesity.
The modes of inheritance of most of the aforementioned genes are autosomal recessive and autosomal dominant. The term monogenic obesity requires some specification and discussion: a ‘monogene’ is by common textbook definition, a gene with a strong effect on the phenotype [Mendelian traits or Mendelian (single gene) conditions], giving rise to a (close to) one-to-one relationship between genotype and phenotype. A ‘major gene’ is defined as a gene harbouring, a variant which is associated with a high lifetime risk for a disease; modifier genes and environmental factors additionally play a role in the aetiology of the respective diseases [4547].
The distinction between monogenic obesity and obesity due to a major gene effect may appear semantic. However, because BMI is a quantitative trait with obesity representing the right tail of the distribution, attempts to address this distinction are warranted.
Consider mutations in the MC4R, which have been reported to result in monogenic obesity [e.g. 48, 49]. In Mc4r knock-out mice the heterozygous animals have a body weight in between the wild-type and homozygous knockout mice [50]; body weight distributions of mice with heterozygous and wild-type genotypes overlap substantially, particularly in male rodents. The obese phenotype in the knock-out mice led to mutation screens of the human MC4R. The initial detections of human mutation carriers were all based on small pedigrees [5153]. Subsequent studies revealed that mutation carriers are not necessarily obese [e.g. 54, 55]. An epidemiological study based on approximately 4,000 individuals assessed within the framework of a population-based study revealed that none of the 6 individuals with functionally relevant MC4R mutations were obese [56]. Finally, in an attempt to adjust for familial background genetic and environmental factors Dempfle et al. [57] analysed current BMI of mutation and wild-type carriers in pedigrees based on index patients with MC4R mutations. The differences amounted to 4.5 and 9.5 kg/m² in males and females, respectively [57]. Accordingly, instead of using the term monogenic obesity, we consider it more appropriate to think of MC4R mutations as resulting in strong effect sizes conveying a high lifetime risk for obesity. The effect sizes of these mutations are lower than those of the leptin or leptin receptor gene mutations [43]. In contrast, the few individuals who were found to be homozygous or compound heterozygous for functionally relevant MC4R mutations were all extremely obese [55, 58].
Over the past 10–15 years mutations in several genes such as those coding for leptin (LEP [15, 59]), leptin receptor (LEPR [60]), prohormone convertase 1 (PC1 [61, 62]) and pro-opiomelanocortin (POMC [63, 64]) have been shown to lead to autosomal recessive forms of obesity [14]. These mutations are all rare and lead, apart from extreme obesity with an early onset, to additional phenotypical manifestations, including red hair (POMC), reduced or impaired fertility (PC1, leptin and LEPR), adrenal insufficiency (POMC), and impaired immunity (LEP) [16, 43, 65]. The pleiotropic effects warrant the consideration that these recessive disorders are classified as syndromal forms of obesity—similar to the Bardet–Biedl, Prader–Willi and other genetic syndromes associated with obesity [43, 66]. The pleiotropic Bardet–Biedl syndrome, of which obesity is one of the main clinical features, has been shown to have an oligogenic basis [14, 19, 43]. It was expected that the syndromic forms of obesity could help to unravel novel genes relevant for idiopathic obesity. However, although the genes for several of the syndromic forms have been detected, the relevance of these genes for general obesity is still unclear [14].
Both young [16, 17] and adult [67] individuals with inborn leptin deficiency have been treated successfully with recombinant leptin; weight loss (primarily fat mass) was shown to be substantial. Farooqi et al. [16, 18, 68, 69] described the rapidly altered eating behaviour upon initiation of treatment with recombinant leptin; food ingestion ceased to be the major focus of daily (and nightly) activity of the thus treated children [16, 18, 68, 69]. This is the first and until now only genetic form of human obesity that is amenable to successful pharmacological treatment.
The MC4R has been shown to comprise mutations entailing a substantially elevated body weight (see above) and two polymorphisms, which both have a small protective effect for the development of obesity (see below; e.g. [70, 71]. Currently, over 130 functionally relevant mutations have been detected in the human MC4R gene [72]. About 2–6% of extremely obese children and adolescents harbour such mutations. Among obese adults the prevalence is seemingly lower (1–2%). Approximately 0.5% of the normal weight population also harbour such mutations [56]. In vitro analyses have shown that most of these mutations lead to either total or partial loss of function [73, 74]. Thus, the endogenous agonist alpha-melanocyte stimulating hormone (α-MSH) is not able to induce its satiating effect to a full extent. Children with MC4R mutations have been shown to eat larger meals [58]. A negative effect of the mutations on energy expenditure also appears likely [75].
Phenotypical effects of MC4R mutations other than obesity have been shown to encompass hyperinsulinemia, elevated growth rate and higher bone density [58]. An initial report that MC4R mutations induce binge eating was not confirmed in subsequent investigations [19, 7679].

Statistical approaches

To detect new obesity gene variants, different approaches have been employed [80]:
(1)
Association studies
 
Several candidate genes have been analysed because of a priori biological, physiological or pharmacological evidence of their involvement in central or peripheral pathways controlling energy intake or expenditure [14]. A large number of conventional association studies involving obese cases and normal weight and/or lean controls, or less frequently studies of families have been performed [14]; meta-analyses have been reported for only a small number of such identified genetic variants. Unfortunately, classical candidate gene studies have proven largely unsuccessful in reliably detecting novel genetic variants relevant for obesity. Identification of a candidate gene can also rely on its location within a linkage region or a region identified via genome wide association studies (GWAS). In GWAS up to 2,000,000 genetic variants can currently be analysed for association with a given phenotype. This approach has been extremely successful for various phenotypes [81]. As the effect sizes are usually small, large groups need to be screened. Because in the pre-GWAS era sample sizes rarely exceeded 1,000 cases and 1,000 controls and in light of the small effect sizes of polygenic variants detected recently in GWAS, it must be concluded that most classical candidate gene studies were underpowered. It remains to be seen to what extent such genetic variants (and subsequently candidate genes) will be picked up in future large scale GWAS meta-analyses.
(2)
Genome wide approaches
 
(a) Genome wide linkage studies aim to identify chromosomal regions harbouring one or more genes relevant for a respective phenotype by making use of linkage data. Those regions underlying linkage peaks are narrowed down by fine mapping, so that candidate gene analyses can be pursued. Although more than 40 microsatellite-based genome wide linkage scans have been performed and single candidate genes have been detected, none of the results have been validated unequivocally [82]. This supports the contention that the effect sizes of genes influencing adiposity are small and/or that substantial heterogeneity exists [83].
(b) GWAS provide a better design to identify common variants with low to moderate penetrance which are relevant as risk factors for the trait of interest. Within a short time span, GWAS have proven to be very successful for the detection of polygenic variants. The production of high density SNP-chips has made GWAS feasible, which only recently have led to the identification of a number of confirmed genes for different complex disorders, thereby revolutionizing the molecular genetic analyses of complex disorders [81]. Stringent efforts to confirm (or reject) an original finding are absolutely crucial; due to the low effect sizes and possible gene–environment interactions it is to be expected that not every study will be able to confirm a true positive finding; meta-analyses are required. GWAS performed for obesity or body mass index (see Table 1) have already documented that this approach is extremely powerful to detect genetic variants relevant for the analysed phenotype(s). Currently known polygenic variants have a mean effect on BMI of approximately 0.03–0.5 kg/m²; it appears reasonable that these effect sizes represent the upper limit of such common variants [8486]. Independent gene variants with small but replicable effects on body weight have been identified unambiguously in 17 gene regions so far [86].
Table 1
Genetic variants with a polygenic effect on body weight in humans
SNP
Chr
Position
Nearest gene
Sample size in the original publicationa
Frequency of the risk allele (risk allele)
Effect on BMI in the original publication
References
rs2815752
1
72,524,461
NEGR1
32,387
62% (A)
+0.10 kg/m² per A alleleb
[92]
rs2568958
1
72,537,704
NEGR1
25,344
58% (A)
+0.43 kg/m² for AA genotypec
[84]
rs10913469
1
176,180,142
SEC16B, RASAL2
25,344
20% (C)
+0.50 kg/m² for CC genotypec
[84]
rs6548238
2
624,905
TMEM18
32,387
84% (C)
+0.26 kg/m² per C alleleb
[92]
rs7561317
2
634,953
TMEM18
25,344
84% (G)
+0.70 kg/m² for GG genotypec
[84]
rs7566605
2
118,552,495
INSIG2
9,881
37% (C)
+1.00 kg/m² for CC genotype
[121]
rs7647305
3
187,316,984
SFRS10, ETV5, DGKG
25,344
77% (C)
+0.54 kg/m² for CC genotypec
[84]
rs10938397
4
45,023,455
GNPDA2
32,387
48% (G)
+0.19 kg/m² per G alleleb
[92]
rs4712652
6
22,186,593
PRL
2,796
41% (A)
+0.031 kg/m² per A allele in childrend
[126]
rs10508503
10
16,339,956
PTER
2,796
8.5% (C)
+0.144 kg/m² per C allele in childrend
[126]
rs6265 (V66M)
11
27,636,492
BDNF
25,344
85% (G)
+0.67 kg/m² for GG genotypec
[84]
rs10838738
11
47,619,625
MTCH2
32,387
34% (G)
+0.07 kg/m² per G alleleb
[92]
rs7138803
12
48,533,735
BCDIN3D, FAIM2
25,344
37% (A)
+0.54 kg/m² for AA genotypec
[84]
rs7498665
16
28,790,742
SH2B1
32,387
41% (G)
+0.15 kg/m² per G alleleb
[92]
rs7498665
16
28,790,742
SH2B1, ATP2A1
25,344
44% (G)
+0.45 kg/m² for GG genotypec
[84]
rs8050136
16
52,373,776
FTO
25,344
41% (A)
+1.07 kg/m² for AA genotypec
[84]
rs9939609
16
52,378,028
FTO
38,759
40% (A)
+0.40 kg/m² per A allele
[95]
rs9939609
16
52,378,028
FTO
32,387
41% (A)
+0.33 kg/m² per A alleleb
[92]
rs1421085
16
52,358,455
FTO
2,796
40% (C)
+0.112 kg/m² per C alleled
[126]
rs1424233
16
78,240,251
MAF
2,796
43% (A)
+0.091 kg/m² per A allele in childrend
[126]
rs1805081
18
19,394,429
NPC1
2,796
44% (A)
−0.087 kg/m² per A allele in childrend
[126]
rs17782313
18
56,002,077
MC4R
16,876
24% (C)
+0.22 kg/m2 per C allele
[85]
rs17782313
18
56,002,077
MC4R
32,387
22% (C)
+0.22 kg/m² per C alleleb
[92]
rs17782313
18
56,002,077
MC4R
2,796
17,5% (C)
+0.097 kg/m² per C alleled
[126]
rs12970134
18
56,035,730
MC4R
25,344
30% (A)
+0.36 kg/m² for AA genotypec
[82]
rs52820871 (I251L)
18
56,189,806
MC4R
16,797
0.75% (251L)
−0.35 SD of their BMI Z-score per 251L allele
[71]
rs2229616 (V103I)
18
56,190,256
MC4R
7,713
2% (103I)
−0.48 kg/m² per 103I allele
[70]
rs29941
19
39,001,372
CHST8, KCTD15
25,344
70% (C)
+0.46 kg/m² for CC genotypec
[84]
rs11084753
19
39,013,977
KCTD15
32,387
67% (G)
+0.06 kg/m² per G alleleb
[92]
aEither in the GWAS or the initial sample
bReported in the population-based cohorts EPIC, FINRISK97, BPPP and METSIM (N = 18,812; [92])
cReported for the Islandic sample (N = 25,344; [84])
dreported for children from the Northern Finland Birth Cohort (N = 5,291; [126]), adapted from [86]
NEGR1: neuronal growth factor regulator 1; SEC16B; cerevisiae, homolog of, B; RASAL2: RAS protein activator like 2; TMEM18: transmembrane protein 18, INSIG2: insulin induced gene 2, SFRS10: splicing factor, arginine/serine-rich, 10; ETV5: ets variant 5; DGKG diacylglycerol kinase, gamma, 90kD, GNPDA2: glucosamine-6-phosphate deaminase 2; PRL: prolactin; PTER: phosphotriesterase related; BDNF: brain derived neurotrophic factor; MTCH2: mitochondrial carrier homolog 2 (C. elegans); BCDIN3D: BCDIN3 domain containing; FAIM2: Fas apoptotic inhibitory molecule 2; SH2B1: SH2B adaptor protein 1; ATP2A1: ATPase, Ca++ transporting, cardiac muscle, fast twitch 1; FTO: fat mass and obesity associated; MAF: v-maf musculoaponeurotic fibrosarcoma oncogene homolog (avian); NPC1: Niemann-Pick disease, type C1; MC4R: melanocortin 4 receptor; CHST8: carbohydrate (N-acetylgalactosamine 4-0) sulfotransferase 8; KCTD15: potassium channel tetramerisation domain containing 15

Polygenic obesity

Some traits can be due to the simultaneous presence of DNA variation in multiple genes. Any of a group of alleles at distinct gene loci that collectively control the inheritance of a quantitative phenotype or modify the expression of a qualitative character are termed ‘polygenic’ variants. It is generally assumed that for quantitative traits, each allele has a small effect and the allelic effects can be additive or non-additive. Potentially, many such polygenic variants play a role in body weight regulation. It is estimated that the total number of genes with a small effect most likely exceeds 100. The lower limit of effect sizes will most likely be well below 100 g. If an individual harbours many polygenic variants that increase body weight, obesity can ensue. Any single variant will have a higher frequency in obese than in normal weight and lean individuals [83]. A polygenic basis of obesity implies that the specific set of polygenic variants relevant for obesity in one individual is unlikely to be the same in another obese subject.

Melanocortin-4 receptor gene (MC4R)

The coding region of the MC4R harbours, in addition to the ‘obesity mutations’ mentioned above, two polygenic variants for weight regulation: the minor alleles of the polymorphisms coding for isoleucine instead of valine at amino acid position 103 (103I) and for leucine instead of isoleucine at position 251 (251L) of the receptor protein are negatively associated with obesity [70, 71, 8789], the mean BMI of such carriers is slightly reduced in comparison to homozygous carriers of the major allele. Heterozygosity for the I103 variant was found in 2–9% of subjects from different populations [70]. A family-based association test (TDT) in 520 trios ascertained via a young obese offspring originally revealed under-transmission of the allele coding for the I103 variant. A first meta-analysis comprising 7,713 cases and controls substantiated the evidence for a negative association of the I103 variant with obesity (odds ratio: 0.69). An effect estimate of −0.48 kg/m² was calculated for I103 carriers [70]. This negative association of I103 with obesity was subsequently confirmed in a single large epidemiological study group comprising 7,937 individuals [87] and two further meta-analyses encompassing a total of 55,195 individuals [88, 89]. The MC4 receptor variant harbouring I103 revealed a modest (twofold) decrease in antagonist hAGRP(87–132) potency [90], which is consistent with the obesity protective effect conferred by this variant.
The influence of the second non-synonymous coding polymorphism (I251L; rs52820871) on body weight was detected in 16,797 individuals of European origin from nine independent case–control, population-based studies and family cohorts. A consistent negative association of the L251 variant (minor allele frequency in cases and controls ranged from 0.41 to 1.21%) with both childhood and adult extreme obesity (odds ratio ranging from 0.25 to 0.76) was detected in eight of the nine studies; the variant was also associated with reduced BMI in population-based samples. A meta-analysis supported the evidence of the obesity protective effect of MC4R L251 (OR = 0.52) [71].
Most recently, SNP rs17782313 188 kb downstream of MC4R was detected via a large scaled GWAS [85], thus further substantiating the important contribution of genetic variation at the MC4R locus to inter-individual differences in BMI. For the first step, GWAS data from seven studies including 16,876 European individuals had been analyzed jointly. The second strongest association signal (p = 2.9 × 10−6, after FTO, see below) was at rs17782313. Due to its position downstream of the MC4R its influence on weight regulation is likely to be mediated through effects on MC4R expression. The finding was confirmed in 60,352 adults and 5,988 children and 660 German nuclear families encompassing one or more obese offspring and both parents. Each copy of the rs17782313 C-allele was associated with a mean difference in BMI of approximately +0.22 kg/m2 (+760 g). One copy of the C-allele resulted in an 8 and 12% increased odds ratio for overweight and obesity, respectively; no significant gender differences were detected. The effect on weight was disproportionately due to fat mass.
The effect of the C-allele was not detectable in children at birth and up to 42 months of age in the Avon Longitudinal Study of Children and Parents (almost 6,000 children) [85]. However, the effect size of one C-allele in children aged 7–11 was twice the amount observed in adults. Interestingly, in adults—but not in children—one copy of the C-allele also resulted in a higher mean height (0.21 cm) implying that this SNP [or the functionally relevant SNP(s) in linkage disequilibrium] also affects overall adult size. This finding has been replicated [9194].

Fat mass and obesity associated gene (FTO)

FTO was among the genes highlighted in GWAS for type 2 diabetes mellitus (T2DM) [95, 96]. By adjustment for BMI it was [95] found that the association to T2DM was actually due to the higher BMI of diabetic cases in comparison to non-diabetic controls. Confirmation of this effect on body weight was obtained in 13 cohorts with a total of 38,759 individuals within a meta-analysis showing that the A-allele of the variant rs9939609 (intron 1) was associated with a 31% increased risk to develop obesity. Among the adults 16% were homozygous for this risk-allele. When compared with individuals harbouring no copy of the risk-allele, they had a 1.67-fold increased odds ratio for obesity; mean body weights of these two groups differed by approximately 3 kg.
The association of SNPs in intron 1 of FTO has been confirmed in all sufficiently sized subsequent replication studies [e.g. 9799]. The first GWAS specifically designed with a primary focus on obesity was performed in 487 extremely obese German children and adolescents and 442 lean controls [99]. Six SNPs in FTO showed the strongest evidence for association with obesity, one of which remained significant after correcting for multiple testing. The odds ratios for obesity for heterozygosity and homozygosity for the risk allele were 1.67 and 2.76, respectively. A longitudinal study of children aged 7 months (n = 640) at baseline to 15 years, showed that the FTO effect became evident from age 7 years on. In this study, FTO variants were not associated with energy intake or physical activity [100]. However, association was found between FTO risk variants and increased energy intake in adults [101103] and decreased satiety [103], but not for energy expenditure [101, 102] or physical activity [100, 104].
Among 190 children and adolescents, those with at least one FTO risk allele reported episodes of loss of control over eating more frequently than individuals homozygous for the non-risk allele. Additionally, those children with risk alleles selected foods with a higher fat content at a buffet meal [105]. A large scale study (n = 4,839 population-based adults) revealed that low physical activity and high-fat diets might be especially obesity promoting in FTO risk allele carriers [106]. FTO risk alleles did not predict weight loss in obese children undergoing a 1-year intervention or their baseline fasting levels of blood glucose, triglycerides and cholesterol [107]. However, within the 280 participants the homozygous carriers of risk variants of both FTO and INSIG2 (insulin induced gene 2; see below) showed the lowest degree of overweight reduction [108]. As some of the studies mentioned above were rather small scale (n < 500 e.g. [100, 101, 105, 107109], additional confirmatory studies are warranted.
The strong association of genetic variation within FTO to obesity has led to a series of functional studies: FTO expression in skeletal muscle and adipose tissue in humans is seemingly not influenced by this variation [110]. An age-dependent decline in FTO expression was found to concur with peripheral defects of glucose and fat metabolism [112]. One of the obesity SNPs in intron 1 was associated with a reduced insulin effect on beta activity measured by magneto encephalography, which implicates a lower cerebro cortical response to insulin. This might be a mechanism by which variation in FTO contributes to the pathogenesis of obesity [111]. Wåhlén et al. [112] suggested a role of the FTO in fat cell lipolysis, thus providing a functional link to body weight regulation.
Human FTO is apparently a member of the non-heme dioxygenase (Fe(II)- and 2-oxoglutarate-dependent dioxygenases) superfamily [113, 114]. Amino acid conservation patterns support this hypothesis [114]. FTO belongs to the AlkB-related protein family [Gerken]. Recombinant murine Fto catalyzes demethylation of 3-methylthymine and 3-methyluracil residues in single-stranded DNA and RNA in vitro, although the efficiency is poor. Consistent with a potential role in nucleic acid demethylation, Fto localizes to the nucleus in transfected cells. Initially, it was criticised that 3-methylthymine and 3-methyluracil residues were analysed although they are rather uncommon in humans. Later it was shown, that Fto was also working, although less actively, on the more common forms of methylated DNA base damage (1-methyladenine and 3-metylcytosine) [112, 115]. Fto messenger RNA (mRNA) is most abundant (in comparison to all analysed tissues) in mouse brain and in particular in hypothalamic nuclei governing energy balance. In the arcuate nucleus, Fto mRNA levels have been found to be regulated by feeding and fasting [113]. FTO is ubiquitously expressed in human embryonic and adult tissues [116, 117].
Recently, it was shown that the loss of Fto in mice leads to postnatal growth retardation and a significant reduction in adipose tissue and lean body mass [118]. The leanness of Fto deficient mice (Fto /) develops as a consequence of increased energy expenditure and systemic sympathetic activation, despite decreased spontaneous locomotor activity and relative hyperphagia. The heterozygous Fto +/ mice have a reduced Fto expression leading to a significantly reduced weight gain after 12 weeks compared to wild-type mice. Thus, the animal model for the first time demonstrated that Fto is functionally involved in energy homeostasis by the control of energy expenditure [118]. A dominant point mutation induced by ENU (N-ethyl-N-nitrosourea) mutagenesis in the murine Fto gene also resulted in reduced fat mass and increased energy expenditure. In this model, locomotor activity was unaltered. High-fat diet led to increased lean mass and reduced fat mass in comparison to controls [119]. Based on these findings it appears that the human FTO risk variants in intron 1 are associated with an increased expression of FTO.
FTO is essential for normal development of the central nervous and cardiovascular systems in humans as shown by individuals with loss of function mutations in FTO: Boissel et al. [117] ascertained a large Palestinian Arab consanguineous multiplex family in which nine affected individuals presented with a novel polymalformation syndrome including postnatal growth retardation, microcephaly, severe psychomotor delay, functional brain deficits and characteristic facial dysmorphic features. Some patients also presented with structural brain malformations, cardiac defects, genital anomalies and cleft palate. Intercurrent infection or unidentified causes led to early lethality which occurred at 1–30 months of age. By linkage and sequence analyses a causative non-synonymous FTO mutation (R316Q) was detected homozygously in affected individuals. The mutation is located in an evolutionarily conserved region of FTO and leads to an inactivation of the enzymatic activity. Unfortunately, anthropometric data were not available for the unaffected family members; none of the heterozygous parents were clinically obese. Heterozygous carriers of loss of function mutations in FTO might be relatively resistant to become obese [117].
Recently, a large scale mutation screen in the coding region of FTO in 1,433 severely obese and 1,433 lean humans revealed novel mutations [120]. Functional in vitro studies showed that two of these mutations led to a reduced FTO function. Although the animal models implied that FTO deficiency should be related to leanness [118, 119] the two mutations were detected in both lean and obese individuals [120].

Insulin induced gene 2 (INSIG2)

The first GWAS for body weight (100k, Affymetrix) was conducted in 694 individuals from 288 families of the Framingham Heart Study [121]. In both children and adults, homozygosity for the minor allele of a common SNP (rs7566605) in the vicinity of INSIG2 was found to be associated with obesity. The initial finding was confirmed in four of five separate samples comprised of individuals of Western European ancestry and African-Americans. Approximately 10% of the individuals harboured the obesity prone CC genotype, which conferred an average increase in BMI of approximately one BMI unit. A meta-analysis comprising all case–control samples showed that under a recessive model, the CC genotype was significantly associated with obesity (OR = 1.22) [121].
Several attempts to replicate the INSIG2 finding have been undertaken. Both confirmations as well as negative findings have been reported [122125]. INSIG2 was not detected in any of the (joint) GWAS for obesity or BMI or in case–control studies [84, 86, 126]. A recent meta-analysis comprised data on a total of 74,345 individuals. Evidence for an overall association of rs7566605 with obesity was not detected. However, the data suggested an association with extreme degrees of obesity [127].

Other loci detected in genome wide association studies

Novel loci were identified by genome wide association studies in large study groups and via meta-analyses. Apart from the first large-scale meta-analysis mentioned above [85], three more recent GWAS reported novel obesity genes with small effects on human body weight [84, 92, 126]. A total of more than 150,000 individuals was analysed (Table 1).
A GWAS that was mainly based on individuals of the Icelandic population [84] resulted in a total of 29 variants in 11 chromosomal regions that reached genome wide significance, amongst them, variants located in seven new loci for obesity. Furthermore, FTO and MC4R were reconfirmed as well as the two obesity candidate genes brain derived neurotrophic factor gene (BDNF) and SH2B adaptor protein 1 gene (SH2B1) [84]. Four of the novel seven loci were not detected in the other two recent GWAS (see below and Table 1). Parallel to the Icelandic study [84], the meta-analysis of 15 GWAS for BMI (n = 32,387) was extended by the ‘Genetic Investigation of ANthropometric Traits’ consortium (GIANT) [92]; the study was also based on the GWAS data analysed by Loos et al. [85]. The best signals were now followed-up in 14 additional cohorts (n = 59,082). Again, strong confirmation was detected for FTO and MC4R. Additionally, six novel loci were identified (see Table 1).
Interestingly, for the first time a copy number variation (CNV) was described as potentially relevant for obesity [128] CNVs are by definition repeats of a size of 1 kb and more [129]. It is assumed that they might be involved in the regulation of neighbouring genes. The frequent ‘obesity-CNV’ (a 45-kb deletion) is located in a region near the neuronal growth regulator 1 gene (NEGR1). Further studies are required to unravel the underlying molecular mechanisms.
The newly detected variants lead to an increase of 0.06–0.33 kg/m² BMI units per allele. In adults with a height of 170 cm this corresponds to 173–954 g per allele. According to the recent GIANT study [92], the six newly discovered loci account for 0.40% and in combination with FTO and MC4R for a total of 0.84% of the BMI variance. The assessment of the combined impact of these loci on BMI revealed that individuals with 13 or more obesity predisposing alleles across the eight loci were on average 1.46 BMI units (equivalent to 3.7–4.7 kg for an adult of average height) heavier than those individuals with less than three of these alleles [92]; the effects proved to be additive.
The third recent GWAS [126] comprising roughly 3,000 Europeans and a large confirmatory study group revealed, in addition to FTO and MC4R, significant association with obesity for three new risk loci (see Table 1). The polymorphism coding for 103I of the MC4R [e.g. 17] was also reconfirmed.

Conclusions and perspectives

Animal models and family studies led to the identification of rare monogenic forms of human obesity. The more common MC4R mutations in our opinion are more consistent with a ‘major gene effect’, as the near one-to-one relationship between genotype and phenotype is not evident among carriers of the risk allele. Instead, these individuals have a substantially increased risk of developing obesity. This initial work led to the discovery of the leptinergic–melanocortinergic pathway, which figures prominently in body weight regulation. Genome wide linkage and classical candidate gene studies were largely unsuccessful to identify obesity genes; these studies were underpowered. The emergence of GWAS has recently revolutionized the field in terms of detection of variants with small effect sizes.
Seventeen solidly confirmed polygenic variants for body weight regulation have been reported as of October 2009. It is obvious that more polygenic variants await detection; undoubtedly large scale GWAS will prove powerful for their elucidation. It will be interesting to determine if the effect sizes of the currently known polygenic variants represent the upper limit. Effect sizes of many as yet undetected variants could well be in the range of <50 g only. The detection of such variants will require meta-analyses based on hundreds of thousands of individuals. For those currently known polygenes, studies on gene–gene interactions can be undertaken. Finally, gene–environment interactions also become amenable to statistical analyses.
If a large number of genetic variants are detected in the future, prediction will potentially become feasible. In most cases, a straightforward but detailed family history would currently provide much more predictive power. However, the analysis of a family history cannot indicate the involvement of specific genetic or biochemical mechanisms. Hence, genetic tests could provide much more detailed data pertaining to the pathways predisposing to obesity within the analysed family.
As obviously only a small fraction of the variance of BMI can currently be explained by variation at the DNA level, one might speculate about the molecular mechanisms underlying the large proportion of as yet unexplained variance. As already pointed out, the effect sizes of gene variants might be very small. Another explanation is that obesity frequently results from infrequent variants with larger effect sizes; substantial heterogeneity might preclude their detection. Additionally, one has to bear in mind that the highly dense SNP-chips only cover a major part but not all the variation in the human genome. Furthermore, haplotypes instead of SNPs might prove more successful for unraveling novel genes for obesity. Complex gene–gene and gene–environment interactions may also contribute to the currently low explained BMI variance. If at any given locus relevant for body weight regulation, some SNPs lead to an elevated expression, others to a reduced expression, such loci might also escape detection using current technology. Finally the role played by CNVs in obesity requires scrutiny and the influence of epigenetic factors on the development of obesity merits close attention.
The future will show to what extent BMI variance at the population level can be explained at the molecular level [130]; this will have a profound impact as to the feasibility of diagnostic kits to detect obesity genes. The current obesity intervention programs are not very efficient, so that novel approaches for treatment have to be developed. Gene products of every detected polygene can be considered as a potential drug target for the treatment of obesity and associated disorders.

Acknowledgments

This work was supported by grants from the Bundesministerium für Bildung und Forschung (NGFNplus 01GS0820 and 01KU0903), the Deutsche Forschungsgemeinschaft (HE 1446/4-1) and the European Union (FP6 LSHMCT-2003-503041). We thank Prof. Robert D. Oades for carefully reading and improving the manuscript.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Psychiatrie

Kombi-Abonnement

Mit e.Med Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Psychiatrie, den Premium-Inhalten der psychiatrischen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Walley AJ, Asher JE, Froguel P (2009) The genetic contribution to non-syndromic human obesity. Nat Rev Genet 10:431–442PubMed Walley AJ, Asher JE, Froguel P (2009) The genetic contribution to non-syndromic human obesity. Nat Rev Genet 10:431–442PubMed
2.
Zurück zum Zitat Fröhlich A (1901) Ein Fall von Tumor der Hypophysis cerebri ohne Akromegalie. Wiener Klinische Rundschau 15(833–836):906–908 Fröhlich A (1901) Ein Fall von Tumor der Hypophysis cerebri ohne Akromegalie. Wiener Klinische Rundschau 15(833–836):906–908
3.
Zurück zum Zitat Bruch H (1941) Obesity in childhood and personality development. Obes Res 5:157–161 Bruch H (1941) Obesity in childhood and personality development. Obes Res 5:157–161
4.
Zurück zum Zitat Bruch H (1964) Psychological aspects of overeating and obesity. Psychosomatics 5:269–274PubMed Bruch H (1964) Psychological aspects of overeating and obesity. Psychosomatics 5:269–274PubMed
5.
Zurück zum Zitat Hebebrand J, Hinney A (2009) Environmental and genetic risk factors in obesity. Child Adolesc Psychiatr Clin N Am 18:83–94PubMed Hebebrand J, Hinney A (2009) Environmental and genetic risk factors in obesity. Child Adolesc Psychiatr Clin N Am 18:83–94PubMed
6.
Zurück zum Zitat Garn SM, Clark D (1975) Nutrition, growth, development, and maturation: findings of the Ten State Nutritional Survey of 1968–70. Pediatrics 56:306–319PubMed Garn SM, Clark D (1975) Nutrition, growth, development, and maturation: findings of the Ten State Nutritional Survey of 1968–70. Pediatrics 56:306–319PubMed
7.
Zurück zum Zitat Lissau-Lind I, Sörensen TIA (1994) Parental neglect during childhood and increased risk of obesity in young adulthood. Lancet 343:324–327 Lissau-Lind I, Sörensen TIA (1994) Parental neglect during childhood and increased risk of obesity in young adulthood. Lancet 343:324–327
8.
Zurück zum Zitat Sobal J, Stunkard AJ (1989) Socioeconomic status and obesity: a review of the literature. Psychol Bull 105:260–275PubMed Sobal J, Stunkard AJ (1989) Socioeconomic status and obesity: a review of the literature. Psychol Bull 105:260–275PubMed
9.
Zurück zum Zitat Strauss RS, Knight J (1999) Influence of the home environment on the development of obesity in children. Pediatrics 103:e85PubMed Strauss RS, Knight J (1999) Influence of the home environment on the development of obesity in children. Pediatrics 103:e85PubMed
10.
Zurück zum Zitat Bouchard C, Tremblay A, Despres JP, Nadeau A, Lupien PJ, Theriault G, Dussault J, Moorjani S, Pinault S, Fournier G (1990) The response to long-term overfeeding in identical twins. N Engl J Med 322:1477–1482PubMed Bouchard C, Tremblay A, Despres JP, Nadeau A, Lupien PJ, Theriault G, Dussault J, Moorjani S, Pinault S, Fournier G (1990) The response to long-term overfeeding in identical twins. N Engl J Med 322:1477–1482PubMed
11.
Zurück zum Zitat Stunkard AJ, Sorensen TI, Hanis C, Teasdale TW, Chakraborty R, Schull WJ, Schulsinger F (1986) An adoption study of human obesity. N Engl J Med 314:193–198PubMedCrossRef Stunkard AJ, Sorensen TI, Hanis C, Teasdale TW, Chakraborty R, Schull WJ, Schulsinger F (1986) An adoption study of human obesity. N Engl J Med 314:193–198PubMedCrossRef
12.
Zurück zum Zitat Stunkard AJ, Harris JR, Pedersen NL, McClearn GE (1990) The body-mass index of twins who have been reared apart. N Engl J Med 322:1483–1487PubMed Stunkard AJ, Harris JR, Pedersen NL, McClearn GE (1990) The body-mass index of twins who have been reared apart. N Engl J Med 322:1483–1487PubMed
13.
Zurück zum Zitat Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM (1994) Positional cloning of the mouse obese gene and its human homologue. Nature 372:425–432PubMed Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM (1994) Positional cloning of the mouse obese gene and its human homologue. Nature 372:425–432PubMed
14.
Zurück zum Zitat Rankinen T, Zuberi A, Chagnon YC, Weisnagel SJ, Argyropoulos G, Walts B, Pérusse L, Bouchard C (2006) The human obesity gene map: the 2005 update. Obesity (Silver Spring) 14:529–644 Rankinen T, Zuberi A, Chagnon YC, Weisnagel SJ, Argyropoulos G, Walts B, Pérusse L, Bouchard C (2006) The human obesity gene map: the 2005 update. Obesity (Silver Spring) 14:529–644
15.
Zurück zum Zitat Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham NJ, Sewter CP, Digby JE, Mohammed SN, Hurst JA, Cheetham CH, Earley AR, Barnett AH, Prins JB, O’Rahilly S (1997) Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387:903–908PubMed Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham NJ, Sewter CP, Digby JE, Mohammed SN, Hurst JA, Cheetham CH, Earley AR, Barnett AH, Prins JB, O’Rahilly S (1997) Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387:903–908PubMed
16.
Zurück zum Zitat Farooqi IS, Jebb SA, Langmack G, Lawrence E, Cheetham CH, Prentice AM, Hughes IA, McCamish MA, O’Rahilly S (1999) Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med 341:879–884PubMed Farooqi IS, Jebb SA, Langmack G, Lawrence E, Cheetham CH, Prentice AM, Hughes IA, McCamish MA, O’Rahilly S (1999) Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med 341:879–884PubMed
17.
Zurück zum Zitat Gibson WT, Farooqi IS, Moreau M, DePaoli AM, Lawrence E, O’Rahilly S, Trussell RA (2004) Congenital leptin deficiency due to homozygosity for the Delta133G mutation: report of another case and evaluation of response to four years of leptin therapy. J Clin Endocrinol Metab 89:4821–4826PubMed Gibson WT, Farooqi IS, Moreau M, DePaoli AM, Lawrence E, O’Rahilly S, Trussell RA (2004) Congenital leptin deficiency due to homozygosity for the Delta133G mutation: report of another case and evaluation of response to four years of leptin therapy. J Clin Endocrinol Metab 89:4821–4826PubMed
18.
Zurück zum Zitat Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C, Sanna V, Jebb SA, Perna F, Fontana S, Lechler RI, DePaoli AM, O’Rahilly S (2002) Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest 110:1093–1103PubMed Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C, Sanna V, Jebb SA, Perna F, Fontana S, Lechler RI, DePaoli AM, O’Rahilly S (2002) Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest 110:1093–1103PubMed
19.
Zurück zum Zitat Hebebrand J (2007) Obesity. In: Martin A, Volkmar FR, Lewis M (eds) Lewis’s child and adolescent psychiatry: a comprehensive textbook. Lippincott Williams & Wilkins, Philadelphia, pp 602–614 Hebebrand J (2007) Obesity. In: Martin A, Volkmar FR, Lewis M (eds) Lewis’s child and adolescent psychiatry: a comprehensive textbook. Lippincott Williams & Wilkins, Philadelphia, pp 602–614
20.
Zurück zum Zitat Hebebrand J, Friedel S, Schäuble N, Geller F, Hinney A (2003) Perspectives: molecular genetic research in human obesity. Obes Rev 4:139–146PubMed Hebebrand J, Friedel S, Schäuble N, Geller F, Hinney A (2003) Perspectives: molecular genetic research in human obesity. Obes Rev 4:139–146PubMed
21.
Zurück zum Zitat Taubes G (1998) As obesity rates rise, experts struggle to explain why. Science 280:1367–1368PubMed Taubes G (1998) As obesity rates rise, experts struggle to explain why. Science 280:1367–1368PubMed
22.
Zurück zum Zitat Neel JV (1962) Diabetes mellitus: “thrifty” genotype rendered detrimental by “progress”? Am J Hum Genet 14:353–362PubMed Neel JV (1962) Diabetes mellitus: “thrifty” genotype rendered detrimental by “progress”? Am J Hum Genet 14:353–362PubMed
23.
Zurück zum Zitat Andreasen CH, Andersen G (2009) Gene-environment interactions and obesity–further aspects of genomewide association studies. Nutrition 25:998–1003PubMed Andreasen CH, Andersen G (2009) Gene-environment interactions and obesity–further aspects of genomewide association studies. Nutrition 25:998–1003PubMed
24.
Zurück zum Zitat Hebebrand J (2009) Diagnostic issues in eating disorders and obesity. Child Adolesc Psychiatric Clin N Am 18:1–16 Hebebrand J (2009) Diagnostic issues in eating disorders and obesity. Child Adolesc Psychiatric Clin N Am 18:1–16
25.
Zurück zum Zitat Reilly JJ, Armstrong J, Dorosty AR, Emmett PM, Ness A, Rogers I, Steer C, Sherriff A, Avon Longitudinal Study of Parents, Children Study Team (2005) Early life risk factors for obesity in childhood: cohort study. Br Med J 330:1357 Reilly JJ, Armstrong J, Dorosty AR, Emmett PM, Ness A, Rogers I, Steer C, Sherriff A, Avon Longitudinal Study of Parents, Children Study Team (2005) Early life risk factors for obesity in childhood: cohort study. Br Med J 330:1357
26.
Zurück zum Zitat Whitaker RC, Wright JA, Koepsell TD, Finch AJ, Psaty BM (1994) Characteristics of children selecting low-fat foods in an elementary school lunch program. Arch Pediatr Adolesc Med 148:1085–1091PubMed Whitaker RC, Wright JA, Koepsell TD, Finch AJ, Psaty BM (1994) Characteristics of children selecting low-fat foods in an elementary school lunch program. Arch Pediatr Adolesc Med 148:1085–1091PubMed
27.
Zurück zum Zitat Magnusson PK, Rasmussen F (2002) Familial resemblance of body mass index and familial risk of high and low body mass index. A study of young men in Sweden. Int J Obes Relat Metab Disord 26:1225–1231PubMed Magnusson PK, Rasmussen F (2002) Familial resemblance of body mass index and familial risk of high and low body mass index. A study of young men in Sweden. Int J Obes Relat Metab Disord 26:1225–1231PubMed
28.
Zurück zum Zitat Mamun AA, O’Callaghan M, Callaway L, Williams G, Najman J, Lawlor DA (2009) Associations of gestational weight gain with offspring body mass index and blood pressure at 21 years of age: evidence from a birth cohort study. Circulation 119:1720–1727PubMed Mamun AA, O’Callaghan M, Callaway L, Williams G, Najman J, Lawlor DA (2009) Associations of gestational weight gain with offspring body mass index and blood pressure at 21 years of age: evidence from a birth cohort study. Circulation 119:1720–1727PubMed
29.
Zurück zum Zitat Maes HH, Neale MC, Eaves LJ (1997) Genetic and environmental factors in relative body weight and human adiposity. Behav Genet 27:325–351PubMed Maes HH, Neale MC, Eaves LJ (1997) Genetic and environmental factors in relative body weight and human adiposity. Behav Genet 27:325–351PubMed
30.
Zurück zum Zitat Stunkard AJ, Sørensen TI (1993) Obesity and socioeconomic status—a complex relation. N Engl J Med 329:1036–1037PubMed Stunkard AJ, Sørensen TI (1993) Obesity and socioeconomic status—a complex relation. N Engl J Med 329:1036–1037PubMed
31.
Zurück zum Zitat Hebebrand J, Wulftange H, Goerg T, Ziegler A, Hinney A, Barth N, Mayer H, Remschmidt H (2000) Epidemic obesity: are genetic factors involved via increased rates of assortative mating? Int J Obes Relat Metab Disord 24:345–353PubMed Hebebrand J, Wulftange H, Goerg T, Ziegler A, Hinney A, Barth N, Mayer H, Remschmidt H (2000) Epidemic obesity: are genetic factors involved via increased rates of assortative mating? Int J Obes Relat Metab Disord 24:345–353PubMed
32.
Zurück zum Zitat Vlietinck R, Derom R, Neale MC, Maes H, van Loon H, Derom C, Thiery M (1989) Genetic and environmental variation in the birth weight of twins. Behav Genet 19:151–161PubMed Vlietinck R, Derom R, Neale MC, Maes H, van Loon H, Derom C, Thiery M (1989) Genetic and environmental variation in the birth weight of twins. Behav Genet 19:151–161PubMed
33.
Zurück zum Zitat Haworth CM, Carnell S, Meaburn EL, Davis OS, Plomin R, Wardle J (2008) Increasing heritability of BMI and stronger associations with the FTO gene over childhood. Obesity (Silver Spring) 16:2663–2668 Haworth CM, Carnell S, Meaburn EL, Davis OS, Plomin R, Wardle J (2008) Increasing heritability of BMI and stronger associations with the FTO gene over childhood. Obesity (Silver Spring) 16:2663–2668
34.
Zurück zum Zitat Pietiläinen KH, Kaprio J, Rissanen A, Winter T, Rimpelä A, Viken RJ, Rose RJ (1999) Distribution and heritability of BMI in Finnish adolescents aged 16y and 17y: a study of 4884 twins and 2509 singletons. Int J Obes Relat Metab Disord 23:107–115PubMed Pietiläinen KH, Kaprio J, Rissanen A, Winter T, Rimpelä A, Viken RJ, Rose RJ (1999) Distribution and heritability of BMI in Finnish adolescents aged 16y and 17y: a study of 4884 twins and 2509 singletons. Int J Obes Relat Metab Disord 23:107–115PubMed
35.
Zurück zum Zitat Reed DR, Bachmanov AA, Beauchamp GK, Tordoff MG, Price RA (1997) Heritable variation in food preferences and their contribution to obesity. Behav Genet 27:373–387PubMed Reed DR, Bachmanov AA, Beauchamp GK, Tordoff MG, Price RA (1997) Heritable variation in food preferences and their contribution to obesity. Behav Genet 27:373–387PubMed
36.
Zurück zum Zitat Pérusse L, Tremblay A, Leblanc C, Bouchard C (1989) Genetic and environmental influences on level of habitual physical activity and exercise participation. Am J Epidemiol 129:1012–1022PubMed Pérusse L, Tremblay A, Leblanc C, Bouchard C (1989) Genetic and environmental influences on level of habitual physical activity and exercise participation. Am J Epidemiol 129:1012–1022PubMed
37.
Zurück zum Zitat Rutherford J, McGuffin P, Katz RJ, Murray RM (1993) Genetic influences on eating attitudes in a normal female twin population. Psychol Med 23:425–436PubMed Rutherford J, McGuffin P, Katz RJ, Murray RM (1993) Genetic influences on eating attitudes in a normal female twin population. Psychol Med 23:425–436PubMed
38.
Zurück zum Zitat Plomin R, Corley R, Carey G, DeFries JC, Fulker DW (1990) Individual differences in television viewing in early childhood: nature as well as nurture. Psychol Sci 1:371–377 Plomin R, Corley R, Carey G, DeFries JC, Fulker DW (1990) Individual differences in television viewing in early childhood: nature as well as nurture. Psychol Sci 1:371–377
39.
Zurück zum Zitat Haemer MA, Huang TT, Daniels SR (2009) The effect of neurohormonal factors, epigenetic factors, and gut microbiota on risk of obesity. Prev Chronic Dis 6:A96PubMed Haemer MA, Huang TT, Daniels SR (2009) The effect of neurohormonal factors, epigenetic factors, and gut microbiota on risk of obesity. Prev Chronic Dis 6:A96PubMed
40.
Zurück zum Zitat Newnham JP, Pennell CE, Lye SJ, Rampono J, Challis JR (2009) Early life origins of obesity. Obstet Gynecol Clin North Am 36:227–244PubMed Newnham JP, Pennell CE, Lye SJ, Rampono J, Challis JR (2009) Early life origins of obesity. Obstet Gynecol Clin North Am 36:227–244PubMed
41.
Zurück zum Zitat Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, Heine-Suñer D, Cigudosa JC, Urioste M, Benitez J, Boix-Chornet M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, Spector TD, Wu YZ, Plass C, Esteller M (2005) Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci USA A102:10604–10609 Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, Heine-Suñer D, Cigudosa JC, Urioste M, Benitez J, Boix-Chornet M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, Spector TD, Wu YZ, Plass C, Esteller M (2005) Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci USA A102:10604–10609
42.
Zurück zum Zitat Kaminsky ZA, Tang T, Wang SC, Ptak C, Oh GH, Wong AH, Feldcamp LA, Virtanen C, Halfvarson J, Tysk C, McRae AF, Visscher PM, Montgomery GW, Gottesman II, Martin NG, Petronis A (2009) DNA methylation profiles in monozygotic and dizygotic twins. Nat Genet 41:240–245PubMed Kaminsky ZA, Tang T, Wang SC, Ptak C, Oh GH, Wong AH, Feldcamp LA, Virtanen C, Halfvarson J, Tysk C, McRae AF, Visscher PM, Montgomery GW, Gottesman II, Martin NG, Petronis A (2009) DNA methylation profiles in monozygotic and dizygotic twins. Nat Genet 41:240–245PubMed
43.
Zurück zum Zitat Farooqi IS, O’Rahilly S (2005) Monogenic obesity in humans. Annu Rev Med 56:443–458PubMed Farooqi IS, O’Rahilly S (2005) Monogenic obesity in humans. Annu Rev Med 56:443–458PubMed
44.
Zurück zum Zitat Farooqi IS, O’Rahilly S (2008) Mutations in ligands and receptors of the leptin-melanocortin pathway that lead to obesity. Nat Clin Pract Endocrinol Metab 4:569–577PubMed Farooqi IS, O’Rahilly S (2008) Mutations in ligands and receptors of the leptin-melanocortin pathway that lead to obesity. Nat Clin Pract Endocrinol Metab 4:569–577PubMed
45.
Zurück zum Zitat Narod SA (2002) Modifiers of risk of hereditary breast and ovarian cancer. Nat Rev Cancer 2:113–123PubMed Narod SA (2002) Modifiers of risk of hereditary breast and ovarian cancer. Nat Rev Cancer 2:113–123PubMed
46.
Zurück zum Zitat Lalouel JM, Rao DC, Morton NE, Elston RC (1983) A unified model for complex segregation analysis. Am J Hum Genet 35:816–826PubMed Lalouel JM, Rao DC, Morton NE, Elston RC (1983) A unified model for complex segregation analysis. Am J Hum Genet 35:816–826PubMed
47.
Zurück zum Zitat Khoury MJ, Beaty TH, Cohen BH (1993) Fundamentals of genetic epidemiology, vi. Oxford University Press, New York, p 383 Khoury MJ, Beaty TH, Cohen BH (1993) Fundamentals of genetic epidemiology, vi. Oxford University Press, New York, p 383
48.
Zurück zum Zitat Farooqi IS (2008) Monogenic human obesity. Front Horm Res 36:1–11PubMed Farooqi IS (2008) Monogenic human obesity. Front Horm Res 36:1–11PubMed
49.
Zurück zum Zitat Farooqi IS, O’Rahilly S (2006) Genetics of obesity in humans. Endocr Rev 27:710–718PubMed Farooqi IS, O’Rahilly S (2006) Genetics of obesity in humans. Endocr Rev 27:710–718PubMed
50.
Zurück zum Zitat Huszar D, Lynch CA, Fairchild-Huntress V, Dunmore JH, Fang Q, Berkemeier LR, Gu W, Kesterson RA, Boston BA, Cone RD, Smith FJ, Campfield LA, Burn P, Lee F (1997) Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell 88:131–141PubMed Huszar D, Lynch CA, Fairchild-Huntress V, Dunmore JH, Fang Q, Berkemeier LR, Gu W, Kesterson RA, Boston BA, Cone RD, Smith FJ, Campfield LA, Burn P, Lee F (1997) Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell 88:131–141PubMed
51.
Zurück zum Zitat Yeo GS, Farooqi IS, Aminian S, Halsall DJ, Stanhope RG, O’Rahilly S (1998) A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nat Genet 20:111–112PubMed Yeo GS, Farooqi IS, Aminian S, Halsall DJ, Stanhope RG, O’Rahilly S (1998) A frameshift mutation in MC4R associated with dominantly inherited human obesity. Nat Genet 20:111–112PubMed
52.
Zurück zum Zitat Vaisse C, Clement K, Guy-Grand B, Froguel P (1998) A frameshift mutation in human MC4R is associated with a dominant form of obesity. Nat Genet 20:113–114PubMed Vaisse C, Clement K, Guy-Grand B, Froguel P (1998) A frameshift mutation in human MC4R is associated with a dominant form of obesity. Nat Genet 20:113–114PubMed
53.
Zurück zum Zitat Hinney A, Schmidt A, Nottebom K, Heibült O, Becker I, Ziegler A, Gerber G, Sina M, Görg T, Mayer H, Siegfried W, Fichter M, Remschmidt H, Hebebrand J (1999) Several mutations in the melanocortin-4 receptor gene including a nonsense and a frameshift mutation associated with dominantly inherited obesity in humans. J Clin Endocrinol Metab 84:1483–1486PubMed Hinney A, Schmidt A, Nottebom K, Heibült O, Becker I, Ziegler A, Gerber G, Sina M, Görg T, Mayer H, Siegfried W, Fichter M, Remschmidt H, Hebebrand J (1999) Several mutations in the melanocortin-4 receptor gene including a nonsense and a frameshift mutation associated with dominantly inherited obesity in humans. J Clin Endocrinol Metab 84:1483–1486PubMed
54.
Zurück zum Zitat Vaisse C, Clement K, Durand E, Hercberg S, Guy-Grand B, Froguel P (2000) Melanocortin-4 receptor mutations are a frequent and heterogeneous cause of morbid obesity. J Clin Invest 106:253–262PubMed Vaisse C, Clement K, Durand E, Hercberg S, Guy-Grand B, Froguel P (2000) Melanocortin-4 receptor mutations are a frequent and heterogeneous cause of morbid obesity. J Clin Invest 106:253–262PubMed
55.
Zurück zum Zitat Hinney A, Hohmann S, Geller F, Vogel C, Hess C, Wermter AK, Brokamp B, Goldschmidt H, Siegfried W, Remschmidt H, Schäfer H, Gudermann T, Hebebrand J (2003) Melanocortin-4 receptor gene: case-control study and transmission disequilibrium test confirm that functionally relevant mutations are compatible with a major gene effect for extreme obesity. J Clin Endocrinol Metab 88:4258–4267PubMed Hinney A, Hohmann S, Geller F, Vogel C, Hess C, Wermter AK, Brokamp B, Goldschmidt H, Siegfried W, Remschmidt H, Schäfer H, Gudermann T, Hebebrand J (2003) Melanocortin-4 receptor gene: case-control study and transmission disequilibrium test confirm that functionally relevant mutations are compatible with a major gene effect for extreme obesity. J Clin Endocrinol Metab 88:4258–4267PubMed
56.
Zurück zum Zitat Hinney A, Bettecken T, Tarnow P, Brumm H, Reichwald K, Lichtner P, Scherag A, Nguyen TT, Schlumberger P, Rief W, Vollmert C, Illig T, Wichmann HE, Schäfer H, Platzer M, Biebermann H, Meitinger T, Hebebrand J (2006) Prevalence, spectrum, and functional characterization of melanocortin-4 receptor gene mutations in a representative populationbased sample and obese adults from Germany. J Clin Endocrinol Metab 91:1761–1769PubMed Hinney A, Bettecken T, Tarnow P, Brumm H, Reichwald K, Lichtner P, Scherag A, Nguyen TT, Schlumberger P, Rief W, Vollmert C, Illig T, Wichmann HE, Schäfer H, Platzer M, Biebermann H, Meitinger T, Hebebrand J (2006) Prevalence, spectrum, and functional characterization of melanocortin-4 receptor gene mutations in a representative populationbased sample and obese adults from Germany. J Clin Endocrinol Metab 91:1761–1769PubMed
57.
Zurück zum Zitat Dempfle A, Hinney A, Heinzel-Gutenbrunner M, Raab M, Geller F, Gudermann T, Schäfer H, Hebebrand J (2004) Large quantitative effect of melanocortin-4 receptor gene mutations on body mass index. J Med Genet 41:795–800PubMed Dempfle A, Hinney A, Heinzel-Gutenbrunner M, Raab M, Geller F, Gudermann T, Schäfer H, Hebebrand J (2004) Large quantitative effect of melanocortin-4 receptor gene mutations on body mass index. J Med Genet 41:795–800PubMed
58.
Zurück zum Zitat Farooqi IS, Keogh JM, Yeo GS, Lank EJ, Cheetham T, O’Rahilly S (2003) Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene. N Engl J Med 348:1085–1095PubMed Farooqi IS, Keogh JM, Yeo GS, Lank EJ, Cheetham T, O’Rahilly S (2003) Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene. N Engl J Med 348:1085–1095PubMed
59.
Zurück zum Zitat Strobel A, Issad T, Camoin L, Ozata M, Strosberg AD (1998) A leptin missense mutation associated with hypogonadism and morbid obesity. Nat Genet 18:213–215PubMed Strobel A, Issad T, Camoin L, Ozata M, Strosberg AD (1998) A leptin missense mutation associated with hypogonadism and morbid obesity. Nat Genet 18:213–215PubMed
60.
Zurück zum Zitat Clément K, Vaisse C, Lahlou N, Cabrol S, Pelloux V, Cassuto D, Gourmelen M, Dina C, Chambaz J, Lacorte JM, Basdevant A, Bougnères P, Lebouc Y, Froguel P, Guy-Grand B (1998) A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature 392:398–401PubMed Clément K, Vaisse C, Lahlou N, Cabrol S, Pelloux V, Cassuto D, Gourmelen M, Dina C, Chambaz J, Lacorte JM, Basdevant A, Bougnères P, Lebouc Y, Froguel P, Guy-Grand B (1998) A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature 392:398–401PubMed
61.
Zurück zum Zitat Jackson RS, Creemers JW, Ohagi S, Raffin-Sanson ML, Sanders L, Montague CT, Hutton JC, O’Rahilly S (1997) Obesity and impaired prohormone processing associated with mutations in the human prohormone convertase 1 gene. Nat Genet 16:303–306PubMed Jackson RS, Creemers JW, Ohagi S, Raffin-Sanson ML, Sanders L, Montague CT, Hutton JC, O’Rahilly S (1997) Obesity and impaired prohormone processing associated with mutations in the human prohormone convertase 1 gene. Nat Genet 16:303–306PubMed
62.
Zurück zum Zitat Jackson RS, Creemers JW, Farooqi IS, Raffin-Sanson ML, Varro A, Dockray GJ, Holst JJ, Brubaker PL, Corvol P, Polonsky KS, Ostrega D, Becker KL, Bertagna X, Hutton JC, White A, Dattani MT, Hussain K, Middleton SJ, Nicole TM, Milla PJ, Lindley KJ, O’Rahilly S (2003) Small-intestinal dysfunction accompanies the complex endocrinopathy of human proprotein convertase 1 deficiency. J Clin Invest 112:1550–1560PubMed Jackson RS, Creemers JW, Farooqi IS, Raffin-Sanson ML, Varro A, Dockray GJ, Holst JJ, Brubaker PL, Corvol P, Polonsky KS, Ostrega D, Becker KL, Bertagna X, Hutton JC, White A, Dattani MT, Hussain K, Middleton SJ, Nicole TM, Milla PJ, Lindley KJ, O’Rahilly S (2003) Small-intestinal dysfunction accompanies the complex endocrinopathy of human proprotein convertase 1 deficiency. J Clin Invest 112:1550–1560PubMed
63.
Zurück zum Zitat Krude H, Biebermann H, Luck W, Horn R, Brabant G, Grüters A (1998) Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nat Genet 19:155–157PubMed Krude H, Biebermann H, Luck W, Horn R, Brabant G, Grüters A (1998) Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nat Genet 19:155–157PubMed
64.
Zurück zum Zitat Challis BG, Pritchard LE, Creemers JW, Delplanque J, Keogh JM, Luan J, Wareham NJ, Yeo GS, Bhattacharyya S, Froguel P, White A, Farooqi IS, O’Rahilly S (2002) A missense mutation disrupting a dibasic prohormone processing site in pro-opiomelanocortin (POMC) increases susceptibility to early-onset obesity through a novel molecular mechanism. Hum Mol Genet 11:1997–2004PubMed Challis BG, Pritchard LE, Creemers JW, Delplanque J, Keogh JM, Luan J, Wareham NJ, Yeo GS, Bhattacharyya S, Froguel P, White A, Farooqi IS, O’Rahilly S (2002) A missense mutation disrupting a dibasic prohormone processing site in pro-opiomelanocortin (POMC) increases susceptibility to early-onset obesity through a novel molecular mechanism. Hum Mol Genet 11:1997–2004PubMed
65.
Zurück zum Zitat Farooqi IS (2006) Monogenic human obesity syndromes. Prog Brain Res 153:119–125PubMed Farooqi IS (2006) Monogenic human obesity syndromes. Prog Brain Res 153:119–125PubMed
66.
Zurück zum Zitat Kousta E, Hadjiathanasiou CG, Tolis G, Papathanasiou A (2009) Pleiotropic genetic syndromes with developmental abnormalities associated with obesity. J Pediatr Endocrinol Metab 22:581–592PubMed Kousta E, Hadjiathanasiou CG, Tolis G, Papathanasiou A (2009) Pleiotropic genetic syndromes with developmental abnormalities associated with obesity. J Pediatr Endocrinol Metab 22:581–592PubMed
67.
Zurück zum Zitat Licinio J, Caglayan S, Ozata M, Yildiz BO, de Miranda PB, O’Kirwan F, Whitby R, Liang L, Cohen P, Bhasin S, Krauss RM, Veldhuis JD, Wagner AJ, DePaoli AM, McCann SM, Wong ML (2004) Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin-deficient adults. Proc Natl Acad Sci USA 101:4531–4536PubMed Licinio J, Caglayan S, Ozata M, Yildiz BO, de Miranda PB, O’Kirwan F, Whitby R, Liang L, Cohen P, Bhasin S, Krauss RM, Veldhuis JD, Wagner AJ, DePaoli AM, McCann SM, Wong ML (2004) Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin-deficient adults. Proc Natl Acad Sci USA 101:4531–4536PubMed
68.
Zurück zum Zitat Farooqi IS, O’Rahilly S (2009) Leptin: a pivotal regulator of human energy homeostasis. Am J Clin Nutr 89:980S–984SPubMed Farooqi IS, O’Rahilly S (2009) Leptin: a pivotal regulator of human energy homeostasis. Am J Clin Nutr 89:980S–984SPubMed
69.
Zurück zum Zitat Farooqi S (2007) Insights from the genetics of severe childhood obesity. Horm Res 68(Suppl 5):5–7PubMed Farooqi S (2007) Insights from the genetics of severe childhood obesity. Horm Res 68(Suppl 5):5–7PubMed
70.
Zurück zum Zitat Geller F, Reichwald K, Dempfl e A, Illig T, Vollmert C, Herpertz S, Siffert W, Platzer M, Hess C, Gudermann T, Biebermann H, Wichmann HE, Schäfer H, Hinney A, Hebebrand J (2004) Melanocortin-4 receptor gene variant I103 is negatively associated with obesity. Am J Hum Genet 74:572–581PubMed Geller F, Reichwald K, Dempfl e A, Illig T, Vollmert C, Herpertz S, Siffert W, Platzer M, Hess C, Gudermann T, Biebermann H, Wichmann HE, Schäfer H, Hinney A, Hebebrand J (2004) Melanocortin-4 receptor gene variant I103 is negatively associated with obesity. Am J Hum Genet 74:572–581PubMed
71.
Zurück zum Zitat Stutzmann F, Vatin V, Cauchi S, Morandi A, Jouret B, Landt O, Tounian P, Levy-Marchal C, Buzzetti R, Pinelli L, Balkau B, Horber F, Bougnères P, Froguel P, Meyre D (2007) Non-synonymous polymorphisms in melanocortin-4 receptor protect against obesity: the two facets of a Janus obesity gene. Hum Mol Genet 16:1837–1844PubMed Stutzmann F, Vatin V, Cauchi S, Morandi A, Jouret B, Landt O, Tounian P, Levy-Marchal C, Buzzetti R, Pinelli L, Balkau B, Horber F, Bougnères P, Froguel P, Meyre D (2007) Non-synonymous polymorphisms in melanocortin-4 receptor protect against obesity: the two facets of a Janus obesity gene. Hum Mol Genet 16:1837–1844PubMed
73.
Zurück zum Zitat Tao YX (2005) Molecular mechanisms of the neural melanocortin receptor dysfunction in severe early onset obesity. Mol Cell Endocrinol 239:1–14PubMed Tao YX (2005) Molecular mechanisms of the neural melanocortin receptor dysfunction in severe early onset obesity. Mol Cell Endocrinol 239:1–14PubMed
74.
Zurück zum Zitat Tao YX, Segaloff DL (2003) Functional characterization of melanocortin-4 receptor mutations associated with childhood obesity. Endocrinology 144:4544–4551PubMed Tao YX, Segaloff DL (2003) Functional characterization of melanocortin-4 receptor mutations associated with childhood obesity. Endocrinology 144:4544–4551PubMed
75.
Zurück zum Zitat Cai G, Cole SA, Butte N, Bacino C, Diego V, Tan K, Göring HH, O’Rahilly S, Farooqi IS, Comuzzie AG (2006) A quantitative trait locus on chromosome 18q for physical activity and dietary intake in Hispanic children. Obesity (Silver Spring) 14:1596–1604 Cai G, Cole SA, Butte N, Bacino C, Diego V, Tan K, Göring HH, O’Rahilly S, Farooqi IS, Comuzzie AG (2006) A quantitative trait locus on chromosome 18q for physical activity and dietary intake in Hispanic children. Obesity (Silver Spring) 14:1596–1604
76.
Zurück zum Zitat Branson R, Potoczna N, Kral JG, Lentes KU, Hoehe MR, Horber FF (2003) Binge eating as a major phenotype of melanocortin 4 receptor gene mutations. N Engl J Med 348:1096–1103PubMed Branson R, Potoczna N, Kral JG, Lentes KU, Hoehe MR, Horber FF (2003) Binge eating as a major phenotype of melanocortin 4 receptor gene mutations. N Engl J Med 348:1096–1103PubMed
77.
Zurück zum Zitat Potoczna N, Branson R, Kral JG, Piec G, Steffen R, Ricklin T, Hoehe MR, Lentes KU, Horber FF (2004) Gene variants and binge eating as predictors of comorbidity and outcome of treatment in severe obesity. J Gastrointest Surg 8:971–981PubMed Potoczna N, Branson R, Kral JG, Piec G, Steffen R, Ricklin T, Hoehe MR, Lentes KU, Horber FF (2004) Gene variants and binge eating as predictors of comorbidity and outcome of treatment in severe obesity. J Gastrointest Surg 8:971–981PubMed
78.
Zurück zum Zitat Herpertz S, Siffert W, Hebebrand J (2003) Binge eating as a phenotype of melanocortin 4 receptor gene mutations. N Engl J Med 349:606–609PubMed Herpertz S, Siffert W, Hebebrand J (2003) Binge eating as a phenotype of melanocortin 4 receptor gene mutations. N Engl J Med 349:606–609PubMed
79.
Zurück zum Zitat Hebebrand J, Geller F, Dempfle A, Heinzel-Gutenbrunner M, Raab M, Gerber G, Wermter AK, Horro FF, Blundell J, Schäfer H, Remschmidt H, Hinney A (2004) Binge-eating episodes are not characteristic of carriers of melanocortin-4 receptor gene mutations. Mol Psychiatry 9:796–800PubMed Hebebrand J, Geller F, Dempfle A, Heinzel-Gutenbrunner M, Raab M, Gerber G, Wermter AK, Horro FF, Blundell J, Schäfer H, Remschmidt H, Hinney A (2004) Binge-eating episodes are not characteristic of carriers of melanocortin-4 receptor gene mutations. Mol Psychiatry 9:796–800PubMed
80.
Zurück zum Zitat Lander ES, Schork NJ (1994) Genetic dissection of complex traits. Science 265:2037–2048PubMed Lander ES, Schork NJ (1994) Genetic dissection of complex traits. Science 265:2037–2048PubMed
81.
Zurück zum Zitat Frayling TM (2007) Genome-wide association studies provide new insights into type 2 diabetes aetiology. Nat Rev Genet 8:657–662PubMed Frayling TM (2007) Genome-wide association studies provide new insights into type 2 diabetes aetiology. Nat Rev Genet 8:657–662PubMed
82.
Zurück zum Zitat Saunders CL, Chiodini BD, Sham P, Lewis CM, Abkevich V, Adeyemo AA, de Andrade M, Arya R, Berenson GS, Blangero J, Boehnke M, Borecki IB, Chagnon YC, Chen W, Comuzzie AG, Deng H-W, Duggirala R, Feitosa MF, Froguel P, Hanson RL, Hebebrand J, Huezo-Dias P, Kissebah AH, Li W, Luke A, Martin LJ, Nash M, Öhman M, Palmer LJ, Peltonen L, Perola M, Price RA, Redline S, Srinivasan SR, Stern MP, Stone S, Stringham H, Turner S, Wijmenga C, Collier DA (2007) Meta-analysis of genome-wide linkage studies in BMI and obesity. Obesity (Silver Spring) 15:2263–2275 Saunders CL, Chiodini BD, Sham P, Lewis CM, Abkevich V, Adeyemo AA, de Andrade M, Arya R, Berenson GS, Blangero J, Boehnke M, Borecki IB, Chagnon YC, Chen W, Comuzzie AG, Deng H-W, Duggirala R, Feitosa MF, Froguel P, Hanson RL, Hebebrand J, Huezo-Dias P, Kissebah AH, Li W, Luke A, Martin LJ, Nash M, Öhman M, Palmer LJ, Peltonen L, Perola M, Price RA, Redline S, Srinivasan SR, Stern MP, Stone S, Stringham H, Turner S, Wijmenga C, Collier DA (2007) Meta-analysis of genome-wide linkage studies in BMI and obesity. Obesity (Silver Spring) 15:2263–2275
83.
Zurück zum Zitat Hinney A, Hebebrand J (2008) Polygenic obesity in humans. Obes Facts 1:35–42PubMed Hinney A, Hebebrand J (2008) Polygenic obesity in humans. Obes Facts 1:35–42PubMed
84.
Zurück zum Zitat Thorleifsson G, Walters GB, Gudbjartsson DF, Steinthorsdottir V, Sulem P, Helgadottir A, Styrkarsdottir U, Gretarsdottir S, Thorlacius S, Jonsdottir I, Jonsdottir T, Olafsdottir EJ, Olafsdottir GH, Jonsson T, Jonsson F, Borch-Johnsen K, Hansen T, Andersen G, Jorgensen T, Lauritzen T, Aben KK, Verbeek AL, Roeleveld N, Kampman E, Yanek LR, Becker LC, Tryggvadottir L, Rafnar T, Becker DM, Gulcher J, Kiemeney LA, Pedersen O, Kong A, Thorsteinsdottir U, Stefansson K (2009) Genome-wide association yields new sequence variants at seven loci that associate with measures of obesity. Nat Genet 41:18–24PubMed Thorleifsson G, Walters GB, Gudbjartsson DF, Steinthorsdottir V, Sulem P, Helgadottir A, Styrkarsdottir U, Gretarsdottir S, Thorlacius S, Jonsdottir I, Jonsdottir T, Olafsdottir EJ, Olafsdottir GH, Jonsson T, Jonsson F, Borch-Johnsen K, Hansen T, Andersen G, Jorgensen T, Lauritzen T, Aben KK, Verbeek AL, Roeleveld N, Kampman E, Yanek LR, Becker LC, Tryggvadottir L, Rafnar T, Becker DM, Gulcher J, Kiemeney LA, Pedersen O, Kong A, Thorsteinsdottir U, Stefansson K (2009) Genome-wide association yields new sequence variants at seven loci that associate with measures of obesity. Nat Genet 41:18–24PubMed
85.
Zurück zum Zitat Loos RJF, Lindgren CM, Li S, Wheeler E, Zhao JH, Prokopenko I, Inouye M, Freathy RM, Attwood AP, Beckmann JS, Berndt SI, The Prostate, Lung, and Colorectal, Ovarian (PLCO) Cancer Screening Trial, Bergmann S, Bennett AJ, Bingham SA, Bochud M, Brown M, Cauchi S, Connell JM, Cooper C, Smith GD, Day I, Dina C, De S, Dermitzakis ET, Doney ASF, Elliott KS, Elliott P, Evans DM, Farooqi IS, Froguel P, Ghori J, Groves CJ, Gwilliam R, Hadley D, Hall AS, Hattersley AT, Hebebrand J, Heid IM, KORA, Herrera B, Hinney A, Hunt SE, Jarvelin MR, Johnson T, Jolley JDM, Karpe F, Keniry A, Khaw KT, Luben RN Mangino M, Mangino M, Marchini J, McArdle WL, McGinnis R, Meyre D, Munroe PB, Morris AD, Ness AR, Neville MJ, Nica AC, Ong KK, O’Rahilly S, Owen KR, Palmer CNA, Papadakis K, Potter S, Pouta A, Qi L, Nurses’ Health Study, Randall JC, Rayner NW, Ring SM, Sandhu MS, Scherag A, Sims MA, Song K, Soranzo N, Speliotes EK, Initiative Diabetes Genetics, Syddall HE, Teichmann SA, Timpson NJ, Tobias JH, Uda M, The SardiNIA Study, Ganz Vogel CI, Wallace C, Waterworth DM, Weedon MN, The Wellcome Trust Case Control Consortium, FUSION, Willer CJ, Wraight VL, Yuan X, Zeggini E, Hirschhorn JN, Strachan DP, Ouwehand WH, Caulfield MJ, Samani NJ, Frayling TM, Vollenweider P, Waeber G, Mooser V, Deloukas P, McCarthy MI, Wareham NJ, Barroso I, Jacobs KB, Chanock SJ, Hayes RB, Lamina C, Gieger C, Illig T, Meitinger T, Wichmann HE, Kraft P, Hankinson SE, Hunter DJ, Hu FB, Lyon HN, Voight BF, Ridderstrale M, Groop L, Scheet P, Sanna S, Abecasis GR, Albai G, Nagaraja R, Schlessinger D, Jackson AU, Tuomilehto J, Collins FS, Boehnke M, Mohlke KL (2008) Common variants near MC4R are associated with fat mass, weight and risk of obesity. Nat Genet 40:768–775PubMed Loos RJF, Lindgren CM, Li S, Wheeler E, Zhao JH, Prokopenko I, Inouye M, Freathy RM, Attwood AP, Beckmann JS, Berndt SI, The Prostate, Lung, and Colorectal, Ovarian (PLCO) Cancer Screening Trial, Bergmann S, Bennett AJ, Bingham SA, Bochud M, Brown M, Cauchi S, Connell JM, Cooper C, Smith GD, Day I, Dina C, De S, Dermitzakis ET, Doney ASF, Elliott KS, Elliott P, Evans DM, Farooqi IS, Froguel P, Ghori J, Groves CJ, Gwilliam R, Hadley D, Hall AS, Hattersley AT, Hebebrand J, Heid IM, KORA, Herrera B, Hinney A, Hunt SE, Jarvelin MR, Johnson T, Jolley JDM, Karpe F, Keniry A, Khaw KT, Luben RN Mangino M, Mangino M, Marchini J, McArdle WL, McGinnis R, Meyre D, Munroe PB, Morris AD, Ness AR, Neville MJ, Nica AC, Ong KK, O’Rahilly S, Owen KR, Palmer CNA, Papadakis K, Potter S, Pouta A, Qi L, Nurses’ Health Study, Randall JC, Rayner NW, Ring SM, Sandhu MS, Scherag A, Sims MA, Song K, Soranzo N, Speliotes EK, Initiative Diabetes Genetics, Syddall HE, Teichmann SA, Timpson NJ, Tobias JH, Uda M, The SardiNIA Study, Ganz Vogel CI, Wallace C, Waterworth DM, Weedon MN, The Wellcome Trust Case Control Consortium, FUSION, Willer CJ, Wraight VL, Yuan X, Zeggini E, Hirschhorn JN, Strachan DP, Ouwehand WH, Caulfield MJ, Samani NJ, Frayling TM, Vollenweider P, Waeber G, Mooser V, Deloukas P, McCarthy MI, Wareham NJ, Barroso I, Jacobs KB, Chanock SJ, Hayes RB, Lamina C, Gieger C, Illig T, Meitinger T, Wichmann HE, Kraft P, Hankinson SE, Hunter DJ, Hu FB, Lyon HN, Voight BF, Ridderstrale M, Groop L, Scheet P, Sanna S, Abecasis GR, Albai G, Nagaraja R, Schlessinger D, Jackson AU, Tuomilehto J, Collins FS, Boehnke M, Mohlke KL (2008) Common variants near MC4R are associated with fat mass, weight and risk of obesity. Nat Genet 40:768–775PubMed
86.
Zurück zum Zitat Hinney A, Hebebrand J (2009) Three at one swoop! Obes Facts 2:3–8PubMed Hinney A, Hebebrand J (2009) Three at one swoop! Obes Facts 2:3–8PubMed
87.
Zurück zum Zitat Heid IM, Vollmert C, Hinney A, Döring A, Geller F, Löwel H, Wichmann HE, Illig T, Hebebrand J, Kronenberg F, KORA Group (2005) Association of the 103I MC4R allele with decreased body mass in 7937 participants of two population based surveys. J Med Genet 42:e21PubMed Heid IM, Vollmert C, Hinney A, Döring A, Geller F, Löwel H, Wichmann HE, Illig T, Hebebrand J, Kronenberg F, KORA Group (2005) Association of the 103I MC4R allele with decreased body mass in 7937 participants of two population based surveys. J Med Genet 42:e21PubMed
88.
Zurück zum Zitat Young EH, Wareham NJ, Farooqi S, Hinney A, Hebebrand J, Scherag A, O’Rahilly S, Barroso I, Sandhu MS (2007) The V103I polymorphism of the MC4R gene and obesity: population based studies and meta-analysis of 29 563 individuals. Int J Obes (Lond) 31:1437–1441 Young EH, Wareham NJ, Farooqi S, Hinney A, Hebebrand J, Scherag A, O’Rahilly S, Barroso I, Sandhu MS (2007) The V103I polymorphism of the MC4R gene and obesity: population based studies and meta-analysis of 29 563 individuals. Int J Obes (Lond) 31:1437–1441
89.
Zurück zum Zitat Wang D, Ma J, Zhang S, Hinney A, Hebebrand J, Wang Y, Wang HJ (2009) Association of the MC4R V103I Polymorphism With Obesity: A Chinese case-control study and meta-analysis in 55,195 individuals. Obesity [Epub ahead of print] Wang D, Ma J, Zhang S, Hinney A, Hebebrand J, Wang Y, Wang HJ (2009) Association of the MC4R V103I Polymorphism With Obesity: A Chinese case-control study and meta-analysis in 55,195 individuals. Obesity [Epub ahead of print]
90.
Zurück zum Zitat Xiang Z, Litherland SA, Sorensen NB, Proneth B, Wood MS, Shaw AM, Millard WJ, Haskell-Luevano C (2006) Pharmacological characterization of 40 human melanocortin-4 receptor polymorphisms with the endogenous proopiomelanocortin-derived agonists and the agouti-related protein (AGRP) antagonist. Biochemistry 45:7277–7288PubMed Xiang Z, Litherland SA, Sorensen NB, Proneth B, Wood MS, Shaw AM, Millard WJ, Haskell-Luevano C (2006) Pharmacological characterization of 40 human melanocortin-4 receptor polymorphisms with the endogenous proopiomelanocortin-derived agonists and the agouti-related protein (AGRP) antagonist. Biochemistry 45:7277–7288PubMed
91.
Zurück zum Zitat Qi L, Kraft P, Hunter DJ, Hu FB (2008) The common obesity variant near MC4R gene is associated with higher intakes of total energy and dietary fat, weight change and diabetes risk in women. Hum Mol Genet 17:3502–3508PubMed Qi L, Kraft P, Hunter DJ, Hu FB (2008) The common obesity variant near MC4R gene is associated with higher intakes of total energy and dietary fat, weight change and diabetes risk in women. Hum Mol Genet 17:3502–3508PubMed
92.
Zurück zum Zitat Willer CJ, Speliotes EK, Loos RJ, Li S, Lindgren CM, Heid IM, Berndt SI, Elliott AL, Jackson AU, Lamina C, Lettre G, Lim N, Lyon HN, McCarroll SA, Papadakis K, Qi L, Randall JC, Roccasecca RM, Sanna S, Scheet P, Weedon MN, Wheeler E, Zhao JH, Jacobs LC, Prokopenko I, Soranzo N, Tanaka T, Timpson NJ, Almgren P, Bennett A, Bergman RN, Bingham SA, Bonnycastle LL, Brown M, Burtt NP, Chines P, Coin L, Collins FS, Connell JM, Cooper C, Smith GD, Dennison EM, Deodhar P, Elliott P, Erdos MR, Estrada K, Evans DM, Gianniny L, Gieger C, Gillson CJ, Guiducci C, Hackett R, Hadley D, Hall AS, Havulinna AS, Hebebrand J, Hofman A, Isomaa B, Jacobs KB, Johnson T, Jousilahti P, Jovanovic Z, Khaw KT, Kraft P, Kuokkanen M, Kuusisto J, Laitinen J, Lakatta EG, Luan J, Luben RN, Mangino M, McArdle WL, Meitinger T, Mulas A, Munroe PB, Narisu N, Ness AR, Northstone K, O’Rahilly S, Purmann C, Rees MG, Ridderstråle M, Ring SM, Rivadeneira F, Ruokonen A, Sandhu MS, Saramies J, Scott LJ, Scuteri A, Silander K, Sims MA, Song K, Stephens J, Stevens S, Stringham HM, Tung YC, Valle TT, Van Duijn CM, Vimaleswaran KS, Vollenweider P, Waeber G, Wallace C, Watanabe RM, Waterworth DM, Watkins N, WellcomeTrustCaseControl Consortium, Witteman JC, Zeggini E, Zhai G, Zillikens MC, Altshuler D, Caulfield MJ, Chanock SJ, Farooqi IS, Ferrucci L, Guralnik JM, Hattersley AT, Hu FB, Jarvelin MR, Laakso M, Mooser V, Ong KK, Ouwehand WH, Salomaa V, Samani NJ, Spector TD, Tuomi T, Tuomilehto J, Uda M, Uitterlinden AG, Wareham NJ, Deloukas P, Frayling TM, Groop LC, Hayes RB, Hunter DJ, Mohlke KL, Peltonen L, Schlessinger D, Strachan DP, Wichmann HE, McCarthy MI, Boehnke M, Barroso I, Abecasis GR, Hirschhorn JN, Genetic Investigation of ANthropometric Traits Consortium (2009) Six new loci associated with body mass index highlight a neuronal influence on body weight regulation. Nat Genet 41:25–34PubMed Willer CJ, Speliotes EK, Loos RJ, Li S, Lindgren CM, Heid IM, Berndt SI, Elliott AL, Jackson AU, Lamina C, Lettre G, Lim N, Lyon HN, McCarroll SA, Papadakis K, Qi L, Randall JC, Roccasecca RM, Sanna S, Scheet P, Weedon MN, Wheeler E, Zhao JH, Jacobs LC, Prokopenko I, Soranzo N, Tanaka T, Timpson NJ, Almgren P, Bennett A, Bergman RN, Bingham SA, Bonnycastle LL, Brown M, Burtt NP, Chines P, Coin L, Collins FS, Connell JM, Cooper C, Smith GD, Dennison EM, Deodhar P, Elliott P, Erdos MR, Estrada K, Evans DM, Gianniny L, Gieger C, Gillson CJ, Guiducci C, Hackett R, Hadley D, Hall AS, Havulinna AS, Hebebrand J, Hofman A, Isomaa B, Jacobs KB, Johnson T, Jousilahti P, Jovanovic Z, Khaw KT, Kraft P, Kuokkanen M, Kuusisto J, Laitinen J, Lakatta EG, Luan J, Luben RN, Mangino M, McArdle WL, Meitinger T, Mulas A, Munroe PB, Narisu N, Ness AR, Northstone K, O’Rahilly S, Purmann C, Rees MG, Ridderstråle M, Ring SM, Rivadeneira F, Ruokonen A, Sandhu MS, Saramies J, Scott LJ, Scuteri A, Silander K, Sims MA, Song K, Stephens J, Stevens S, Stringham HM, Tung YC, Valle TT, Van Duijn CM, Vimaleswaran KS, Vollenweider P, Waeber G, Wallace C, Watanabe RM, Waterworth DM, Watkins N, WellcomeTrustCaseControl Consortium, Witteman JC, Zeggini E, Zhai G, Zillikens MC, Altshuler D, Caulfield MJ, Chanock SJ, Farooqi IS, Ferrucci L, Guralnik JM, Hattersley AT, Hu FB, Jarvelin MR, Laakso M, Mooser V, Ong KK, Ouwehand WH, Salomaa V, Samani NJ, Spector TD, Tuomi T, Tuomilehto J, Uda M, Uitterlinden AG, Wareham NJ, Deloukas P, Frayling TM, Groop LC, Hayes RB, Hunter DJ, Mohlke KL, Peltonen L, Schlessinger D, Strachan DP, Wichmann HE, McCarthy MI, Boehnke M, Barroso I, Abecasis GR, Hirschhorn JN, Genetic Investigation of ANthropometric Traits Consortium (2009) Six new loci associated with body mass index highlight a neuronal influence on body weight regulation. Nat Genet 41:25–34PubMed
93.
Zurück zum Zitat Zobel DP, Andreasen CH, Grarup N, Eiberg H, Sørensen TI, Sandbaek A, Lauritzen T, Borch-Johnsen K, Jørgensen T, Pedersen O, Hansen T (2009) Variants near MC4R are associated with obesity and influence obesity-related quantitative traits in a population of middle-aged people: studies of 14, 940 Danes. Diabetes 58:757–764PubMed Zobel DP, Andreasen CH, Grarup N, Eiberg H, Sørensen TI, Sandbaek A, Lauritzen T, Borch-Johnsen K, Jørgensen T, Pedersen O, Hansen T (2009) Variants near MC4R are associated with obesity and influence obesity-related quantitative traits in a population of middle-aged people: studies of 14, 940 Danes. Diabetes 58:757–764PubMed
94.
Zurück zum Zitat Renström F, Payne F, Nordström A, Brito EC, Rolandsson O, Hallmans G, Barroso I, Nordström P, Franks PW, GIANT Consortium (2009) Replication and extension of genome-wide association study results for obesity in 4923 adults from northern Sweden. Hum Mol Genet 18:1489–1596PubMed Renström F, Payne F, Nordström A, Brito EC, Rolandsson O, Hallmans G, Barroso I, Nordström P, Franks PW, GIANT Consortium (2009) Replication and extension of genome-wide association study results for obesity in 4923 adults from northern Sweden. Hum Mol Genet 18:1489–1596PubMed
95.
Zurück zum Zitat Frayling TM, Timpson NJ, Weedon MN, Zeggini E, Freathy RM, Lindgren CM, Perry JR, Elliott KS, Lango H, Rayner NW, Shields B, Harries LW, Barrett JC, Ellard S, Groves CJ, Knight B, Patch AM, Ness AR, Ebrahim S, Lawlor DA, Ring SM, Ben-Shlomo Y, Jarvelin MR, Sovio U, Bennett AJ, Melzer D, Ferrucci L, Loos RJ, Barroso I, Wareham NJ, Karpe F, Owen KR, Cardon LR, Walker M, Hitman GA, Palmer CN, Doney AS, Morris AD, Smith GD, Hattersley AT, McCarthy MI (2007) A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 316:889–894PubMed Frayling TM, Timpson NJ, Weedon MN, Zeggini E, Freathy RM, Lindgren CM, Perry JR, Elliott KS, Lango H, Rayner NW, Shields B, Harries LW, Barrett JC, Ellard S, Groves CJ, Knight B, Patch AM, Ness AR, Ebrahim S, Lawlor DA, Ring SM, Ben-Shlomo Y, Jarvelin MR, Sovio U, Bennett AJ, Melzer D, Ferrucci L, Loos RJ, Barroso I, Wareham NJ, Karpe F, Owen KR, Cardon LR, Walker M, Hitman GA, Palmer CN, Doney AS, Morris AD, Smith GD, Hattersley AT, McCarthy MI (2007) A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 316:889–894PubMed
96.
Zurück zum Zitat Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, Erdos MR, Stringham HM, Chines PS, Jackson AU, Prokunina-Olsson L, Ding CJ, Swift AJ, Narisu N, Hu T, Pruim R, Xiao R, Li XY, Conneely KN, Riebow NL, Sprau AG, Tong M, White PP, Hetrick KN, Barnhart MW, Bark CW, Goldstein JL, Watkins L, Xiang F, Saramies J, Buchanan TA, Watanabe RM, Valle TT, Kinnunen L, Abecasis GR, Pugh EW, Doheny KF, Bergman RN, Tuomilehto J, Collins FS, Boehnke M (2007) A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 316:1341–1345PubMed Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, Erdos MR, Stringham HM, Chines PS, Jackson AU, Prokunina-Olsson L, Ding CJ, Swift AJ, Narisu N, Hu T, Pruim R, Xiao R, Li XY, Conneely KN, Riebow NL, Sprau AG, Tong M, White PP, Hetrick KN, Barnhart MW, Bark CW, Goldstein JL, Watkins L, Xiang F, Saramies J, Buchanan TA, Watanabe RM, Valle TT, Kinnunen L, Abecasis GR, Pugh EW, Doheny KF, Bergman RN, Tuomilehto J, Collins FS, Boehnke M (2007) A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 316:1341–1345PubMed
97.
Zurück zum Zitat Dina C, Meyre D, Gallina S, Durand E, Korner A, Jacobson P, Carlsson LM, Kiess W, Vatin V, Lecoeur C, Delplanque J, Vaillant E, Pattou F, Ruiz J, Weill J, Levy-Marchal C, Horber F, Potoczna N, Hercberg S, Le Stunff C, Bougneres P, Kovacs P, Marre M, Balkau B, Cauchi S, Chevre JC, Froguel P (2007) Variation in FTO contributes to childhood obesity and severe adult obesity. Nat Genet 39:724–726PubMed Dina C, Meyre D, Gallina S, Durand E, Korner A, Jacobson P, Carlsson LM, Kiess W, Vatin V, Lecoeur C, Delplanque J, Vaillant E, Pattou F, Ruiz J, Weill J, Levy-Marchal C, Horber F, Potoczna N, Hercberg S, Le Stunff C, Bougneres P, Kovacs P, Marre M, Balkau B, Cauchi S, Chevre JC, Froguel P (2007) Variation in FTO contributes to childhood obesity and severe adult obesity. Nat Genet 39:724–726PubMed
98.
Zurück zum Zitat Scuteri A, Sanna S, Chen WM, Uda M, Albai G, Strait J, Najjar S, Nagaraja R, Orru M, Usala G, Dei M, Lai S, Maschio A, Busonero F, Mulas A, Ehret GB, Fink AA, Weder AB, Cooper RS, Galan P, Chakravarti A, Schlessinger D, Cao A, Lakatta E, Abecasis GR (2007) Genome-wide Association scan shows genetic variants in the FTO gene are associated with obesity-related traits. PLoS Genetics 3:e115PubMed Scuteri A, Sanna S, Chen WM, Uda M, Albai G, Strait J, Najjar S, Nagaraja R, Orru M, Usala G, Dei M, Lai S, Maschio A, Busonero F, Mulas A, Ehret GB, Fink AA, Weder AB, Cooper RS, Galan P, Chakravarti A, Schlessinger D, Cao A, Lakatta E, Abecasis GR (2007) Genome-wide Association scan shows genetic variants in the FTO gene are associated with obesity-related traits. PLoS Genetics 3:e115PubMed
99.
Zurück zum Zitat Hinney A, Nguyen TT, Scherag A, Friedel S, Brönner G, Müller TD, Grallert H, Illig T, Wichmann H-E, Rief W, Schäfer H, Hebebrand J (2007) Genome Wide Association (GWA) study for early onset extreme obesity supports the role of fat mass and obesity associated gene (FTO) variants. PLoS ONE 2:e1361PubMed Hinney A, Nguyen TT, Scherag A, Friedel S, Brönner G, Müller TD, Grallert H, Illig T, Wichmann H-E, Rief W, Schäfer H, Hebebrand J (2007) Genome Wide Association (GWA) study for early onset extreme obesity supports the role of fat mass and obesity associated gene (FTO) variants. PLoS ONE 2:e1361PubMed
100.
Zurück zum Zitat Hakanen M, Raitakari OT, Lehtimäki T, Peltonen N, Pahkala K, Sillanmäki L, Lagström H, Viikari J, Simell O, Rönnemaa T (2009) FTO genotype is associated with body mass index after the age of seven years but not with energy intake or leisure-time physical activity. J Clin Endocrinol Metab 94:1281–1287PubMed Hakanen M, Raitakari OT, Lehtimäki T, Peltonen N, Pahkala K, Sillanmäki L, Lagström H, Viikari J, Simell O, Rönnemaa T (2009) FTO genotype is associated with body mass index after the age of seven years but not with energy intake or leisure-time physical activity. J Clin Endocrinol Metab 94:1281–1287PubMed
101.
Zurück zum Zitat Speakman JR, Rance KA, Johnstone AM (2008) Polymorphisms of the FTO gene are associated with variation in energy intake, but not energy expenditure. Obesity (Silver Spring) 16:1961–1965 Speakman JR, Rance KA, Johnstone AM (2008) Polymorphisms of the FTO gene are associated with variation in energy intake, but not energy expenditure. Obesity (Silver Spring) 16:1961–1965
102.
Zurück zum Zitat Cecil JE, Tavendale R, Watt P, Hetherington MM, Palmer CN (2008) An obesity-associated FTO gene variant and increased energy intake in children. N Engl J Med 359:2558–2566PubMed Cecil JE, Tavendale R, Watt P, Hetherington MM, Palmer CN (2008) An obesity-associated FTO gene variant and increased energy intake in children. N Engl J Med 359:2558–2566PubMed
103.
Zurück zum Zitat Wardle J, Carnell S, Haworth CM, Farooqi IS, O’Rahilly S, Plomin R (2008) Obesity associated genetic variation in FTO is associated with diminished satiety. J Clin Endocrinol Metab 93:3640–3643PubMed Wardle J, Carnell S, Haworth CM, Farooqi IS, O’Rahilly S, Plomin R (2008) Obesity associated genetic variation in FTO is associated with diminished satiety. J Clin Endocrinol Metab 93:3640–3643PubMed
104.
Zurück zum Zitat Jonsson A, Renström F, Lyssenko V, Brito EC, Isomaa B, Berglund G, Nilsson PM, Groop L, Franks PW (2009) Assessing the effect of interaction between an FTO variant (rs9939609) and physical activity on obesity in 15,925 Swedish and 2,511 Finnish adults. Diabetologia 52:1334–1348PubMed Jonsson A, Renström F, Lyssenko V, Brito EC, Isomaa B, Berglund G, Nilsson PM, Groop L, Franks PW (2009) Assessing the effect of interaction between an FTO variant (rs9939609) and physical activity on obesity in 15,925 Swedish and 2,511 Finnish adults. Diabetologia 52:1334–1348PubMed
105.
Zurück zum Zitat Tanofsky-Kraff M, Han JC, Anandalingam K, Shomaker LB, Columbo KM, Wolkoff LE, Kozlosky M, Elliott C, Ranzenhofer LM, Roza CA, Yanovski SZ, Yanovski JA (2009) The FTO gene rs9939609 obesity-risk allele and loss of control over eating. Am J Clin Nutr 90:1483–1488PubMed Tanofsky-Kraff M, Han JC, Anandalingam K, Shomaker LB, Columbo KM, Wolkoff LE, Kozlosky M, Elliott C, Ranzenhofer LM, Roza CA, Yanovski SZ, Yanovski JA (2009) The FTO gene rs9939609 obesity-risk allele and loss of control over eating. Am J Clin Nutr 90:1483–1488PubMed
106.
Zurück zum Zitat Sonestedt E, Roos C, Gullberg B, Ericson U, Wirfält E, Orho-Melander M (2009) Fat and carbohydrate intake modify the association between genetic variation in the FTO genotype and obesity. Am J Clin Nutr 90:1418–1425PubMed Sonestedt E, Roos C, Gullberg B, Ericson U, Wirfält E, Orho-Melander M (2009) Fat and carbohydrate intake modify the association between genetic variation in the FTO genotype and obesity. Am J Clin Nutr 90:1418–1425PubMed
107.
Zurück zum Zitat Müller TD, Hinney A, Scherag A, Nguyen TT, Schreiner F, Schäfer H, Hebebrand J, Roth CL, Reinehr T (2008) ‘Fat mass and obesity associated’ gene (FTO): no significant association of variant rs9939609 with weight loss in a lifestyle intervention and lipid metabolism markers in German obese children and adolescents. BMC Med Genet 9:85PubMed Müller TD, Hinney A, Scherag A, Nguyen TT, Schreiner F, Schäfer H, Hebebrand J, Roth CL, Reinehr T (2008) ‘Fat mass and obesity associated’ gene (FTO): no significant association of variant rs9939609 with weight loss in a lifestyle intervention and lipid metabolism markers in German obese children and adolescents. BMC Med Genet 9:85PubMed
108.
Zurück zum Zitat Reinehr T, Hinney A, Toschke AM, Hebebrand J (2009) Aggravating effect of INSIG2 and FTO on overweight reduction in a one-year lifestyle intervention. Arch Dis Child 94:965–967PubMed Reinehr T, Hinney A, Toschke AM, Hebebrand J (2009) Aggravating effect of INSIG2 and FTO on overweight reduction in a one-year lifestyle intervention. Arch Dis Child 94:965–967PubMed
109.
Zurück zum Zitat Mitchell JA, Church TS, Rankinen T, Earnest CP, Sui X, Blair SN (2009) FTO Genotype and the weight loss benefits of moderate intensity exercise. Obesity (Silver Spring). Doi:10.1038/oby.2009.311. Mitchell JA, Church TS, Rankinen T, Earnest CP, Sui X, Blair SN (2009) FTO Genotype and the weight loss benefits of moderate intensity exercise. Obesity (Silver Spring). Doi:10.​1038/​oby.​2009.​311.
110.
Zurück zum Zitat Grunnet LG, Nilsson E, Ling C, Hansen T, Pedersen O, Groop L, Vaag A, Poulsen P (2009) Regulation and function of FTO mRNA expression in human skeletal muscle and subcutaneous adipose tissue. Diabetes 58:2402–2408PubMed Grunnet LG, Nilsson E, Ling C, Hansen T, Pedersen O, Groop L, Vaag A, Poulsen P (2009) Regulation and function of FTO mRNA expression in human skeletal muscle and subcutaneous adipose tissue. Diabetes 58:2402–2408PubMed
111.
Zurück zum Zitat Tschritter O, Preissl H, Yokoyama Y, Machicao F, Häring HU, Fritsche A (2007) Variation in the FTO gene locus is associated with cerebrocortical insulin resistance in humans. Diabetologia 50:2602–2603PubMed Tschritter O, Preissl H, Yokoyama Y, Machicao F, Häring HU, Fritsche A (2007) Variation in the FTO gene locus is associated with cerebrocortical insulin resistance in humans. Diabetologia 50:2602–2603PubMed
112.
Zurück zum Zitat Wåhlén K, Sjölin E, Hoffstedt J (2008) The common rs9939609 gene variant of the fat mass- and obesity-associated gene FTO is related to fat cell lipolysis. J Lipid Res 49:607–611PubMed Wåhlén K, Sjölin E, Hoffstedt J (2008) The common rs9939609 gene variant of the fat mass- and obesity-associated gene FTO is related to fat cell lipolysis. J Lipid Res 49:607–611PubMed
113.
Zurück zum Zitat Gerken T, Girard CA, Tung YC, Webby CJ, Saudek V, Hewitson KS, Yeo GS, McDonough MA, Cunliffe S, McNeill LA, Galvanovskis J, Rorsman P, Robins P, Prieur X, Coll AP, Ma M, Jovanovic Z, Farooqi IS, Sedgwick B, Barroso I, Lindahl T, Ponting CP, Ashcroft FM, O’Rahilly S, Schofield CJ (2007) The obesity-associated FTO gene encodes a 2-oxoglutarate-dependent nucleic acid demethylase. Science 318:1469–1472PubMed Gerken T, Girard CA, Tung YC, Webby CJ, Saudek V, Hewitson KS, Yeo GS, McDonough MA, Cunliffe S, McNeill LA, Galvanovskis J, Rorsman P, Robins P, Prieur X, Coll AP, Ma M, Jovanovic Z, Farooqi IS, Sedgwick B, Barroso I, Lindahl T, Ponting CP, Ashcroft FM, O’Rahilly S, Schofield CJ (2007) The obesity-associated FTO gene encodes a 2-oxoglutarate-dependent nucleic acid demethylase. Science 318:1469–1472PubMed
114.
Zurück zum Zitat Sanchez-Pulido L, Andrade-Navarro MA (2007) The FTO (fat mass and obesity associated) gene codes for a novel member of the non-heme dioxygenase superfamily. BMC Biochem 8:23PubMed Sanchez-Pulido L, Andrade-Navarro MA (2007) The FTO (fat mass and obesity associated) gene codes for a novel member of the non-heme dioxygenase superfamily. BMC Biochem 8:23PubMed
115.
Zurück zum Zitat Jia G, Yang CG, Yang S, Jian X, Yi C, Zhou Z, He C (2008) Oxidative demethylation of 3-methylthymine and 3-methyluracil in single-stranded DNA and RNA by mouse and human FTO. FEBS Lett 582:3313–3319PubMed Jia G, Yang CG, Yang S, Jian X, Yi C, Zhou Z, He C (2008) Oxidative demethylation of 3-methylthymine and 3-methyluracil in single-stranded DNA and RNA by mouse and human FTO. FEBS Lett 582:3313–3319PubMed
116.
Zurück zum Zitat Stratigopoulos G, Padilla SL, LeDuc CA, Watson E, Hattersley AT, McCarthy MI, Zeltser LM, Chung WK, Leibel RL (2008) Regulation of Fto/Ftm gene expression in mice and humans. Am J Physiol Regul Integr Comp Physiol 294:R1185–R1196PubMed Stratigopoulos G, Padilla SL, LeDuc CA, Watson E, Hattersley AT, McCarthy MI, Zeltser LM, Chung WK, Leibel RL (2008) Regulation of Fto/Ftm gene expression in mice and humans. Am J Physiol Regul Integr Comp Physiol 294:R1185–R1196PubMed
117.
Zurück zum Zitat Boissel S, Reish O, Proulx K, Kawagoe-Takaki H, Sedgwick B, Yeo GS, Meyre D, Golzio C, Molinari F, Kadhom N, Etchevers HC, Saudek V, Farooqi IS, Froguel P, Lindahl T, O’Rahilly S, Munnich A, Colleaux L (2009) Loss-of-function mutation in the dioxygenase-encoding FTO gene causes severe growth retardation and multiple malformations. Am J Hum Genet 85:106–111PubMed Boissel S, Reish O, Proulx K, Kawagoe-Takaki H, Sedgwick B, Yeo GS, Meyre D, Golzio C, Molinari F, Kadhom N, Etchevers HC, Saudek V, Farooqi IS, Froguel P, Lindahl T, O’Rahilly S, Munnich A, Colleaux L (2009) Loss-of-function mutation in the dioxygenase-encoding FTO gene causes severe growth retardation and multiple malformations. Am J Hum Genet 85:106–111PubMed
118.
Zurück zum Zitat Fischer J, Koch L, Emmerling C, Vierkotten J, Peters T, Brüning JC, Rüther U (2009) Inactivation of the Fto gene protects from obesity. Nature 458:894–898PubMed Fischer J, Koch L, Emmerling C, Vierkotten J, Peters T, Brüning JC, Rüther U (2009) Inactivation of the Fto gene protects from obesity. Nature 458:894–898PubMed
119.
Zurück zum Zitat Church C, Lee S, Bagg EA, McTaggart JS, Deacon R, Gerken T, Lee A, Moir L, Mecinović J, Quwailid MM, Schofield CJ, Ashcroft FM, Cox RD (2009) A mouse model for the metabolic effects of the human fat mass and obesity associated FTO gene. PLoS Genet 5:e1000599PubMed Church C, Lee S, Bagg EA, McTaggart JS, Deacon R, Gerken T, Lee A, Moir L, Mecinović J, Quwailid MM, Schofield CJ, Ashcroft FM, Cox RD (2009) A mouse model for the metabolic effects of the human fat mass and obesity associated FTO gene. PLoS Genet 5:e1000599PubMed
120.
Zurück zum Zitat Meyre D, Proulx K, Kawagoe-Takaki H, Vatin V, Gutiérrez-Aguilar R, Lyon D, Ma M, Choquet H, Horber F, Van Hul W, Van Gaal L, Balkau B, Visvikis-Siest S, Pattou F, Farooqi IS, Saudek V, O’Rahilly S, Froguel P, Sedgwick B, Yeo GS (2010) Prevalence of loss of function FTO mutations in lean and obese individuals. Diabetes 59:311–318PubMed Meyre D, Proulx K, Kawagoe-Takaki H, Vatin V, Gutiérrez-Aguilar R, Lyon D, Ma M, Choquet H, Horber F, Van Hul W, Van Gaal L, Balkau B, Visvikis-Siest S, Pattou F, Farooqi IS, Saudek V, O’Rahilly S, Froguel P, Sedgwick B, Yeo GS (2010) Prevalence of loss of function FTO mutations in lean and obese individuals. Diabetes 59:311–318PubMed
121.
Zurück zum Zitat Herbert A, Gerry NP, McQueen MB, Heid IM, Pfeufer A, Illig T, Wichmann HE, Meitinger T, Hunter D, Hu FB, Colditz G, Hinney A, Hebebrand J, Koberwitz K, Zhu X, Cooper R, Ardlie K, Lyon H, Hirschhorn JN, Laird NM, Lenburg ME, Lange C, Christman MF (2006) A common genetic variant is associated with adult and childhood obesity. Science 312:279–283PubMed Herbert A, Gerry NP, McQueen MB, Heid IM, Pfeufer A, Illig T, Wichmann HE, Meitinger T, Hunter D, Hu FB, Colditz G, Hinney A, Hebebrand J, Koberwitz K, Zhu X, Cooper R, Ardlie K, Lyon H, Hirschhorn JN, Laird NM, Lenburg ME, Lange C, Christman MF (2006) A common genetic variant is associated with adult and childhood obesity. Science 312:279–283PubMed
122.
Zurück zum Zitat Dina C, Meyre D, Samson C, Tichet J, Marre M, Jouret B, Charles MA, Balkau B, Froguel P (2007) Comment on ‘A common genetic variant is associated with adult and childhood obesity’. Science 315:187PubMed Dina C, Meyre D, Samson C, Tichet J, Marre M, Jouret B, Charles MA, Balkau B, Froguel P (2007) Comment on ‘A common genetic variant is associated with adult and childhood obesity’. Science 315:187PubMed
123.
Zurück zum Zitat Loos RJ, Barroso I, O’Rahilly S, Wareham NJ (2007) Comment on ‘A common genetic variant is associated with adult and childhood obesity’. Science 315:187PubMed Loos RJ, Barroso I, O’Rahilly S, Wareham NJ (2007) Comment on ‘A common genetic variant is associated with adult and childhood obesity’. Science 315:187PubMed
124.
Zurück zum Zitat Lyon HN, Emilsson V, Hinney A, Heid IM, Lasky-Su J, Zhu X, Thorleifsson G, Gunnarsdottir S, Walters GB, Thorsteinsdottir U, Kong A, Gulcher J, Nguyen TT, Scherag A, Pfeufer A, Meitinger T, Bronner G, Rief W, Soto-Quiros ME, Avila L, Klanderman B, Raby BA, Silverman EK, Weiss ST, Laird N, Ding X, Groop L, Tuomi T, Isomaa B, Bengtsson K, Butler JL, Cooper RS, Fox CS, O’Donnell CJ, Vollmert C, Celedon JC, Wichmann HE, Hebebrand J, Stefansson K, Lange C, Hirschhorn JN (2007) The association of a SNP upstream of INSIG2 with body mass index is reproduced in several but not all cohorts. PLoS Genet 3:e61PubMed Lyon HN, Emilsson V, Hinney A, Heid IM, Lasky-Su J, Zhu X, Thorleifsson G, Gunnarsdottir S, Walters GB, Thorsteinsdottir U, Kong A, Gulcher J, Nguyen TT, Scherag A, Pfeufer A, Meitinger T, Bronner G, Rief W, Soto-Quiros ME, Avila L, Klanderman B, Raby BA, Silverman EK, Weiss ST, Laird N, Ding X, Groop L, Tuomi T, Isomaa B, Bengtsson K, Butler JL, Cooper RS, Fox CS, O’Donnell CJ, Vollmert C, Celedon JC, Wichmann HE, Hebebrand J, Stefansson K, Lange C, Hirschhorn JN (2007) The association of a SNP upstream of INSIG2 with body mass index is reproduced in several but not all cohorts. PLoS Genet 3:e61PubMed
125.
Zurück zum Zitat Rosskopf D, Bornhorst A, Rimmbach C, Schwahn C, Kayser A, Krüger A, Tessmann G, Geissler I, Kroemer HK, Völzke H (2007) Comment on ‘A common genetic variant is associated with adult and childhood obesity’. Science 315:187PubMed Rosskopf D, Bornhorst A, Rimmbach C, Schwahn C, Kayser A, Krüger A, Tessmann G, Geissler I, Kroemer HK, Völzke H (2007) Comment on ‘A common genetic variant is associated with adult and childhood obesity’. Science 315:187PubMed
126.
Zurück zum Zitat Meyre D, Delplanque J, Chèvre JC, Lecoeur C, Lobbens S, Gallina S, Durand E, Vatin V, Degraeve F, Proença C, Gaget S, Körner A, Kovacs P, Kiess W, Tichet J, Marre M, Hartikainen AL, Horber F, Potoczna N, Hercberg S, Levy-Marchal C, Pattou F, Heude B, Tauber M, McCarthy MI, Blakemore AI, Montpetit A, Polychronakos C, Weill J, Coin LJ, Asher J, Elliott P, Järvelin MR, Visvikis-Siest S, Balkau B, Sladek R, Balding D, Walley A, Dina C, Froguel P (2009) Genome-wide association study for early-onset and morbid adult obesity identifies three new risk loci in European populations. Nat Genet 41:157–159PubMed Meyre D, Delplanque J, Chèvre JC, Lecoeur C, Lobbens S, Gallina S, Durand E, Vatin V, Degraeve F, Proença C, Gaget S, Körner A, Kovacs P, Kiess W, Tichet J, Marre M, Hartikainen AL, Horber F, Potoczna N, Hercberg S, Levy-Marchal C, Pattou F, Heude B, Tauber M, McCarthy MI, Blakemore AI, Montpetit A, Polychronakos C, Weill J, Coin LJ, Asher J, Elliott P, Järvelin MR, Visvikis-Siest S, Balkau B, Sladek R, Balding D, Walley A, Dina C, Froguel P (2009) Genome-wide association study for early-onset and morbid adult obesity identifies three new risk loci in European populations. Nat Genet 41:157–159PubMed
127.
Zurück zum Zitat Heid IM, Huth C, Loos RJ, Kronenberg F, Adamkova V, Anand SS, Ardlie K, Biebermann H, Bjerregaard P, Boeing H, Bouchard C, Ciullo M, Cooper JA, Corella D, Dina C, Engert JC, Fisher E, Francès F, Froguel P, Hebebrand J, Hegele RA, Hinney A, Hoehe MR, Hu FB, Hubacek JA, Humphries SE, Hunt SC, Illig T, Järvelin MR, Kaakinen M, Kollerits B, Krude H, Kumar J, Lange LA, Langer B, Li S, Luchner A, Lyon HN, Meyre D, Mohlke KL, Mooser V, Nebel A, Nguyen TT, Paulweber B, Perusse L, Qi L, Rankinen T, Rosskopf D, Schreiber S, Sengupta S, Sorice R, Suk A, Thorleifsson G, Thorsteinsdottir U, Völzke H, Vimaleswaran KS, Wareham NJ, Waterworth D, Yusuf S, Lindgren C, McCarthy MI, Lange C, Hirschhorn JN, Laird N, Wichmann HE (2009) Meta-analysis of the INSIG2 association with obesity including 74, 345 individuals: does heterogeneity of estimates relate to study design? PLoS Genet 5(10):e1000694PubMed Heid IM, Huth C, Loos RJ, Kronenberg F, Adamkova V, Anand SS, Ardlie K, Biebermann H, Bjerregaard P, Boeing H, Bouchard C, Ciullo M, Cooper JA, Corella D, Dina C, Engert JC, Fisher E, Francès F, Froguel P, Hebebrand J, Hegele RA, Hinney A, Hoehe MR, Hu FB, Hubacek JA, Humphries SE, Hunt SC, Illig T, Järvelin MR, Kaakinen M, Kollerits B, Krude H, Kumar J, Lange LA, Langer B, Li S, Luchner A, Lyon HN, Meyre D, Mohlke KL, Mooser V, Nebel A, Nguyen TT, Paulweber B, Perusse L, Qi L, Rankinen T, Rosskopf D, Schreiber S, Sengupta S, Sorice R, Suk A, Thorleifsson G, Thorsteinsdottir U, Völzke H, Vimaleswaran KS, Wareham NJ, Waterworth D, Yusuf S, Lindgren C, McCarthy MI, Lange C, Hirschhorn JN, Laird N, Wichmann HE (2009) Meta-analysis of the INSIG2 association with obesity including 74, 345 individuals: does heterogeneity of estimates relate to study design? PLoS Genet 5(10):e1000694PubMed
128.
Zurück zum Zitat Sha BY, Yang TL, Zhao LJ, Chen XD, Guo Y, Chen Y, Pan F, Zhang ZX, Dong SS, Xu XH, Deng HW (2009) Genome-wide association study suggested copy number variation may be associated with body mass index in the Chinese population. J Hum Genet 54:199–202PubMed Sha BY, Yang TL, Zhao LJ, Chen XD, Guo Y, Chen Y, Pan F, Zhang ZX, Dong SS, Xu XH, Deng HW (2009) Genome-wide association study suggested copy number variation may be associated with body mass index in the Chinese population. J Hum Genet 54:199–202PubMed
129.
Zurück zum Zitat Conrad DF, Pinto D, Redon R, Feuk L, Gokcumen O, Zhang Y, Aerts J, Andrews TD, Barnes C, Campbell P, Fitzgerald T, Hu M, Ihm CH, Kristiansson K, Macarthur DG, Macdonald JR, Onyiah I, Pang AW, Robson S, Stirrups K, Valsesia A, Walter K, Wei J; The Wellcome Trust Case Control Consortium, Tyler-Smith C, Carter NP, Lee C, Scherer SW, Hurles ME (2009) Origins and functional impact of copy number variation in the human genome. Nature. doi:10.1038/nature08516. Conrad DF, Pinto D, Redon R, Feuk L, Gokcumen O, Zhang Y, Aerts J, Andrews TD, Barnes C, Campbell P, Fitzgerald T, Hu M, Ihm CH, Kristiansson K, Macarthur DG, Macdonald JR, Onyiah I, Pang AW, Robson S, Stirrups K, Valsesia A, Walter K, Wei J; The Wellcome Trust Case Control Consortium, Tyler-Smith C, Carter NP, Lee C, Scherer SW, Hurles ME (2009) Origins and functional impact of copy number variation in the human genome. Nature. doi:10.​1038/​nature08516.
130.
Zurück zum Zitat Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, McCarthy MI, Ramos EM, Cardon LR, Chakravarti A, Cho JH, Guttmacher AE, Kong A, Kruglyak L, Mardis E, Rotimi CN, Slatkin M, Valle D, Whittemore AS, Boehnke M, Clark AG, Eichler EE, Gibson G, Haines JL, Mackay TF, McCarroll SA, Visscher PM (2009) Finding the missing heritability of complex diseases. Nature 461:747–753PubMed Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, McCarthy MI, Ramos EM, Cardon LR, Chakravarti A, Cho JH, Guttmacher AE, Kong A, Kruglyak L, Mardis E, Rotimi CN, Slatkin M, Valle D, Whittemore AS, Boehnke M, Clark AG, Eichler EE, Gibson G, Haines JL, Mackay TF, McCarroll SA, Visscher PM (2009) Finding the missing heritability of complex diseases. Nature 461:747–753PubMed
Metadaten
Titel
From monogenic to polygenic obesity: recent advances
verfasst von
Anke Hinney
Carla I. G. Vogel
Johannes Hebebrand
Publikationsdatum
01.03.2010
Verlag
Springer-Verlag
Erschienen in
European Child & Adolescent Psychiatry / Ausgabe 3/2010
Print ISSN: 1018-8827
Elektronische ISSN: 1435-165X
DOI
https://doi.org/10.1007/s00787-010-0096-6

Weitere Artikel der Ausgabe 3/2010

European Child & Adolescent Psychiatry 3/2010 Zur Ausgabe

Update Psychiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.