Skip to main content
Current Genomics logoLink to Current Genomics
. 2011 May;12(3):154–168. doi: 10.2174/138920211795677921

Molecular Basis of Obesity: Current Status and Future Prospects

Hélène Choquet 1, David Meyre 2,*
PMCID: PMC3137001  PMID: 22043164

Abstract

Obesity is a global health problem that is gradually affecting each continent of the world. Obesity is a heterogeneous disorder, and the biological causes of obesity are complex. The rapid increase in obesity prevalence during the past few decades is due to major societal changes (sedentary lifestyle, over-nutrition) but who becomes obese at the individual level is determined to a great extent by genetic susceptibility. In this review, we evidence that obesity is a strongly heritable disorder, and provide an update on the molecular basis of obesity. To date, nine loci have been involved in Mendelian forms of obesity and 58 loci contribute to polygenic obesity, and rare and common structural variants have been reliably associated with obesity. Most of the obesity genes remain to be discovered, but promising technologies, methodologies and the use of “deep phenotyping” lead to optimism to chip away at the ‘missing heritability’ of obesity in the near future. In the longer term, the genetic dissection of obesity will help to characterize disease mechanisms, provide new targets for drug design, and lead to an early diagnosis, treatment, and prevention of obesity.

Keywords: Candidate gene, childhood obesity, copy number variation, gwas, heritability, linkage, monogenic, polygenic.

INTRODUCTION

Since 1980, the mean body mass index (BMI) worldwide increased by 0.4-0.5 kg/m² per decade in adults 20 years and older with 502 million adults worldwide classified as obese (BMI ≥ 30 kg/m²) by 2008 [1]. The worldwide prevalence of childhood overweight and obesity have increased from 4.2% in 1990 to 6.7% in 2010 and this trend is expected to reach 9.1% in 2020 [2]. This global health problem is gradually affecting each continent of the world.

High rates of weight gain during infancy may increase a person’s later risk of obesity. There are growing numbers of obese children developing diseases formerly considered to be an “adult” condition, such as type 2 diabetes, nonalcoholic fatty liver disease, sleep apnea and hypertension [3]. Obesity-associated diseases are now believed to lead to a shortened lifespan [4]. As the incidence of obesity-related diseases increase among adults as well as children, the consequences of the obesity epidemic on the economy of developed countries should be considered as a major priority [5].

The environmental causes of childhood obesity involve an unhealthy diet and physical activity patterns as well as early-life factors such as diabetes exposure in utero, larger size for gestational age, shorter breastfeeding duration and more rapid infant weight gain [6]. However, obesity is the result not only of several environmental risk factors, but also genetic predisposition [7]. Understanding the many causal factors leading to the complex trait of obesity may help develop more targeted and effective therapies.

PROGRESS IN DEFINING THE MOLECULAR BASIS OF OBESITY

Obesity is a Heritable Disorder

Family history of obesity is a well-established risk predictor for obesity in childhood. The risk in a child is 2.5-4-fold higher if one of their parents is obese and 10-fold higher if both parents are obese, compared to having both parents of normal weight [8]. The familial risk for obesity (the risk ratio to be obese for an individual if a first degree relative is obese compared with individuals in the population who have only normal-weight first degree relatives) is comprised between 1.5 and 5 depending on the severity of obesity [9]. The familial aggregation of individual’s body size is not of a recent concept: Sir Francis Galton mentioned this observation in his book “Natural Inheritance” in 1889 [10]. However, as familial resemblance can be explained by shared environments as well as genetic factors, the specific influence of genes in determining early-onset obesity was investigated 35 years ago with the emergence of twin and family studies.

In 1977, Feinleib and colleagues [11] studied the correlations for weight in 250 monozygotic (100% of genome shared) and 264 dizygotic (50% of genome shared) male veteran twin pairs and established for the first time that familial aggregation for obesity results mainly from genetic influence. These results were confirmed in more than 4,000 monozygotic and dizygotic twin pairs in 1986. Strong heritability values for body mass index (BMI) were observed for the same subjects at 20 years (h²= 0.77) and at 45 years (h²=0.84). Heritability represents the proportion of phenotypic variation in a population that is attributable to genetic variation among individuals [7]. Adoption studies have strengthened the evidence of a strong genetic influence on human body weight. Body corpulence of adopted children was shown to strongly correlate with BMI of their biologic parents, whereas no correlation was observed with the BMI of their adoptive parents [12]. Furthermore, correlation values for BMI of 0.66-0.7 were estimated by Stunkard et al. [13] for monozygotic twins reared apart, 0.66-0.74 for monozygotic twins reared together, 0.15-0.25 for dizygotic twins reared apart and 0.27-0.33 for dizygotic twins reared together suggesting that genetic influences on body-mass index are substantial, whereas the family environment alone has minor influence. Many twin and family studies including children or adolescents have been published since these pioneering observations, providing heritability values for BMI between 0.20 and 0.86 [14-17].

Longitudinal studies have demonstrated that heritability estimates tend to increase from infancy to childhood [18], from childhood to pre-adolescence [15] and from pre-adolescence to adolescence [17], mirroring the growing exposure to obesity-prone environments that subjects with genetic propensities tend to favor. Heritability studies have also shown that longitudinal BMI change from adolescence to young adulthood is an heritable trait and that genetic factors modulating BMI levels are only partially shared with those modulating change in BMI across time, supporting a complex etiology of BMI and BMI change [19]. Heritability of obesity as a binary trait, as well as for BMI in all ranges of the population, involved overall equates to the same set of genes [15].

Heritability values for BMI / obesity can be modified by specific environmental exposures. A high level of physical activity can substantially reduce the influence of genetic factors on BMI in both young adults and older adults [20, 21]. Even if heritability estimations of BMI are similar in sex-specific analyses, some sets of genes explaining the BMI variation may be different in adolescent as well in adult males and females [14, 22]. Ethnicity may also interact with the genetic predisposition to obesity and ethnic-specific genetic factors are likely to modulate human body weight. The proportion of European genome in admixed African or Indian Native populations is inversely correlated with BMI, supporting the idea that specific gene subsets may account for genetic susceptibility to obesity in different ethnic backgrounds [23, 24]. Surprisingly, the major societal changes that accompanied the transition to an “obesogenic” environment (environments that promote gaining weight) did not have a major impact on the genetic predisposition to obesity. The values of heritability for BMI in childhood remain high (h²=0.77) even in the current “obesogenic” environment [16], and many of the same genes seem to be involved in establishing genetic susceptibility to obesity in the pre- as in the post-obesity epidemic period [25]. Heritability estimates for obesity-related traits are consistent among Nigerian, Jamaican and US black people despite contrasted environmental conditions and different obesity prevalence (5% in Nigeria, 23% in Jamaica and 39% in the USA) [26].

Beyond BMI, heritability studies have also been conducted on endophenotypes related to obesity in young individuals. Strong genetic influences have been evidenced for intermediate traits such as percentage of body fat [27], waist circumference [28], eating behaviour [29], level of physical activity [30] or energy expenditure [31].

Mendelian Forms of Non-Syndromic Obesity

Monogenic forms of obesity refer to a single gene disorder leading to a highly penetrant form of the disease. The study of extreme human obesity caused by a single gene defects has provided a glimpse into the long-term regulation of body weight. For example, defects in eight genes involved in the neuronal differentiation of the paraventricular nucleus and in the leptin/melanocortin pathway lead to monogenic forms of early-onset severe obesity, demonstrating the critical role of the leptin-melanocortin system critical for energy balance in humans [32]. The eight genes are leptin (LEP), leptin receptor (LEPR), proopiomelanocortin (POMC), prohormone convertase 1 (PCSK1), melanocortin 4 receptor (MC4R), single-minded homolog 1 (SIM1), brain-derived neurotrophic factor (BDNF) and its receptor TrkB coded by the neurotrophic tyrosine kinase receptor type 2 gene (NTRK2).

Recessive Forms of Monogenic Obesity

Recessive forms of obesity caused by homozygous / heterozygous compound loss of function mutations in five genes (LEP, LEPR, POMC, PCSK1 and MC4R) have been reported to date [33-37]. Complete inactivation of these five genes invariably results in severe hyperphagia and fully penetrant form of early-onset extreme obesity in humans. Additional phenotypic features associated with these recessive forms of obesity are more specific: individuals with congenital leptin receptor or leptin deficiency present alterations in immune function and frequent childhood infections of the respiratory tract associated with high rates of premature death. They also manifest delayed puberty due to hypogonadotropic hypogonadism, and hypothyroidism [33, 34]. The clinical features of individuals with congenital leptin receptor deficiency are usually less severe than those with congenital homozygous leptin deficiency, and hypothyroidism is less common [38]. Individuals with complete POMC deficiency present in their early life with hypoadrenalism secondary to ACTH deficiency and develop hypoglycemia, jaundice and neonatal death which is associated with severe liver cholestasis. POMC-deficient individuals of European ancestry have pale skin and red hair [35], but those with congenital POMC deficiency can harbor normal hair and skin pigmentation in other ethnic backgrounds [39]. Individuals with complete PCSK1 deficiency harbor reactive hypoglycemia and perturbations in endocrine function including episodes of severe diarrhea which indicate an important role for the PC1/3 enzyme in enteroendocrine cells [36, 40]. Individuals with complete MC4R deficiency harbor an increased lean mass, increased bone mineral density and tall stature [37].

The study of recessive forms of monogenic extreme obesity has been useful in delineating the role of genes from the leptin / melanocortin pathway in human physiology but explains a small percentage of obesity in the population. Indeed these recessive forms of obesity are excessively rare and are mostly identified in families with a high level of consanguinity (consanguinity increases the probability to carry two deleterious copies of the same gene). Thus far, 14 individuals with complete LEP deficiency have been identified worldwide, 13 subjects with complete LEPR deficiency, seven with complete POMC deficiency, three with complete PCSK1 deficiency and 20 with complete MC4R deficiency.

Partial Gene Deficiency and Obesity Features

Heterozygosity for deleterious coding mutations in MC4R [41] or POMC [42] has been associated with a non fully penetrant form of obesity, whereas partial LEP or LEPR deficiency has been associated with a higher percentage of body fat mass [38, 43]. On the opposite side, the eight heterozygous carriers of PCSK1 loss of function mutations reported to date, have been described as clinically unaffected [44]. This result is intriguing, since heterozygous PC1N222D/+ mice present with an obesity-intermediary phenotype [45] and heterozygotes PC1-null mice tend to be mildly obese [46]. Even if heterozygous carriers of deleterious mutations in MC4R, POMC, LEP and LEPR present a milder and incompletely penetrant form of obesity in comparison with the obligatory severe obesity phenotype caused by homozygous / heterozygous compound loss of function mutations in the same genes, they are likely to contribute for a non-negligible fraction of obesity at the population level.

MC4R deficiency is the common cause of monogenic obesity. Starting from a MC4R loss of function mutation frequency of 0.07% reported in the general populations [47], we can expect 426,701 heterozygous carriers and only 149 homozygous / compound heterozygous carriers in the US population (N=305,000,000). Using an average penetrance of 60% for heterozygous carriers and 100% for homozygous and compound heterozygous carriers, as previously reported in literature [41], partial MC4R deficiency may explain obesity in 256,021 individuals, whereas complete MC4R deficiency may be the cause of obesity for only 149 subjects in the US population.

Haplo-insufficiency for BDNF, TrkB and SIM1 has been associated with severe hyperphagic obesity, accompanied by syndromic features in humans [48-51]. BDNF and its receptor TrkB are involved in proliferation, survival, and differentiation of neurons during development and post-natal synaptic plasticity in the central nervous system, especially in hypothalamic neurons. SIM1, the mammalian homologue of Drosophila sim, is a transcription factor playing a major role in neuronal differentiation of the paraventricular nucleus of the hypothalamus, a critical brain region for food intake regulation. No human case of complete deficiency for BDNF, TrkB and SIM1 are reported in literature. This is consistent with the phenotypes observed in mice. Partial deficiency of Bdnf, TrkB or Sim1 in mice induces hyperphagia, obesity and developmental features [52-54], whereas complete deficiency for Bdnf or Sim1 is lethal [52, 53] and complete deficiency for TrkB dramatically reduces life span [55].

Genome Structural Variations

In the last few years, the advent of genome-scanning technologies has led to the discovery that genetic differences among people can derive from lost or duplicated segments of chromosomes, called copy number variants (CNVs) or structural variants [56]. CNVs contribute significantly to the genetic architecture of human obesity. Rare deletions in the region p11.2 of the chromosome 16 have been reported in about 0.5-0.7% of individuals with severe obesity in two independent studies [57, 58], and the link between deletions at 16p11.2 and obesity has been confirmed in three follow-up reports [59-61]. The 16p11.2 deletion interval identified in these studies encompasses about 30 genes. The SH2B adapter protein 1 (SH2B1) is one of these genes and is an excellent candidate gene to link the 16p11.2 deletion to obesity. Indeed, SH2B1 modulates leptin sensitivity and Sh2b1 knock-out mice develop hyperphagia and obesity [62]. In addition, the SH2B1 locus was recently associated with common obesity by genome-wide association studies (GWAS) [63, 64].

A recent study identified 17 rare CNV loci previously unreported in the public domain and only found in obese but not in lean children of European ancestry. Eight out of 17 CNVs were also found in obese children of African ancestry, but not in lean control subjects of the same ethnicity [65]. Finally, Wang et al. [60] investigated the potential role of large and rare CNVs in moderate and extreme obesity and demonstrated that rare CNVs > 2 Mb were present in 1.3% of obese subjects but were absent in lean controls. Several CNVs disrupt known candidate genes for obesity, such as NAP1L5, UCP1 and IL15 [60]. The examination of rare CNVs offers novel insights into the genetic architecture of obesity.

Polygenic Forms of Obesity

The search for gene variants associated with polygenic forms of obesity is based on the common disease-common variant hypothesis. This hypothesis states that multiple common, interacting disease alleles contribute to common diseases (each variant at each gene having a modest effect on the disease phenotype) and are well represented in human populations [66]. Three main approaches have been used to identify novel gene variants associated with polygenic obesity: candidate gene, genome-wide linkage and genome-wide association studies.

Candidate Gene Studies

Association studies with candidate genes have been widely used for the study of complex traits but have proven largely unsuccessful. Several hundreds of obesity candidate genes have been selected for genetic association study on the biological, physiological or pharmacological evidence of their role in body weight regulation, but most of the positive association signals initially reported have never been consistently replicated in follow-up replication studies [67]. Large meta-analyses have been reported for only a small number of variants, and so far only two genes have been reliably associated with obesity phenotypes using this approach.

Targeted disruption of Mc4r results in obesity in mice, loss-of-function coding mutations in MC4R lead to the more common form of monogenic human obesity [41, 68] and significant evidence of linkage for obesity-related traits has been reported on chromosome 18, where the MC4R gene resides [69]. MC4R is therefore a highly relevant candidate gene for common obesity. Two gain-of-function [70] infrequent coding polymorphisms (I251L and V103I) in MC4R have been negatively associated with obesity [71, 72]. The 103I variant was associated with a 20% lower risk for obesity in a meta-analysis of 39,879 subjects of European ancestry [72] and the 103I allele carriers had a 31% lower risk for obesity in a meta-analysis of 3,526 individuals from six East Asian studies [73]. The 251L allele was associated with a 50% lower risk for obesity in a meta-analysis of 11,435 European subjects [72].

Loss-of-function rare mutations in the PCSK1 gene have been associated with obesity both in humans and rodents [36, 45]. In addition, four independent genome-wide linkage studies for obesity-related traits delineate a common 5.6-Mb interval on chromosome 5q where PCSK1 resides [74]. We recently showed that the relatively infrequent (minor allele frequency: 5%) single nucleotide polymorphism (SNP) N221D induces a 10.4% significant reduction of PC1/3 catalytic activity, and is associated with a 34% increased risk for obesity in European populations [74]. The N221D variant was also associated with BMI level in an independent sample of 32,000 European subjects [64]. These two examples indicate that the candidate gene approach can be successful if: 1) stringent criteria are applied to gene and SNP selection process and 2) large scale association studies are performed.

Genome-Wide Linkage Studies

Genome-wide linkage scans involve the genotyping of families recruited for the high recurrence of a disease using highly polymorphic microsatellite markers that are regularly spaced across the whole genome, followed by a calculation of the degree of linkage of the marker to a disease trait. Genome-wide linkage approaches led to the successful identification of > 1,200 genes involved in Mendelian human diseases, however the application of genome-wide linkage in the analysis of complex genetic traits has been more controversial [75]. More than 80 linkage studies for obesity-related traits have been reported, showing significant evidence for linkage (LOD-score > 4) for some of them [76, 77]. However, a meta-analysis of 37 published studies containing data on over 31,000 individuals was unable to confirm a major locus for obesity [78], as observed in meta-analyses for other complex diseases [79].

Possible explanations may include genes influencing adiposity are of very small effect, with substantial genetic heterogeneity and variable dependence on environmental factors, but this does not explain why significant peaks of linkage have been reported in individual studies at many different chromosomal locations. A plausible alternative explanation is that rare variants with high disease penetrance may be a common explanation for linkage peaks observed in complex traits like obesity [42, 80]. This is in line with statistical simulations predicting that odds-ratios must be high (OR > 2) to induce significant peaks of linkage in modest family sample sets [75]. Attempts to identify new obesity genes by genome-wide linkage strategies have been mostly unsuccessful, genetic variants in only two genes have shown association with obesity, contribution to the initial linkage peak and have been confirmed in subsequent replication studies.

The only significant evidence of linkage for childhood obesity was obtained on chromosome 6q22.31-q23.2 in 115 French pedigrees [76]. Restriction of the linkage interval under study by comparison of eight independent linkage studies and subsequent positional candidate gene approach led to the identification of a three-allele risk haplotype (K121Q, IVS20delT-11, A→G +1044TGA; QdelTG) in the ectonucleotide pyrophosphatase / phosphodiesterase 1 (ENPP1) gene that showed association with childhood obesity and contribution to the observed linkage with childhood obesity [81]. The ENPP1 QdelTG haplotype was associated with adult moderate and morbid obesity and type 2 diabetes and was also associated with increased serum levels of soluble ENPP1 protein in children [81]. Interestingly, ENPP1 gene variation was found to be independently associated with obesity-related traits in Mexican American pedigrees by a positional candidate gene approach [82]. The function of the gene can be directly related to obesity and type 2 diabetes as ENPP1 inhibits insulin receptor signaling [83]. The link between ENPP1 and insulin resistance has been evidenced in genetic mouse models as well [84]. Insulin resistance in brain induces hyperphagia and obesity in mice [85] and insulin resistance is a strong predictor of subsequent development of obesity in children [86]. The association of the ENPP1 risk haplotype with childhood obesity has been replicated in a German study [87] but further replication is needed to provide an unequivocal confirmation.

In 2002, Stone and colleagues [77] identified a major predisposition locus for sever obesity in White American females on chromosome 4p15-p14. Subsequent positional cloning effort by the same team led to the identification of a non-synonymous polymorphism (R125W) in the TBC1D1 gene associated with severe familial obesity in women only and accounting for the majority of the evidence of linkage on chromosome 4p15-14 [88]. The association of the TBC1D1 R125W polymorphism with familial severe obesity in females and linkage at 4p15-p14 was replicated in an independent French cohort [89]. Further replication is now needed at this stage, and the gender-specific association of the R125W variant with obesity remains unexplained. TBC1D1 protein function involves adipogenesis process [90], insulin signalling [91] and lipid use in skeletal muscle [92] and inactivation of Tbc1d1 confers leanness in mice [92]. These observations further support the candidacy of TBC1D1 as a ‘thrifty’ gene as recently proposed by Koumanov et al. [93].

Genome-Wide Association Studies

Genome-wide association studies (GWAS) are a relatively new way for scientists to identify genes involved in human diseases and have revolutionized the search for genetic common variants contributing to complex diseases. This method interrogates the genome for several hundred-thousand single nucleotide polymorphisms (SNPs) across the genome, and identifies the SNPs that occur more frequently in people with a particular disease than in those without the disease. As of June 2010, there have been more than 900 published genome-wide associations for 165 traits (http://www.genome.gov). The combinations of three major breakthroughs have made genome-wide comprehensive association studies possible: 1) the emergence of the notion of linkage disequilibrium block [94], the determination of the human genome SNP map through the International Hapmap Consortium [95] (this led to the conclusion that 80% of the common genetic variation (>14 million variants) in subjects of European ancestry can be captured by genotyping 300,000 carefully selected single nucleotide polymorphisms (SNPs) [96]); 2) the development and commercialization of new methods for high throughput genotyping using SNP microarrays [97] and 3) the recruitment of large scale case control and population-based cohorts with both DNA and phenotypes available [98].

In 2007, four different approaches led to the identification of variation in the intron 1 of Fat mass and obesity associated (FTO) gene as the major contributor to polygenic obesity in populations of European ancestry [99-102]. Frayling et al. [99] identified FTO through a GWAS for type 2 diabetes in UK subjects, variants in the FTO gene showing a strong association with T2D mediated through BMI [99]. Scuteri et al. [101] identified FTO by using a GWAS approach for BMI in the genetically isolated population of Sardinia. Hinney et al. [102] identified FTO by using GWAS for early-onset extreme obesity in a German case control study. Dina et al. [100] unexpectedly found a strong association between FTO and obesity, by performing a population structure approach with a set of 48 SNPs in a French obesity case control design. The 16% of adults of European ancestry who were homozygous for the risk allele weighed about 3 kilograms more and had 1.67-fold increased odds of obesity when compared with those not inheriting a risk allele [99]. Apart from European populations, FTO was shown to be the top association signal for BMI (P=1 x 10-7) in a recent GWAS including 8,842 Korean subjects [103].

Four large GWAS meta-analyses in general populations of European descent (16,876 < N < 123,865) confirmed the strong association of the FTO locus with BMI and identified 35 additional SNPs in 33 loci robustly (P < 5 x 10-7) associated with BMI (Table 1). Multiple independent association signals were reported at the FTO, MC4R and BDNF loci [63, 104]. Altogether these loci only explained 1.45% of the variance in BMI (0.34% explained by the SNP in intron 1 of FTO alone), suggesting that many additional common genetic variants associated with BMI remain to be discovered [104]. Each additional risk allele increases BMI of 0.17 kg/m² and body weight of 435-551 grams in adults of 160-180 cm in height [104]. Interestingly, SNPs that modulate BMI in general adult populations are also associated with BMI in pediatric populations [105], and are associated with an increased risk for early-onset [64, 104] or adult obesity [106].

Table 1.

Sixty-One Common Gene Variants at 58 Loci are Associated with Obesity Phenotypes at a Genome-Wide Level of Significance (P < 5 x 108)

Nearest Gene Polymorphism Phenotype Reference
FTO rs1421085 / rs9939609 body mass index, extreme obesity [99, 100]
MC4R rs17782313 body mass index, extreme and childhood obesity [107, 109, 130]
PCSK1 rs6232 extreme obesity [74]
PCSK1 rs6234 / rs6235 extreme obesity [74]
CTNNBL1 rs6013029 body mass index [139]
TMEM18 rs6548238 body mass index, childhood obesity [64, 109]
GNPDA2 rs10938397 body mass index [64]
SH2B1 rs7498665 body mass index [64]
KCTD15 rs11084753 body mass index [64]
MTCH2 rs10838738 body mass index [64]
NEGR1 rs2815752 body mass index [64]
NPC1 rs1805081 extreme obesity [107]
MAF rs1424233 extreme obesity [107]
PTER rs10598503 extreme obesity [107]
PRL rs4712652 extreme obesity [107]
SEC16B rs10913469 body mass index [63]
ETV5 rs7647305 body mass index [63]
AIF1 rs2844479 body mass index [63]
BDNF rs6265 body mass index [63]
BDNF rs925946 body mass index [63]
FAIM2 rs7138803 body mass index [63]
FTO rs6499640 body mass index [63]
SDCCAG8 rs12145833 childhood obesity [109]
TNKS rs17150703 childhood obesity [109]
TFAP2B rs987237 waist circumference, body mass index [104, 110]
MSRA rs7826222 waist circumference [110]
LYPLAL1 rs4846567 waist to hip ratio [110, 113]
NRXN3 rs10146997 waist circumference, body mass index [104, 112]
C12orf51 rs2074356 waist to hip ratio [103]
GPRC5BB rs12444979 body mass index [104]
POMC rs713586 body mass index [104]
MAP2K5 rs2241423 body mass index [104]
GIPR rs2287019 body mass index [104]
FANCL rs887912 body mass index [104]
TNNI3K rs1514175 body mass index [104]
LRRN6C rs10968576 body mass index [104]
FLJ35779 rs2112347 body mass index [104]
SLC39A8 rs13107325 body mass index [104]
TMEM160 rs3810291 body mass index [104]
CADM2 rs13078807 body mass index [104]
LRP1B rs2890652 body mass index [104]
PRKD1 rs11847697 body mass index [104]
MTIF3 rs4771122 body mass index [104]
ZNF608 rs48361333 body mass index [104]
PTBP2 rs1555543 body mass index [104]
TUB rs4929949 body mass index [104]
HMGA1 rs206936 body mass index [104]
MC4R rs7227255 body mass index [104]
RSPO3 rs9491696 waist to hip ratio [113]
VEGFA rs6905288 waist to hip ratio [113]
TBX15/WARS2 rs984222 waist to hip ratio [113]
NFE2L3 rs1055144 waist to hip ratio [113]
GRB14 rs10195252 waist to hip ratio [113]
DNM3/PIGC rs1011731 waist to hip ratio [113]
ITPR2/SSPN rs718314 waist to hip ratio [113]
LY86 rs1294421 waist to hip ratio [113]
HOXC13 rs1443512 waist to hip ratio [113]
ADAMTS9 rs6795735 waist to hip ratio [113]
ZNRF3/KREMEN1 rs4823006 waist to hip ratio [113]
NISCH/STAB1 rs6784615 waist to hip ratio [113]
CPEB4 rs6861681 waist to hip ratio [113]

The genetic architecture of extreme childhood and adult obesity has also been investigated using case control GWAS designs [102, 107-109]. Nine loci have been associated with extreme obesity at the genome-wide level, five of them influencing BMI / waist circumference as well as risk for extreme obesity (FTO, MC4R, TMEM18, MSRA, NPC1) [102, 104, 107-110], four loci being more specific of genetic risk for extreme obesity (MAF, PTER, PRL, SDCCAG8) [107, 109] (Table 1). Overall, these data indicate that the genetic architecture of BMI and extreme obesity are mostly overlapping in children as well as adults, and that extreme obesity may represent the extreme of the phenotypic spectrum of BMI rather than a distinct condition [108]. Several of the likely causal obesity genes identified through GWAS studies (FTO, MC4R, POMC, SH2B1, BDNF, NPC1, NRXN3 and NEGR1) are highly expressed or known to act in the central nervous system, indicating a key role for central regulation of food intake in obesity susceptibility in line with monogenic forms of human obesity [64, 104].

Beyond the study of BMI, four GWAS have investigated the genetic architecture of body fat distribution, estimated by waist circumference (WC) or waist to hip ratio (WHR) measurements [110-113]. Nineteen loci have been identified and five of them only are associated with BMI / obesity (FTO, MC4R, NRXN3, TFAP2B, MSRA) (Table 1). However, it is to noted that WC or WHR measurements were adjusted for BMI in the two larger GWAS meta-analyses, to identify SNPs associated with central adiposity and those independently of overall adiposity [110, 113]. The biological role of several likely causal candidate genes identified by GWAS pinpoint a key role of adipose tissue development and function in determining body fat distribution [113].

Common copy number variants (CNVs) are in high linkage disequilibrium with SNPs in the human genome suggesting that disease-associated common structural variants can be identified by SNP-based whole-genome association studies [114]. Using a SNP tagging approach, a 45-kb deletion near the NEGR1 gene and a 21-kb deletion 50 kb upstream of GPRC5BT have been recently and convincingly associated with BMI variation [64, 104], and a 2.8-kb common duplication 87 kb from the LY86 gene has been conclusively associated with WHR [113]. A common CNV on chromosomes 10p11.22 has been associated with BMI in a Chinese population [115] and covers four genes, one of them (PPYR1) being a plausible obesity candidate gene [115]. The association of CNV 10p11.22 with obesity has been confirmed in two independent studies [60, 61]. CNVs are plausible functional causal variants due to their potential impact on gene expression [116], and genes located nearby these CNVs might be prioritized for fine mapping and functional follow-up.

FUTURE PROSPECTS

New Approaches to Chip Away at the “Missing Heritability” of Obesity

Although there is strong evidence that heritability of human body weight is high, only a small fraction of the variance in BMI can be explained by the current list of genetic factors suggesting potential sources of missing heritability [104]. One hypothesis about missing heritability from GWAS has focused on the possible contribution of variants of low minor allele frequency (MAF), defined as 0.5<MAF<5 %, or of rare variants (MAF<0.5 %). GWA genotyping arrays used in the first waves of GWAS classically included 300,000-500,000 SNPs and they provided only an exhaustive coverage of the common SNP variation (MAF > 5%) in the genome [96], signifying that rare variants associated with complex diseases have not been captured in these studies. Low frequency variants may have much stronger effects on the disease risk than common variants without demonstrating clear Mendelian segregation, and may explain an important part of the missing heritability [117]. Common and rare structural variation, including CNVs, such as insertions and deletions and copy neutral variation, may account for some of the unexplained heritability [118]. Imprinted genes [119], gene x gene and gene x environment interactions [120, 121], ethnic-specific disease loci [103] or disease-associated haplotypes that are not identified by single SNP analyses [122] may also account for a substantial part of the missing heritability of obesity.

The establishment of many population-based studies that have collected genome-wide data on genetic variation has recently led to the formation of consortia facilitating powerful GWAS meta-analyses for BMI or WHR [104, 113]. Upcoming GWAS meta-analyses for continuous or extreme obesity phenotypes in larger sample sizes are likely to complete the current list of obesity predisposing genes, and will enable gene x environment or gene x gene well-powered GWAS studies. The completion of family-based GWAS for obesity-related traits will be useful to identify disease-associated imprinted loci and haplotypes [118]. The integration of data issued from expression and DNA arrays offers the possibility to identify true disease-associated SNPs that do not reach a genome-wide significant level of association in initial GWAS analysis. This approach recently led to the identification of F13A1 as a valuable candidate gene for obesity [123], and SNP prioritization programs integrating expression QTL information are now emerging [124]. A new generation of dense DNA arrays (including up to 5 millions SNP and CNVs) based on the detailed information of rare and common single nucleotide and copy number variation in the human genome provided by the 1000 Genome project [125], combined with the development of adequate statistical methodologies [126], will enable the identification of novel rare variants predisposing to obesity. Numerous rare variants may be detected in a gene or region but they may have disparate effects on phenotype. One strategy to study associations between such variants and disease might be pooling rare variants together using logical criteria and analyzing them as a single group [127].

Others strategies may shortly lead to a more exhaustive picture of the rare variants explaining highly penetrant forms of obesity. High-resolution homozygosity mapping in large consanguineous pedigrees is a powerful approach to discover novel obesity loci with a recessive mode of inheritance, as recently exemplified in syndromic forms of obesity [128]. Exome capture and parallel sequencing strategies in carefully selected unrelated cases and controls have proven successful for gene identification [129] and this approach should be successfully extended in the future to large pedigrees with a Mendelian pattern of obesity. The high occurrence of Mendelian patterns of inheritance observed in multigenerational pedigrees with extreme obesity, the fact that > 130 genes lead to a severe obesity phenotype in mice when down or up-regulated, and that every single monogenic gene explains a weak fraction of human obesity suggests that many monogenic genes remain to be discovered [77]. In addition to the deletion at 16p11.2, the identification of additional rare structural variants associated with highly penetrant forms of obesity by genome-wide association approaches are likely to be followed by systematic resequencing approaches for genes located in genome structural variation intervals, that may help to identify additional Mendelian obesity genes. Altogether, these novel methodologies are likely to fill the gap of the “missing heritability” of obesity in the near future.

New Phenotypes for New Genes

On the Use of BMI Variable in GWAS

Heritability studies in young and older adults have recently shown that genetic factors modulating BMI levels are only partially shared with those modulating change in BMI across time, supporting a complex etiology of BMI level and BMI change [19]. As recent GWAS have focused on the BMI level only [63, 64, 104, 130], an interesting perspective would be to perform a GWAS for parameters related to BMI change across life span in order to find novel SNPs robustly associated with more complex obesity phenotypes. The ‘set point’ theory proposes the existence of a feedback biological control system that regulates total body weight to a constant ‘body-inherent’ weight [131]. However, observational studies suggest that a set point in humans, if any, is loose rather than tightly controlled [132] and BMI variance in adulthood is expected to vary significantly in populations, from ‘BMI stable’ to ‘BMI yo-yo’ longitudinal patterns. In line with a growing interest for the genetic analysis on the variance of quantitative traits [133], it may be interesting to estimate the heritability of BMI variance across time and, if substantial, to identify gene variants associated with BMI variance by GWAS.

Recent GWAS meta-analyses for BMI [63, 64, 104, 130] have collected BMI information in individuals living in a wide range of heterogeneous environments. This approach has been successful but gene variants associated with BMI in specific environmental exposures may have been missed. Knowing that the inter-individual response for BMI distribution to a similar environment can differ considerably and genetic factors must explain these differences in populations [16], GWAS for BMI in specific environmental conditions may lead to the discovery of novel predisposing loci. It may be of particular interest to perform GWAS for BMI dynamic change in response to a brutal obesity-prone environment modification. From this point of view, the genetic analysis of BMI response to antipsychotic drugs intake, pregnancy or smoking cessation would be relevant, as 1) large inter-individual variability, likely to be modulated by genetic factors, is observed for BMI variation in response to these specific environmental exposures and 2) these conditions predispose to subsequent obesity development. GWAS for the BMI response to lifestyle intervention, pharmacotherapy and bariatric surgery may also be useful to identify the genes involved in efficient responses to specific treatments.

On the Use of ‘Deep Phenotyping’ in GWAS

Beyond the study of BMI, GWAS for additional phenotypes such as waist circumference or waist to hip ratio led to the identification of 14 novel genetic loci [110-113]. These data strongly argue for the use of different measures of adiposity in GWAS. Although widely used as “the obesity phenotype” in gene-search studies, BMI is not a good proxy of body fat content, especially at the individual level [134]. Whereas BMI is significantly associated with fat mass in obese subjects, there is little or no association between BMI and fat mass in normal weight and underweight subjects. At a given BMI, fat mass may vary by more than 100% [132]. Because phenotypic precision is major for genetic interrogation, the use of "deep phenotyping" information may help to dissect the genetic influences of obesity [135]. GWAS for endophenotypes such as body fat content and percentage of fat mass estimated by dual-emission X-ray absorptiometry, behavioral food intake measured by ad libitum meal test [136] and energy expenditure estimated by room respiration calorimetry [136] may provide highly relevant information.

Müller et al. [137] considered that the genetic basis of body weight regulation is unlikely to be fully discernable in individuals who are at stable energy equilibrium (i.e., at a stable body weight). They proposed to use dynamic phenotypes, including energy intake, energy expenditure and partitioning under conditions of controlled over and underfeeding rather than static phenotypes such as body composition and energy homeostasis. Such dynamic phenotypes should provide a more sensible basis for future genetic studies on obesity [132].

In conclusion, nine loci have been identified in Mendelian forms of non-syndromic obesity, and 58 loci have been robustly associated with polygenic obesity up to now but these loci explain a small fraction of the heritability for obesity. It seems that we are only at ‘the end of the beginning’ of the search for genetic variants that predispose to obesity. Innovative methodologies and technologies presented in detail here lead to optimism, and there is no doubt that we will live a prolific period of discovery in human obesity genetics in the upcoming years. The time required to shift from scientific discoveries to clinical applications is often underestimated [138] but on the longer term, the exhaustive genetic dissection of obesity will help to characterize the disease mechanisms, to provide new targets for drug design, and to lead to an efficient diagnosis, treatment, and prevention of obesity.

ACKNOWLEDGEMENTS

We thank Yoan Gerrard for the editing of the manuscript, and the reviewers for their helpful comments.

AUTHORS’ CONTRIBUTIONS

Both authors contributed to the conception and production of the manuscript and approved the final version.

REFERENCES

  • 1.Finucane MM, Stevens GA, Cowan MJ, Danaei G, Lin JK, Paciorek CJ, Singh GM, Gutierrez HR, Lu Y, Bahalim AN, Farzadfar F, Riley LM, Ezzati M. National, regional, and global trends in body-mass index since 1980: systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9.1 million participants. Lancet. 2011;377(9765):557–567. doi: 10.1016/S0140-6736(10)62037-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.de Onis M, Blossner M, Borghi E. Global prevalence and trends of overweight and obesity among preschool children. Am. J. Clin. Nutr. 2010;92(5):1257–1264. doi: 10.3945/ajcn.2010.29786. [DOI] [PubMed] [Google Scholar]
  • 3.Yanovski SZ, Yanovski JA. Obesity prevalence in the United States--up, down, or sideways? N. Engl. J. Med. 2011;364(11):987–989. doi: 10.1056/NEJMp1009229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Fontaine KR, Redden DT, Wang C, Westfall AO, Allison DB. Years of life lost due to obesity. JAMA. 2003;289(2):187–193. doi: 10.1001/jama.289.2.187. [DOI] [PubMed] [Google Scholar]
  • 5.Cecchini M, Sassi F, Lauer JA, Lee YY, Guajardo-Barron V, Chisholm D. Tackling of unhealthy diets, physical inactivity, and obesity: health effects and cost-effectiveness. Lancet. 2010;376(9754):1775–1784. doi: 10.1016/S0140-6736(10)61514-0. [DOI] [PubMed] [Google Scholar]
  • 6.Lamb MM, Dabelea D, Yin X, Ogden LG, Klingensmith GJ, Rewers M, Norris JM. Early-life predictors of higher body mass index in healthy children. Ann. Nutr. Metab. 2010;56(1):16–22. doi: 10.1159/000261899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Stunkard AJ, Foch TT, Hrubec Z. A twin study of human obesity. JAMA. 1986;256(1):51–54. [PubMed] [Google Scholar]
  • 8.Reilly JJ, Armstrong J, Dorosty AR, Emmett PM, Ness A, Rogers I, Steer C, Sherriff A. Early life risk factors for obesity in childhood: cohort study. BMJ. 2005;330(7504):1357. doi: 10.1136/bmj.38470.670903.E0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lee JH, Reed DR, Price RA. Familial risk ratios for extreme obesity: implications for mapping human obesity genes. Int. J. Obes. Relat. Metab. Disord. 1997;21(10):935–940. doi: 10.1038/sj.ijo.0800498. [DOI] [PubMed] [Google Scholar]
  • 10.Galton F. Natural inheritance. London: Macmillan; 1889. [Google Scholar]
  • 11.Feinleib M, Garrison RJ, Fabsitz R, Christian JC, Hrubec Z, Borhani NO, Kannel WB, Rosenman R, Schwartz JT, Wagner JO. The NHLBI twin study of cardiovascular disease risk factors: methodology and summary of results. Am. J. Epidemiol. 1977;106(4):284–285. doi: 10.1093/oxfordjournals.aje.a112464. [DOI] [PubMed] [Google Scholar]
  • 12.Stunkard AJ, Sorensen TI, Hanis C, Teasdale TW, Chakraborty R, Schull WJ, Schulsinger F. An adoption study of human obesity. N. Engl. J. Med. 1986;314(4):193–198. doi: 10.1056/NEJM198601233140401. [DOI] [PubMed] [Google Scholar]
  • 13.Stunkard AJ, Harris JR, Pedersen NL, McClearn GE. The body-mass index of twins who have been reared apart. N. Engl. J. Med. 1990;322(21):1483–1487. doi: 10.1056/NEJM199005243222102. [DOI] [PubMed] [Google Scholar]
  • 14.Pietilainen KH, Kaprio J, Rissanen A, Winter T, Rimpela A, Viken RJ, Rose RJ. 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. 1999;23(2):107–115. doi: 10.1038/sj.ijo.0800767. [DOI] [PubMed] [Google Scholar]
  • 15.Haworth CM, Plomin R, Carnell S, Wardle J. Childhood obesity: Genetic and environmental overlap with normal-range BMI. Obesity (Silver Spring) 2008;16(7):1585–1590. doi: 10.1038/oby.2008.240. [DOI] [PubMed] [Google Scholar]
  • 16.Wardle J, Carnell S, Haworth CM, Plomin R. Evidence for a strong genetic influence on childhood adiposity despite the force of the obesogenic environment. Am. J. Clin. Nutr. 2008;87(2):398–404. doi: 10.1093/ajcn/87.2.398. [DOI] [PubMed] [Google Scholar]
  • 17.Lajunen HR, Kaprio J, Keski-Rahkonen A, Rose RJ, Pulkkinen L, Rissanen A, Silventoinen K. Genetic and environmental effects on body mass index during adolescence: a prospective study among Finnish twins. Int. J. Obes. (Lond) 2009;33(5):559–567. doi: 10.1038/ijo.2009.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Demerath EW, Choh AC, Czerwinski SA, Lee M, Sun SS, Chumlea WC, Duren D, Sherwood RJ, Blangero J, Towne B, Siervogel RM. Genetic and environmental influences on infant weight and weight change: the Fels Longitudinal Study. Am. J. Hum. Biol. 2007;19(5):692–702. doi: 10.1002/ajhb.20660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.North KE, Graff M, Adair LS, Lange EM, Lange LA, Guo G, Gordon-Larsen P. Genetic epidemiology of BMI and body mass change from adolescence to young adulthood. Obesity (Silver Spring) 2010;18(7):1474–1476. doi: 10.1038/oby.2009.350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Mustelin L, Silventoinen K, Pietilainen K, Rissanen A, Kaprio J. Physical activity reduces the influence of genetic effects on BMI and waist circumference: a study in young adult twins. Int. J. Obes. (Lond) 2009;33(1):29–36. doi: 10.1038/ijo.2008.258. [DOI] [PubMed] [Google Scholar]
  • 21.McCaffery JM, Papandonatos GD, Bond DS, Lyons MJ, Wing RR. Gene X environment interaction of vigorous exercise and body mass index among male Vietnam-era twins. Am. J. Clin. Nutr. 2009;89(4):1011–1018. doi: 10.3945/ajcn.2008.27170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Schousboe K, Willemsen G, Kyvik KO, Mortensen J, Boomsma DI, Cornes BK, Davis CJ, Fagnani C, Hjelmborg J, Kaprio J, De Lange M, Luciano M, Martin NG, Pedersen N, Pietilainen KH, Rissanen A, Saarni S, Sorensen TI, Van Baal GC, Harris JR. Sex differences in heritability of BMI: a comparative study of results from twin studies in eight countries. Twin Res. 2003;6(5):409–421. doi: 10.1375/136905203770326411. [DOI] [PubMed] [Google Scholar]
  • 23.Klimentidis YC, Miller GF, Shriver MD. The relationship between European genetic admixture and body composition among Hispanics and Native Americans. Am. J. Hum. Biol. 2009;21(3):377–382. doi: 10.1002/ajhb.20886. [DOI] [PubMed] [Google Scholar]
  • 24.Cheng CY, Reich D, Coresh J, Boerwinkle E, Patterson N, Li M, North KE, Tandon A, Bailey-Wilson JE, Wilson JG, Kao WH. Admixture mapping of obesity-related traits in African Americans: the Atherosclerosis Risk in Communities (ARIC) Study. Obesity (Silver Spring) 2010;18(3):563–572. doi: 10.1038/oby.2009.282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Martin LJ, Woo JG, Morrison JA. Evidence of shared genetic effects between pre- and postobesity epidemic BMI levels. Obesity (Silver Spring) 2010;18(7):1378–1382. doi: 10.1038/oby.2009.394. [DOI] [PubMed] [Google Scholar]
  • 26.Luke A, Guo X, Adeyemo AA, Wilks R, Forrester T, Lowe W, Jr., Comuzzie AG, Martin LJ, Zhu X, Rotimi CN, Cooper RS. Heritability of obesity-related traits among Nigerians, Jamaicans and US black people. Int. J. Obes. Relat. Metab. Disord. 2001;25(7):1034–1041. doi: 10.1038/sj.ijo.0801650. [DOI] [PubMed] [Google Scholar]
  • 27.Malis C, Rasmussen EL, Poulsen P, Petersen I, Christensen K, Beck-Nielsen H, Astrup A, Vaag AA. Total and regional fat distribution is strongly influenced by genetic factors in young and elderly twins. Obes. Res. 2005;13(12):2139–2145. doi: 10.1038/oby.2005.265. [DOI] [PubMed] [Google Scholar]
  • 28.Beardsall K, Ong KK, Murphy N, Ahmed ML, Zhao JH, Peeters MW, Dunger DB. Heritability of childhood weight gain from birth and risk markers for adult metabolic disease in prepubertal twins. J. Clin. Endocrinol. Metab. 2009;94(10):3708–3713. doi: 10.1210/jc.2009-0757. [DOI] [PubMed] [Google Scholar]
  • 29.Carnell S, Haworth CM, Plomin R, Wardle J. Genetic influence on appetite in children. Int. J. Obes. (Lond) 2008;32(10):1468–1473. doi: 10.1038/ijo.2008.127. [DOI] [PubMed] [Google Scholar]
  • 30.Mustelin L, Latvala A, Pietilainen KH, Piirila P, Sovijarvi AR, Kujala UM, Rissanen A, Kaprio J. Associations between sports participation, cardiorespiratory fitness and adiposity in young adult twins. J. Appl. Physiol. 2011;110(3):681–686. doi: 10.1152/japplphysiol.00753.2010. [DOI] [PubMed] [Google Scholar]
  • 31.Bouchard C, Tremblay A, Nadeau A, Despres JP, Theriault G, Boulay MR, Lortie G, Leblanc C, Fournier G. Genetic effect in resting and exercise metabolic rates. Metabolism. 1989;38(4):364–370. doi: 10.1016/0026-0495(89)90126-1. [DOI] [PubMed] [Google Scholar]
  • 32.Farooqi IS, O'Rahilly S. Mutations in ligands and receptors of the leptin-melanocortin pathway that lead to obesity. Nat. Clin. Pract. Endocrinol. Metab. 2008;4(10):569–577. doi: 10.1038/ncpendmet0966. [DOI] [PubMed] [Google Scholar]
  • 33.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. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature. 1997;387(6636):903–908. doi: 10.1038/43185. [DOI] [PubMed] [Google Scholar]
  • 34.Clement K, Vaisse C, Lahlou N, Cabrol S, Pelloux V, Cassuto D, Gourmelen M, Dina C, Chambaz J, Lacorte JM, Basdevant A, Bougneres P, Lebouc Y, Froguel P, Guy-Grand B. A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature. 1998;392(6674):398–401. doi: 10.1038/32911. [DOI] [PubMed] [Google Scholar]
  • 35.Krude H, Biebermann H, Luck W, Horn R, Brabant G, Gruters A. Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nat. Genet. 1998;19(2):155–157. doi: 10.1038/509. [DOI] [PubMed] [Google Scholar]
  • 36.Jackson RS, Creemers JW, Ohagi S, Raffin-Sanson ML, Sanders L, Montague CT, Hutton JC, O'Rahilly S. Obesity and impaired prohormone processing associated with mutations in the human prohormone convertase 1 gene. Nat. Genet. 1997;16(3):303–306. doi: 10.1038/ng0797-303. [DOI] [PubMed] [Google Scholar]
  • 37.Farooqi IS, Keogh JM, Yeo GS, Lank EJ, Cheetham T, O'Rahilly S. Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene. N. Engl. J. Med. 2003;348(12):1085–1095. doi: 10.1056/NEJMoa022050. [DOI] [PubMed] [Google Scholar]
  • 38.Farooqi IS, Wangensteen T, Collins S, Kimber W, Matarese G, Keogh JM, Lank E, Bottomley B, Lopez-Fernandez J, Ferraz-Amaro I, Dattani MT, Ercan O, Myhre AG, Retterstol L, Stanhope R, Edge JA, McKenzie S, Lessan N, Ghodsi M, De Rosa V, Perna F, Fontana S, Barroso I, Undlien DE, O'Rahilly S. Clinical and molecular genetic spectrum of congenital deficiency of the leptin receptor. N. Engl. J. Med. 2007;356(3):237–247. doi: 10.1056/NEJMoa063988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Clement K, Dubern B, Mencarelli M, Czernichow P, Ito S, Wakamatsu K, Barsh GS, Vaisse C, Leger J. Unexpected endocrine features and normal pigmentation in a young adult patient carrying a novel homozygous mutation in the POMC gene. J. Clin. Endocrinol. Metab. 2008;93(12):4955–4962. doi: 10.1210/jc.2008-1164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.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. Small-intestinal dysfunction accompanies the complex endocrinopathy of human proprotein convertase 1 deficiency. J. Clin. Invest. 2003;112(10):1550–1560. doi: 10.1172/JCI18784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Stutzmann F, Tan K, Vatin V, Dina C, Jouret B, Tichet J, Balkau B, Potoczna N, Horber F, O'Rahilly S, Farooqi IS, Froguel P, Meyre D. Prevalence of melanocortin-4 receptor deficiency in Europeans and their age-dependent penetrance in multigenerational pedigrees. Diabetes. 2008;57(9):2511–2518. doi: 10.2337/db08-0153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Farooqi IS, Drop S, Clements A, Keogh JM, Biernacka J, Lowenbein S, Challis BG, O'Rahilly S. Heterozygosity for a POMC-null mutation and increased obesity risk in humans. Diabetes. 2006;55(9):2549–2553. doi: 10.2337/db06-0214. [DOI] [PubMed] [Google Scholar]
  • 43.Farooqi IS, Keogh JM, Kamath S, Jones S, Gibson WT, Trussell R, Jebb SA, Lip GY, O'Rahilly S. Partial leptin deficiency and human adiposity. Nature. 2001;414(6859):34–35. doi: 10.1038/35102112. [DOI] [PubMed] [Google Scholar]
  • 44.Farooqi IS, Volders K, Stanhope R, Heuschkel R, White A, Lank E, Keogh J, O'Rahilly S, Creemers JW. Hyperphagia and early-onset obesity due to a novel homozygous missense mutation in prohormone convertase 1/3. J. Clin. Endocrinol. Metab. 2007;92(9):3369–3373. doi: 10.1210/jc.2007-0687. [DOI] [PubMed] [Google Scholar]
  • 45.Lloyd DJ, Bohan S, Gekakis N. Obesity, hyperphagia and increased metabolic efficiency in Pc1 mutant mice. Hum. Mol. Genet. 2006;15(11):1884–1893. doi: 10.1093/hmg/ddl111. [DOI] [PubMed] [Google Scholar]
  • 46.Zhu X, Zhou A, Dey A, Norrbom C, Carroll R, Zhang C, Laurent V, Lindberg I, Ugleholdt R, Holst JJ, Steiner DF. Disruption of PC1/3 expression in mice causes dwarfism and multiple neuroendocrine peptide processing defects. Proc. Natl. Acad. Sci. USA. 2002;99(16):10293–10298. doi: 10.1073/pnas.162352599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.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, Schafer H, Platzer M, Biebermann H, Meitinger T, Hebebrand J. Prevalence, spectrum, and functional characterization of melanocortin-4 receptor gene mutations in a representative population-based sample and obese adults from Germany. J. Clin. Endocrinol. Metab. 2006;91(5):1761–1769. doi: 10.1210/jc.2005-2056. [DOI] [PubMed] [Google Scholar]
  • 48.Holder JL, Jr., Butte NF, Zinn AR. Profound obesity associated with a balanced translocation that disrupts the SIM1 gene. Hum. Mol. Genet. 2000;9(1):101–108. doi: 10.1093/hmg/9.1.101. [DOI] [PubMed] [Google Scholar]
  • 49.Yeo GS, Connie Hung CC, Rochford J, Keogh J, Gray J, Sivaramakrishnan S, O'Rahilly S, Farooqi IS. A de novo mutation affecting human TrkB associated with severe obesity and developmental delay. Nat. Neurosci. 2004;7(11):1187–1189. doi: 10.1038/nn1336. [DOI] [PubMed] [Google Scholar]
  • 50.Gray J, Yeo GS, Cox JJ, Morton J, Adlam AL, Keogh JM, Yanovski JA, El Gharbawy A, Han JC, Tung YC, Hodges JR, Raymond FL, O'Rahilly S, Farooqi IS. Hyperphagia, severe obesity, impaired cognitive function, and hyperactivity associated with functional loss of one copy of the brain-derived neurotrophic factor (BDNF) gene. Diabetes. 2006;55(12):3366–3371. doi: 10.2337/db06-0550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Bonaglia MC, Ciccone R, Gimelli G, Gimelli S, Marelli S, Verheij J, Giorda R, Grasso R, Borgatti R, Pagone F, Rodriguez L, Martinez-Frias ML, van Ravenswaaij C, Zuffardi O. Detailed phenotype-genotype study in five patients with chromosome 6q16 deletion: narrowing the critical region for Prader-Willi-like phenotype. Eur. J. Hum. Genet. 2008;16(12):1443–1449. doi: 10.1038/ejhg.2008.119. [DOI] [PubMed] [Google Scholar]
  • 52.Kernie SG, Liebl DJ, Parada LF. BDNF regulates eating behavior and locomotor activity in mice. EMBO J. 2000;19(6):1290–1300. doi: 10.1093/emboj/19.6.1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Michaud JL, Boucher F, Melnyk A, Gauthier F, Goshu E, Levy E, Mitchell GA, Himms-Hagen J, Fan CM. Sim1 haploinsufficiency causes hyperphagia, obesity and reduction of the paraventricular nucleus of the hypothalamus. Hum. Mol. Genet. 2001;10(14):1465–1473. doi: 10.1093/hmg/10.14.1465. [DOI] [PubMed] [Google Scholar]
  • 54.Xu B, Goulding EH, Zang K, Cepoi D, Cone RD, Jones KR, Tecott LH, Reichardt LF. Brain-derived neurotrophic factor regulates energy balance downstream of melanocortin-4 receptor. Nat. Neurosci. 2003;6(7):736–742. doi: 10.1038/nn1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Silos-Santiago I, Fagan AM, Garber M, Fritzsch B, Barbacid M. Severe sensory deficits but normal CNS development in newborn mice lacking TrkB and TrkC tyrosine protein kinase receptors. Eur. J. Neurosci. 1997;9(10):2045–2056. doi: 10.1111/j.1460-9568.1997.tb01372.x. [DOI] [PubMed] [Google Scholar]
  • 56.Feuk L, Carson AR, Scherer SW. Structural variation in the human genome. Nat. Rev. Genet. 2006;7(2):85–97. doi: 10.1038/nrg1767. [DOI] [PubMed] [Google Scholar]
  • 57.Bochukova EG, Huang N, Keogh J, Henning E, Purmann C, Blaszczyk K, Saeed S, Hamilton-Shield J, Clayton-Smith J, O'Rahilly S, Hurles ME, Farooqi IS. Large, rare chromosomal deletions associated with severe early-onset obesity. Nature. 2009;463(7281):666–670. doi: 10.1038/nature08689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Walters RG, Jacquemont S, Valsesia A, de Smith AJ, Martinet D, Andersson J, Falchi M, Chen F, Andrieux J, Lobbens S, Delobel B, Stutzmann F, El-Sayed Moustafa JS, Chevre JC, Lecoeur C, Vatin V, Bouquillon S, Buxton JL, Boute O, Holder-Espinasse M, Cuisset JM, Lemaitre MP, Ambresin AE, Brioschi A, Gaillard M, Giusti V, Fellmann F, Ferrarini A, Hadjikhani N, Campion D, Guilmatre A, Goldenberg A, Calmels N, Mandel JL, Le Caignec C, David A, Isidor B, Cordier MP, Dupuis-Girod S, Labalme A, Sanlaville D, Beri-Dexheimer M, Jonveaux P, Leheup B, Ounap K, Bochukova EG, Henning E, Keogh J, Ellis RJ, Macdermot KD, van Haelst MM, Vincent-Delorme C, Plessis G, Touraine R, Philippe A, Malan V, Mathieu-Dramard M, Chiesa J, Blaumeiser B, Kooy RF, Caiazzo R, Pigeyre M, Balkau B, Sladek R, Bergmann S, Mooser V, Waterworth D, Reymond A, Vollenweider P, Waeber G, Kurg A, Palta P, Esko T, Metspalu A, Nelis M, Elliott P, Hartikainen AL, McCarthy MI, Peltonen L, Carlsson L, Jacobson P, Sjostrom L, Huang N, Hurles ME, O'Rahilly S, Farooqi IS, Mannik K, Jarvelin MR, Pattou F, Meyre D, Walley AJ, Coin LJ, Blakemore AI, Froguel P, Beckmann JS. A new highly penetrant form of obesity due to deletions on chromosome 16p11.2. Nature. 2010;463(7281):671–675. doi: 10.1038/nature08727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Bachmann-Gagescu R, Mefford HC, Cowan C, Glew GM, Hing AV, Wallace S, Bader PI, Hamati A, Reitnauer PJ, Smith R, Stockton DW, Muhle H, Helbig I, Eichler EE, Ballif BC, Rosenfeld J, Tsuchiya KD. Recurrent 200-kb deletions of 16p11.2 that include the SH2B1 gene are associated with developmental delay and obesity. Genet. Med. 2010;12(10):641–647. doi: 10.1097/GIM.0b013e3181ef4286. [DOI] [PubMed] [Google Scholar]
  • 60.Wang K, Li WD, Glessner JT, Grant SF, Hakonarson H, Price RA. Large copy-number variations are enriched in cases with moderate to extreme obesity. Diabetes. 2010;59(10):2690–2694. doi: 10.2337/db10-0192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Jarick I, Vogel CI, Scherag S, Schafer H, Hebebrand J, Hinney A, Scherag A. Novel common copy number variation for early onset extreme obesity on chromosome 11q11 identified by a genome-wide analysis. Hum. Mol. Genet. 2011;20(4):840–852. doi: 10.1093/hmg/ddq518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ren D, Li M, Duan C, Rui L. Identification of SH2-B as a key regulator of leptin sensitivity, energy balance, and body weight in mice. Cell Metab. 2005;2(2):95–104. doi: 10.1016/j.cmet.2005.07.004. [DOI] [PubMed] [Google Scholar]
  • 63.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. Genome-wide association yields new sequence variants at seven loci that associate with measures of obesity. Nat. Genet. 2009;41(1):18–24. doi: 10.1038/ng.274. [DOI] [PubMed] [Google Scholar]
  • 64.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, Ridderstrale 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, 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. Six new loci associated with body mass index highlight a neuronal influence on body weight regulation. Nat. Genet. 2009;41(1):25–34. doi: 10.1038/ng.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Glessner JT, Bradfield JP, Wang K, Takahashi N, Zhang H, Sleiman PM, Mentch FD, Kim CE, Hou C, Thomas KA, Garris ML, Deliard S, Frackelton EC, Otieno FG, Zhao J, Chiavacci RM, Li M, Buxbaum JD, Berkowitz RI, Hakonarson H, Grant SF. A genome-wide study reveals copy number variants exclusive to childhood obesity cases. Am. J. Hum. Genet. 2010;87(5):661–666. doi: 10.1016/j.ajhg.2010.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Reich DE, Lander ES. On the allelic spectrum of human disease. Trends Genet. 2001;17(9):502–510. doi: 10.1016/s0168-9525(01)02410-6. [DOI] [PubMed] [Google Scholar]
  • 67.Rankinen T, Zuberi A, Chagnon YC, Weisnagel SJ, Argyropoulos G, Walts B, Perusse L, Bouchard C. The human obesity gene map: the 2005 update. Obesity (Silver Spring) 2006;14(4):529–644. doi: 10.1038/oby.2006.71. [DOI] [PubMed] [Google Scholar]
  • 68.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. Targeted disruption of the melanocortin-4 receptor results in obesity in mice. Cell. 1997;88(1):131–141. doi: 10.1016/s0092-8674(00)81865-6. [DOI] [PubMed] [Google Scholar]
  • 69.Cai G, Cole SA, Butte N, Bacino C, Diego V, Tan K, Goring HH, O'Rahilly S, Farooqi IS, Comuzzie AG. A quantitative trait locus on chromosome 18q for physical activity and dietary intake in Hispanic children. Obesity (Silver Spring) 2006;14(9):1596–1604. doi: 10.1038/oby.2006.184. [DOI] [PubMed] [Google Scholar]
  • 70.Xiang Z, Litherland SA, Sorensen NB, Proneth B, Wood MS, Shaw AM, Millard WJ, Haskell-Luevano C. Pharmacological characterization of 40 human melanocortin-4 receptor polymorphisms with the endogenous proopiomelanocortin-derived agonists and the agouti-related protein (AGRP) antagonist. Biochemistry. 2006;45(23):7277–7288. doi: 10.1021/bi0600300. [DOI] [PubMed] [Google Scholar]
  • 71.Geller F, Reichwald K, Dempfle A, Illig T, Vollmert C, Herpertz S, Siffert W, Platzer M, Hess C, Gudermann T, Biebermann H, Wichmann HE, Schafer H, Hinney A, Hebebrand J. Melanocortin-4 receptor gene variant I103 is negatively associated with obesity. Am. J. Hum. Genet. 2004;74(3):572–581. doi: 10.1086/382490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.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, Bougneres P, Froguel P, Meyre D. Non-synonymous polymorphisms in melanocortin-4 receptor protect against obesity: the two facets of a Janus obesity gene. Hum. Mol. Genet. 2007;16(15):1837–1844. doi: 10.1093/hmg/ddm132. [DOI] [PubMed] [Google Scholar]
  • 73.Wang D, Ma J, Zhang S, Hinney A, Hebebrand J, Wang Y, Wang HJ. Association of the MC4R V103I polymorphism with obesity: a Chinese case-control study and meta-analysis in 55,195 individuals. Obesity (Silver Spring) 2010;18(3):573–579. doi: 10.1038/oby.2009.268. [DOI] [PubMed] [Google Scholar]
  • 74.Benzinou M, Creemers JW, Choquet H, Lobbens S, Dina C, Durand E, Guerardel A, Boutin P, Jouret B, Heude B, Balkau B, Tichet J, Marre M, Potoczna N, Horber F, Le Stunff C, Czernichow S, Sandbaek A, Lauritzen T, Borch-Johnsen K, Andersen G, Kiess W, Korner A, Kovacs P, Jacobson P, Carlsson LM, Walley AJ, Jorgensen T, Hansen T, Pedersen O, Meyre D, Froguel P. Common nonsynonymous variants in PCSK1 confer risk of obesity. Nat. Genet. 2008;40(8):943–945. doi: 10.1038/ng.177. [DOI] [PubMed] [Google Scholar]
  • 75.Altmuller J, Palmer LJ, Fischer G, Scherb H, Wjst M. Genomewide scans of complex human diseases: true linkage is hard to find. Am. J. Hum. Genet. 2001;69(5):936–950. doi: 10.1086/324069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Meyre D, Lecoeur C, Delplanque J, Francke S, Vatin V, Durand E, Weill J, Dina C, Froguel P. A genome-wide scan for childhood obesity-associated traits in French families shows significant linkage on chromosome 6q22.31-q23.2. Diabetes. 2004;53(3):803–811. doi: 10.2337/diabetes.53.3.803. [DOI] [PubMed] [Google Scholar]
  • 77.Stone S, Abkevich V, Hunt SC, Gutin A, Russell DL, Neff CD, Riley R, Frech GC, Hensel CH, Jammulapati S, Potter J, Sexton D, Tran T, Gibbs D, Iliev D, Gress R, Bloomquist B, Amatruda J, Rae PM, Adams TD, Skolnick MH, Shattuck D. A major predisposition locus for severe obesity, at 4p15-p14. Am. J. Hum. Genet. 2002;70(6):1459–1468. doi: 10.1086/340670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.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 HW, Duggirala R, Feitosa MF, Froguel P, Hanson RL, Hebebrand J, Huezo-Dias P, Kissebah AH, Li W, Luke A, Martin LJ, Nash M, Ohman M, Palmer LJ, Peltonen L, Perola M, Price RA, Redline S, Srinivasan SR, Stern MP, Stone S, Stringham H, Turner S, Wijmenga C, D AC. Meta-analysis of genome-wide linkage studies in BMI and obesity. Obesity (Silver Spring) 2007;15(9):2263–2275. doi: 10.1038/oby.2007.269. [DOI] [PubMed] [Google Scholar]
  • 79.Morahan G, Mehta M, James I, Chen WM, Akolkar B, Erlich HA, Hilner JE, Julier C, Nerup J, Nierras C, Pociot F, Todd JA, Rich SS. Tests for genetic interactions in type 1 diabetes: Linkage and stratification analyses of 4,422 affected sib-pairs. Diabetes. 2011;60(3):1030–1040. doi: 10.2337/db10-1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Bowden DW, An SS, Palmer ND, Brown WM, Norris JM, Haffner SM, Hawkins GA, Guo X, Rotter JI, Chen YD, Wagenknecht LE, Langefeld CD. Molecular basis of a linkage peak: exome sequencing and family-based analysis identify a rare genetic variant in the ADIPOQ gene in the IRAS Family Study. Hum. Mol. Genet. 2010;19(20):4112–4120. doi: 10.1093/hmg/ddq327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Meyre D, Bouatia-Naji N, Tounian A, Samson C, Lecoeur C, Vatin V, Ghoussaini M, Wachter C, Hercberg S, Charpentier G, Patsch W, Pattou F, Charles MA, Tounian P, Clement K, Jouret B, Weill J, Maddux BA, Goldfine ID, Walley A, Boutin P, Dina C, Froguel P. Variants of ENPP1 are associated with childhood and adult obesity and increase the risk of glucose intolerance and type 2 diabetes. Nat. Genet. 2005;37(8):863–867. doi: 10.1038/ng1604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Jenkinson CP, Coletta DK, Flechtner-Mors M, Hu SL, Fourcaudot MJ, Rodriguez LM, Schneider J, Arya R, Stern MP, Blangero J, Duggirala R, DeFronzo RA. Association of genetic variation in ENPP1 with obesity-related phenotypes. Obesity (Silver Spring) 2008;16(7):1708–1713. doi: 10.1038/oby.2008.262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Bacci S, De Cosmo S, Prudente S, Trischitta V. ENPP1 gene, insulin resistance and related clinical outcomes. Curr. Opin. Clin. Nutr. Metab. Care. 2007;10(4):403–409. doi: 10.1097/MCO.0b013e3281e386c9. [DOI] [PubMed] [Google Scholar]
  • 84.Dong H, Maddux BA, Altomonte J, Meseck M, Accili D, Terkeltaub R, Johnson K, Youngren JF, Goldfine ID. Increased hepatic levels of the insulin receptor inhibitor, PC-1/NPP1, induce insulin resistance and glucose intolerance. Diabetes. 2005;54(2):367–372. doi: 10.2337/diabetes.54.2.367. [DOI] [PubMed] [Google Scholar]
  • 85.Bruning JC, Gautam D, Burks DJ, Gillette J, Schubert M, Orban PC, Klein R, Krone W, Muller-Wieland D, Kahn CR. Role of brain insulin receptor in control of body weight and reproduction. Science. 2000;289(5487):2122–2125. doi: 10.1126/science.289.5487.2122. [DOI] [PubMed] [Google Scholar]
  • 86.Odeleye OE, de Courten M, Pettitt DJ, Ravussin E. Fasting hyperinsulinemia is a predictor of increased body weight gain and obesity in Pima Indian children. Diabetes. 1997;46(8):1341–1345. doi: 10.2337/diab.46.8.1341. [DOI] [PubMed] [Google Scholar]
  • 87.Bottcher Y, Korner A, Reinehr T, Enigk B, Kiess W, Stumvoll M, Kovacs P. ENPP1 variants and haplotypes predispose to early onset obesity and impaired glucose and insulin metabolism in German obese children. J. Clin. Endocrinol. Metab. 2006;91(12):4948–4952. doi: 10.1210/jc.2006-0540. [DOI] [PubMed] [Google Scholar]
  • 88.Stone S, Abkevich V, Russell DL, Riley R, Timms K, Tran T, Trem D, Frank D, Jammulapati S, Neff CD, Iliev D, Gress R, He G, Frech GC, Adams TD, Skolnick MH, Lanchbury JS, Gutin A, Hunt SC, Shattuck D. TBC1D1 is a candidate for a severe obesity gene and evidence for a gene/gene interaction in obesity predisposition. Hum. Mol. Genet. 2006;15(18):2709–2720. doi: 10.1093/hmg/ddl204. [DOI] [PubMed] [Google Scholar]
  • 89.Meyre D, Farge M, Lecoeur C, Proenca C, Durand E, Allegaert F, Tichet J, Marre M, Balkau B, Weill J, Delplanque J, Froguel P. R125W coding variant in TBC1D1 confers risk for familial obesity and contributes to linkage on chromosome 4p14 in the French population. Hum. Mol. Genet. 2008;17(12):1798–1802. doi: 10.1093/hmg/ddn070. [DOI] [PubMed] [Google Scholar]
  • 90.Ross SE, Erickson RL, Gerin I, DeRose PM, Bajnok L, Longo KA, Misek DE, Kuick R, Hanash SM, Atkins KB, Andresen SM, Nebb HI, Madsen L, Kristiansen K, MacDougald OA. Microarray analyses during adipogenesis: understanding the effects of Wnt signaling on adipogenesis and the roles of liver X receptor alpha in adipocyte metabolism. Mol. Cell Biol. 2002;22(16):5989–5999. doi: 10.1128/MCB.22.16.5989-5999.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Zhou QL, Jiang ZY, Holik J, Chawla A, Hagan GN, Leszyk J, Czech MP. Akt substrate TBC1D1 regulates GLUT1 expression through the mTOR pathway in 3T3-L1 adipocytes. Biochem. J. 2008;411(3):647–655. doi: 10.1042/BJ20071084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Chadt A, Leicht K, Deshmukh A, Jiang LQ, Scherneck S, Bernhardt U, Dreja T, Vogel H, Schmolz K, Kluge R, Zierath JR, Hultschig C, Hoeben RC, Schurmann A, Joost HG, Al-Hasani H. Tbc1d1 mutation in lean mouse strain confers leanness and protects from diet-induced obesity. Nat. Genet. 2008;40(11):1354–1359. doi: 10.1038/ng.244. [DOI] [PubMed] [Google Scholar]
  • 93.Koumanov F, Holman GD. Thrifty Tbc1d1 and Tbc1d4 proteins link signalling and membrane trafficking pathways. Biochem. J. 2007;403(2):e9–11. doi: 10.1042/BJ20070271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Reich DE, Cargill M, Bolk S, Ireland J, Sabeti PC, Richter DJ, Lavery T, Kouyoumjian R, Farhadian SF, Ward R, Lander ES. Linkage disequilibrium in the human genome. Nature. 2001;411(6834):199–204. doi: 10.1038/35075590. [DOI] [PubMed] [Google Scholar]
  • 95.The International HapMap Project. Nature. 2003;426(6968):789–796. doi: 10.1038/nature02168. [DOI] [PubMed] [Google Scholar]
  • 96.Barrett JC, Cardon LR. Evaluating coverage of genome-wide association studies. Nat. Genet. 2006;38(6):659–662. doi: 10.1038/ng1801. [DOI] [PubMed] [Google Scholar]
  • 97.Ewis AA, Zhelev Z, Bakalova R, Fukuoka S, Shinohara Y, Ishikawa M, Baba Y. A history of microarrays in biomedicine. Expert Rev. Mol. Diagn. 2005;5(3):315–328. doi: 10.1586/14737159.5.3.315. [DOI] [PubMed] [Google Scholar]
  • 98.Wright AF, Carothers AD, Campbell H. Gene-environment interactions--the BioBank UK study. Pharmacogenomics J. 2002;2(2):75–82. doi: 10.1038/sj.tpj.6500085. [DOI] [PubMed] [Google Scholar]
  • 99.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. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science. 2007;316(5826):889–894. doi: 10.1126/science.1141634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.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. Variation in FTO contributes to childhood obesity and severe adult obesity. Nat. Genet. 2007;39(6):724–726. doi: 10.1038/ng2048. [DOI] [PubMed] [Google Scholar]
  • 101.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. Genome-wide association scan shows genetic variants in the FTO gene are associated with obesity-related traits. PLoS Genet. 2007;3(7):e115. doi: 10.1371/journal.pgen.0030115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hinney A, Nguyen TT, Scherag A, Friedel S, Bronner G, Muller TD, Grallert H, Illig T, Wichmann HE, Rief W, Schafer H, Hebebrand J. Genome wide association (GWA) study for early onset extreme obesity supports the role of fat mass and obesity associated gene (FTO) variants. PLoS ONE. 2007;2(12):e1361. doi: 10.1371/journal.pone.0001361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Cho YS, Go MJ, Kim YJ, Heo JY, Oh JH, Ban HJ, Yoon D, Lee MH, Kim DJ, Park M, Cha SH, Kim JW, Han BG, Min H, Ahn Y, Park MS, Han HR, Jang HY, Cho EY, Lee JE, Cho NH, Shin C, Park T, Park JW, Lee JK, Cardon L, Clarke G, McCarthy MI, Lee JY, Lee JK, Oh B, Kim HL. A large-scale genome-wide association study of Asian populations uncovers genetic factors influencing eight quantitative traits. Nat. Genet. 2009;41(5):527–534. doi: 10.1038/ng.357. [DOI] [PubMed] [Google Scholar]
  • 104.Speliotes EK, Willer CJ, Berndt SI, Monda KL, Thorleifsson G, Jackson AU, Allen HL, Lindgren CM, Luan J, Magi R, Randall JC, Vedantam S, Winkler TW, Qi L, Workalemahu T, Heid IM, Steinthorsdottir V, Stringham HM, Weedon MN, Wheeler E, Wood AR, Ferreira T, Weyant RJ, Segre AV, Estrada K, Liang L, Nemesh J, Park JH, Gustafsson S, Kilpelainen TO, Yang J, Bouatia-Naji N, Esko T, Feitosa MF, Kutalik Z, Mangino M, Raychaudhuri S, Scherag A, Smith AV, Welch R, Zhao JH, Aben KK, Absher DM, Amin N, Dixon AL, Fisher E, Glazer NL, Goddard ME, Heard-Costa NL, Hoesel V, Hottenga JJ, Johansson A, Johnson T, Ketkar S, Lamina C, Li S, Moffatt MF, Myers RH, Narisu N, Perry JR, Peters MJ, Preuss M, Ripatti S, Rivadeneira F, Sandholt C, Scott LJ, Timpson NJ, Tyrer JP, van Wingerden S, Watanabe RM, White CC, Wiklund F, Barlassina C, Chasman DI, Cooper MN, Jansson JO, Lawrence RW, Pellikka N, Prokopenko I, Shi J, Thiering E, Alavere H, Alibrandi MT, Almgren P, Arnold AM, Aspelund T, Atwood LD, Balkau B, Balmforth AJ, Bennett AJ, Ben-Shlomo Y, Bergman RN, Bergmann S, Biebermann H, Blakemore AI, Boes T, Bonnycastle LL, Bornstein SR, Brown MJ, Buchanan TA, Busonero F, Campbell H, Cappuccio FP, Cavalcanti-Proenca C, Chen YD, Chen CM, Chines PS, Clarke R, Coin L, Connell J, Day IN, Heijer M, Duan J, Ebrahim S, Elliott P, Elosua R, Eiriksdottir G, Erdos MR, Eriksson JG, Facheris MF, Felix SB, Fischer-Posovszky P, Folsom AR, Friedrich N, Freimer NB, Fu M, Gaget S, Gejman PV, Geus EJ, Gieger C, Gjesing AP, Goel A, Goyette P, Grallert H, Grassler J, Greenawalt DM, Groves CJ, Gudnason V, Guiducci C, Hartikainen AL, Hassanali N, Hall AS, Havulinna AS, Hayward C, Heath AC, Hengstenberg C, Hicks AA, Hinney A, Hofman A, Homuth G, Hui J, Igl W, Iribarren C, Isomaa B, Jacobs KB, Jarick I, Jewell E, John U, Jorgensen T, Jousilahti P, Jula A, Kaakinen M, Kajantie E, Kaplan LM, Kathiresan S, Kettunen J, Kinnunen L, Knowles JW, Kolcic I, Konig IR, Koskinen S, Kovacs P, Kuusisto J, Kraft P, Kvaloy K, Laitinen J, Lantieri O, Lanzani C, Launer LJ, Lecoeur C, Lehtimaki T, Lettre G, Liu J, Lokki ML, Lorentzon M, Luben RN, Ludwig B, Manunta P, Marek D, Marre M, Martin NG, McArdle WL, McCarthy A, McKnight B, Meitinger T, Melander O, Meyre D, Midthjell K, Montgomery GW, Morken MA, Morris AP, Mulic R, Ngwa JS, Nelis M, Neville MJ, Nyholt DR, O'Donnell CJ, O'Rahilly S, Ong KK, Oostra B, Pare G, Parker AN, Perola M, Pichler I, Pietilainen KH, Platou CG, Polasek O, Pouta A, Rafelt S, Raitakari O, Rayner NW, Ridderstrale M, Rief W, Ruokonen A, Robertson NR, Rzehak P, Salomaa V, Sanders AR, Sandhu MS, Sanna S, Saramies J, Savolainen MJ, Scherag S, Schipf S, Schreiber S, Schunkert H, Silander K, Sinisalo J, Siscovick DS, Smit JH, Soranzo N, Sovio U, Stephens J, Surakka I, Swift AJ, Tammesoo ML, Tardif JC, Teder-Laving M, Teslovich TM, Thompson JR, Thomson B, Tonjes A, Tuomi T, van Meurs JB, van Ommen GJ, Vatin V, Viikari J, Visvikis-Siest S, Vitart V, Vogel CI, Voight BF, Waite LL, Wallaschofski H, Walters GB, Widen E, Wiegand S, Wild SH, Willemsen G, Witte DR, Witteman JC, Xu J, Zhang Q, Zgaga L, Ziegler A, Zitting P, Beilby JP, Farooqi IS, Hebebrand J, Huikuri HV, James AL, Kahonen M, Levinson DF, Macciardi F, Nieminen MS, Ohlsson C, Palmer LJ, Ridker PM, Stumvoll M, Beckmann JS, Boeing H, Boerwinkle E, Boomsma DI, Caulfield MJ, Chanock SJ, Collins FS, Cupples LA, Smith GD, Erdmann J, Froguel P, Gronberg H, Gyllensten U, Hall P, Hansen T, Harris TB, Hattersley AT, Hayes RB, Heinrich J, Hu FB, Hveem K, Illig T, Jarvelin MR, Kaprio J, Karpe F, Khaw KT, Kiemeney LA, Krude H, Laakso M, Lawlor DA, Metspalu A, Munroe PB, Ouwehand WH, Pedersen O, Penninx BW, Peters A, Pramstaller PP, Quertermous T, Reinehr T, Rissanen A, Rudan I, Samani NJ, Schwarz PE, Shuldiner AR, Spector TD, Tuomilehto J, Uda M, Uitterlinden A, Valle TT, Wabitsch M, Waeber G, Wareham NJ, Watkins H, Wilson JF, Wright AF, Zillikens MC, Chatterjee N, McCarroll SA, Purcell S, Schadt EE, Visscher PM, Assimes TL, Borecki IB, Deloukas P, Fox CS, Groop LC, Haritunians T, Hunter DJ, Kaplan RC, Mohlke KL, O'Connell JR, Peltonen L, Schlessinger D, Strachan DP, van Duijn CM, Wichmann HE, Frayling TM, Thorsteinsdottir U, Abecasis GR, Barroso I, Boehnke M, Stefansson K, North KE, McCarthy MI, Hirschhorn JN, Ingelsson E, Loos RJ. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat. Genet. 2010;42(11):937–948. doi: 10.1038/ng.686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.den Hoed M, Ekelund U, Brage S, Grontved A, Zhao JH, Sharp SJ, Ong KK, Wareham NJ, Loos RJ. Genetic susceptibility to obesity and related traits in childhood and adolescence: influence of loci identified by genome-wide association studies. Diabetes. 2010;59(11):2980–2988. doi: 10.2337/db10-0370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Sandholt CH, Sparso T, Grarup N, Albrechtsen A, Almind K, Hansen L, Toft U, Jorgensen T, Hansen T, Pedersen O. Combined analyses of 20 common obesity susceptibility variants. Diabetes. 2010;59(7):1667–1673. doi: 10.2337/db09-1042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Meyre D, Delplanque J, Chevre JC, Lecoeur C, Lobbens S, Gallina S, Durand E, Vatin V, Degraeve F, Proenca C, Gaget S, Korner 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, Jarvelin MR, Visvikis-Siest S, Balkau B, Sladek R, Balding D, Walley A, Dina C, Froguel P. Genome-wide association study for early-onset and morbid adult obesity identifies three new risk loci in European populations. Nat. Genet. 2009;41(2):157–159. doi: 10.1038/ng.301. [DOI] [PubMed] [Google Scholar]
  • 108.Cotsapas C, Speliotes EK, Hatoum IJ, Greenawalt DM, Dobrin R, Lum PY, Suver C, Chudin E, Kemp D, Reitman M, Voight BF, Neale BM, Schadt EE, Hirschhorn JN, Kaplan LM, Daly MJ. Common body mass index-associated variants confer risk of extreme obesity. Hum. Mol. Genet. 2009;18(18):3502–3507. doi: 10.1093/hmg/ddp292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Scherag A, Dina C, Hinney A, Vatin V, Scherag S, Vogel CI, Muller TD, Grallert H, Wichmann HE, Balkau B, Heude B, Jarvelin MR, Hartikainen AL, Levy-Marchal C, Weill J, Delplanque J, Korner A, Kiess W, Kovacs P, Rayner NW, Prokopenko I, McCarthy MI, Schafer H, Jarick I, Boeing H, Fisher E, Reinehr T, Heinrich J, Rzehak P, Berdel D, Borte M, Biebermann H, Krude H, Rosskopf D, Rimmbach C, Rief W, Fromme T, Klingenspor M, Schurmann A, Schulz N, Nothen MM, Muhleisen TW, Erbel R, Jockel KH, Moebus S, Boes T, Illig T, Froguel P, Hebebrand J, Meyre D. Two new Loci for body-weight regulation identified in a joint analysis of genome-wide association studies for early-onset extreme obesity in French and german study groups. PLoS Genet. 2010;6(4):e1000916. doi: 10.1371/journal.pgen.1000916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Lindgren CM, Heid IM, Randall JC, Lamina C, Steinthorsdottir V, Qi L, Speliotes EK, Thorleifsson G, Willer CJ, Herrera BM, Jackson AU, Lim N, Scheet P, Soranzo N, Amin N, Aulchenko YS, Chambers JC, Drong A, Luan J, Lyon HN, Rivadeneira F, Sanna S, Timpson NJ, Zillikens MC, Zhao JH, Almgren P, Bandinelli S, Bennett AJ, Bergman RN, Bonnycastle LL, Bumpstead SJ, Chanock SJ, Cherkas L, Chines P, Coin L, Cooper C, Crawford G, Doering A, Dominiczak A, Doney AS, Ebrahim S, Elliott P, Erdos MR, Estrada K, Ferrucci L, Fischer G, Forouhi NG, Gieger C, Grallert H, Groves CJ, Grundy S, Guiducci C, Hadley D, Hamsten A, Havulinna AS, Hofman A, Holle R, Holloway JW, Illig T, Isomaa B, Jacobs LC, Jameson K, Jousilahti P, Karpe F, Kuusisto J, Laitinen J, Lathrop GM, Lawlor DA, Mangino M, McArdle WL, Meitinger T, Morken MA, Morris AP, Munroe P, Narisu N, Nordstrom A, Nordstrom P, Oostra BA, Palmer CN, Payne F, Peden JF, Prokopenko I, Renstrom F, Ruokonen A, Salomaa V, Sandhu MS, Scott LJ, Scuteri A, Silander K, Song K, Yuan X, Stringham HM, Swift AJ, Tuomi T, Uda M, Vollenweider P, Waeber G, Wallace C, Walters GB, Weedon MN, Witteman JC, Zhang C, Zhang W, Caulfield MJ, Collins FS, Davey Smith G, Day IN, Franks PW, Hattersley AT, Hu FB, Jarvelin MR, Kong A, Kooner JS, Laakso M, Lakatta E, Mooser V, Morris AD, Peltonen L, Samani NJ, Spector TD, Strachan DP, Tanaka T, Tuomilehto J, Uitterlinden AG, van Duijn CM, Wareham NJ, Hugh W, Waterworth DM, Boehnke M, Deloukas P, Groop L, Hunter DJ, Thorsteinsdottir U, Schlessinger D, Wichmann HE, Frayling TM, Abecasis GR, Hirschhorn JN, Loos RJ, Stefansson K, Mohlke KL, Barroso I, McCarthy MI. Genome-wide association scan meta-analysis identifies three Loci influencing adiposity and fat distribution. PLoS Genet. 2009;5(6):e1000508. doi: 10.1371/journal.pgen.1000508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Chambers JC, Elliott P, Zabaneh D, Zhang W, Li Y, Froguel P, Balding D, Scott J, Kooner JS. Common genetic variation near MC4R is associated with waist circumference and insulin resistance. Nat. Genet. 2008;40(6):716–718. doi: 10.1038/ng.156. [DOI] [PubMed] [Google Scholar]
  • 112.Heard-Costa NL, Zillikens MC, Monda KL, Johansson A, Harris TB, Fu M, Haritunians T, Feitosa MF, Aspelund T, Eiriksdottir G, Garcia M, Launer LJ, Smith AV, Mitchell BD, McArdle PF, Shuldiner AR, Bielinski SJ, Boerwinkle E, Brancati F, Demerath EW, Pankow JS, Arnold AM, Chen YD, Glazer NL, McKnight B, Psaty BM, Rotter JI, Amin N, Campbell H, Gyllensten U, Pattaro C, Pramstaller PP, Rudan I, Struchalin M, Vitart V, Gao X, Kraja A, Province MA, Zhang Q, Atwood LD, Dupuis J, Hirschhorn JN, Jaquish CE, O'Donnell CJ, Vasan RS, White CC, Aulchenko YS, Estrada K, Hofman A, Rivadeneira F, Uitterlinden AG, Witteman JC, Oostra BA, Kaplan RC, Gudnason V, O'Connell JR, Borecki IB, van Duijn CM, Cupples LA, Fox CS, North KE. NRXN3 is a novel locus for waist circumference: a genome-wide association study from the CHARGE Consortium. PLoS Genet. 2009;5(6):e1000539. doi: 10.1371/journal.pgen.1000539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Heid IM, Jackson AU, Randall JC, Winkler TW, Qi L, Steinthorsdottir V, Thorleifsson G, Zillikens MC, Speliotes EK, Magi R, Workalemahu T, White CC, Bouatia-Naji N, Harris TB, Berndt SI, Ingelsson E, Willer CJ, Weedon MN, Luan J, Vedantam S, Esko T, Kilpelainen TO, Kutalik Z, Li S, Monda KL, Dixon AL, Holmes CC, Kaplan LM, Liang L, Min JL, Moffatt MF, Molony C, Nicholson G, Schadt EE, Zondervan KT, Feitosa MF, Ferreira T, Allen HL, Weyant RJ, Wheeler E, Wood AR, Estrada K, Goddard ME, Lettre G, Mangino M, Nyholt DR, Purcell S, Smith AV, Visscher PM, Yang J, McCarroll SA, Nemesh J, Voight BF, Absher D, Amin N, Aspelund T, Coin L, Glazer NL, Hayward C, Heard-Costa NL, Hottenga JJ, Johansson A, Johnson T, Kaakinen M, Kapur K, Ketkar S, Knowles JW, Kraft P, Kraja AT, Lamina C, Leitzmann MF, McKnight B, Morris AP, Ong KK, Perry JR, Peters MJ, Polasek O, Prokopenko I, Rayner NW, Ripatti S, Rivadeneira F, Robertson NR, Sanna S, Sovio U, Surakka I, Teumer A, van Wingerden S, Vitart V, Zhao JH, Cavalcanti-Proenca C, Chines PS, Fisher E, Kulzer JR, Lecoeur C, Narisu N, Sandholt C, Scott LJ, Silander K, Stark K, Tammesoo ML, Teslovich TM, Timpson NJ, Watanabe RM, Welch R, Chasman DI, Cooper MN, Jansson JO, Kettunen J, Lawrence RW, Pellikka N, Perola M, Vandenput L, Alavere H, Almgren P, Atwood LD, Bennett AJ, Biffar R, Bonnycastle LL, Bornstein SR, Buchanan TA, Campbell H, Day IN, Dei M, Dorr M, Elliott P, Erdos MR, Eriksson JG, Freimer NB, Fu M, Gaget S, Geus EJ, Gjesing AP, Grallert H, Grassler J, Groves CJ, Guiducci C, Hartikainen AL, Hassanali N, Havulinna AS, Herzig KH, Hicks AA, Hui J, Igl W, Jousilahti P, Jula A, Kajantie E, Kinnunen L, Kolcic I, Koskinen S, Kovacs P, Kroemer HK, Krzelj V, Kuusisto J, Kvaloy K, Laitinen J, Lantieri O, Lathrop GM, Lokki ML, Luben RN, Ludwig B, McArdle WL, McCarthy A, Morken MA, Nelis M, Neville MJ, Pare G, Parker AN, Peden JF, Pichler I, Pietilainen KH, Platou CG, Pouta A, Ridderstrale M, Samani NJ, Saramies J, Sinisalo J, Smit JH, Strawbridge RJ, Stringham HM, Swift AJ, Teder-Laving M, Thomson B, Usala G, van Meurs JB, van Ommen GJ, Vatin V, Volpato CB, Wallaschofski H, Walters GB, Widen E, Wild SH, Willemsen G, Witte DR, Zgaga L, Zitting P, Beilby JP, James AL, Kahonen M, Lehtimaki T, Nieminen MS, Ohlsson C, Palmer LJ, Raitakari O, Ridker PM, Stumvoll M, Tonjes A, Viikari J, Balkau B, Ben-Shlomo Y, Bergman RN, Boeing H, Smith GD, Ebrahim S, Froguel P, Hansen T, Hengstenberg C, Hveem K, Isomaa B, Jorgensen T, Karpe F, Khaw KT, Laakso M, Lawlor DA, Marre M, Meitinger T, Metspalu A, Midthjell K, Pedersen O, Salomaa V, Schwarz PE, Tuomi T, Tuomilehto J, Valle TT, Wareham NJ, Arnold AM, Beckmann JS, Bergmann S, Boerwinkle E, Boomsma DI, Caulfield MJ, Collins FS, Eiriksdottir G, Gudnason V, Gyllensten U, Hamsten A, Hattersley AT, Hofman A, Hu FB, Illig T, Iribarren C, Jarvelin MR, Kao WH, Kaprio J, Launer LJ, Munroe PB, Oostra B, Penninx BW, Pramstaller PP, Psaty BM, Quertermous T, Rissanen A, Rudan I, Shuldiner AR, Soranzo N, Spector TD, Syvanen AC, Uda M, Uitterlinden A, Volzke H, Vollenweider P, Wilson JF, Witteman JC, Wright AF, Abecasis GR, Boehnke M, Borecki IB, Deloukas P, Frayling TM, Groop LC, Haritunians T, Hunter DJ, Kaplan RC, North KE, O'Connell JR, Peltonen L, Schlessinger D, Strachan DP, Hirschhorn JN, Assimes TL, Wichmann HE, Thorsteinsdottir U, van Duijn CM, Stefansson K, Cupples LA, Loos RJ, Barroso I, McCarthy MI, Fox CS, Mohlke KL, Lindgren CM. Meta-analysis identifies 13 new loci associated with waist-hip ratio and reveals sexual dimorphism in the genetic basis of fat distribution. Nat. Genet. 2010;42(11):949–960. doi: 10.1038/ng.685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.McCarroll SA, Hadnott TN, Perry GH, Sabeti PC, Zody MC, Barrett JC, Dallaire S, Gabriel SB, Lee C, Daly MJ, Altshuler DM. Common deletion polymorphisms in the human genome. Nat. Genet. 2006;38(1):86–92. doi: 10.1038/ng1696. [DOI] [PubMed] [Google Scholar]
  • 115.Sha BY, Yang TL, Zhao LJ, Chen XD, Guo Y, Chen Y, Pan F, Zhang ZX, Dong SS, Xu XH, Deng HW. Genome-wide association study suggested copy number variation may be associated with body mass index in the Chinese population. J. Hum. Genet. 2009;54(4):199–202. doi: 10.1038/jhg.2009.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Stranger BE, Forrest MS, Dunning M, Ingle CE, Beazley C, Thorne N, Redon R, Bird CP, de Grassi A, Lee C, Tyler-Smith C, Carter N, Scherer SW, Tavare S, Deloukas P, Hurles ME, Dermitzakis ET. Relative impact of nucleotide and copy number variation on gene expression phenotypes. Science. 2007;315(5813):848–853. doi: 10.1126/science.1136678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.McCarthy MI, Abecasis GR, Cardon LR, Goldstein DB, Little J, Ioannidis JP, Hirschhorn JN. Genome-wide association studies for complex traits: consensus, uncertainty and challenges. Nat. Rev. Genet. 2008;9(5):356–369. doi: 10.1038/nrg2344. [DOI] [PubMed] [Google Scholar]
  • 118.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. Finding the missing heritability of complex diseases. Nature. 2009;461(7265):747–753. doi: 10.1038/nature08494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Kong A, Steinthorsdottir V, Masson G, Thorleifsson G, Sulem P, Besenbacher S, Jonasdottir A, Sigurdsson A, Kristinsson KT, Jonasdottir A, Frigge ML, Gylfason A, Olason PI, Gudjonsson SA, Sverrisson S, Stacey SN, Sigurgeirsson B, Benediktsdottir KR, Sigurdsson H, Jonsson T, Benediktsson R, Olafsson JH, Johannsson OT, Hreidarsson AB, Sigurdsson G, Voight BF, Scott LJ, Steinthorsdottir V, Dina C, Zeggini E, Huth C, Aulchenko YS, Welch RP, Thorleifsson G, McCulloch LJ, Ferreira T, Grallert H, Amin N, Wu G, Willer CJ, Raychaudhuri S, Purcell S, McCarroll SA, Langenberg C, Hoffmann OM, Dupuis J, Qi L, Segre AV, van Hoek M, Navarro P, Ardlie K, Balkau B, Benediktsson R, Bennett AJ, Blagieva R, Boerwinkle E, Bonnycastle LL, Bostrom KB, Bravenboer B, Bumpstead S, Burtt NP, Charpentier G, Chines PS, Cornelis M, Couper DJ, Crawford G, Doney AS, Elliott KS, Elliott AL, Erdos MR, Fox CS, Franklin CS, Ganser M, Gieger C, Grarup N, Green T, Griffin S, Groves CJ, Guiducci C, Hadjadj S, Hassanali N, Herder C, Isomaa B, Jackson AU, Johnson PR, Jorgensen T, Kao WH, Klopp N, Kong A, Kraft P, Kuusisto J, Lauritzen T, Li M, Lieverse A, Lindgren CM, Lyssenko V, Marre M, Meitinger T, Midthjell K, Morken MA, Narisu N, Nilsson P, Owen KR, Payne F, Perry JR, Petersen AK, Platou C, Proenca C, Prokopenko I, Rathmann W, William Rayner N, Robertson NR, Rocheleau G, Roden M, Sampson MJ, Saxena R, Shields BM, Shrader P, Sigurdsson G, Smith N, Sparso T, Strassburger K, Stringham HM, Sun Q, Swift AJ, Thorand B, Tichet J, Tuomi T, van Dam R, van Herpt T, Walters GB, Weedon MN, Witteman J, Bergman RN, Cauchi S, Collins FS, Gloyn AL, Gyllensten U, Hansen T, Hide WA, Hitman GA, Hofman A, Hunter D, Hveem K, Laakso M, Mohlke KL, Morris AD, Palmer CN, Pramstaller PP, Rudan I, Sijbrands E, Stein LD, Tuomilehto J, Uitterlinden A, Walker M, Wareham NJ, Watanabe RM, Abecasis GR, Barroso I, Boehm BO, Campbell H, Daly MJ, Florez JC, Frayling TM, Groop L, Hattersley AT, Hu FB, Meigs JB, Morris AP, Pankow JS, Pedersen O, Sladek R, Thorsteinsdottir U, Wichmann HE, Wilson JF, Illig T, Froguel P, van Duijn CM, Stefansson K, Altshuler D, Boehnke M, McCarthy MI, Ferguson-Smith AC, Gudbjartsson DF, Thorsteinsdottir U, Stefansson K. Parental origin of sequence variants associated with complex diseases. Nature. 2009;462(7275):868–874. doi: 10.1038/nature08625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Manning AK, LaValley M, Liu CT, Rice K, An P, Liu Y, Miljkovic I, Rasmussen-Torvik L, Harris TB, Province MA, Borecki IB, Florez JC, Meigs JB, Cupples LA, Dupuis J. Meta-analysis of gene-environment interaction: joint estimation of SNP and SNP x environment regression coefficients. Genet. Epidemiol. 2011;35(1):11–18. doi: 10.1002/gepi.20546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kooperberg C, Leblanc M, Dai JY, Rajapakse I. Structures and Assumptions: Strategies to Harness Gene x Gene and Gene x Environment Interactions in GWAS. Stat. Sci. 2009;24(4):472–488. doi: 10.1214/09-sts287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Tregouet DA, Konig IR, Erdmann J, Munteanu A, Braund PS, Hall AS, Grosshennig A, Linsel-Nitschke P, Perret C, DeSuremain M, Meitinger T, Wright BJ, Preuss M, Balmforth AJ, Ball SG, Meisinger C, Germain C, Evans A, Arveiler D, Luc G, Ruidavets JB, Morrison C, van der Harst P, Schreiber S, Neureuther K, Schafer A, Bugert P, El Mokhtari NE, Schrezenmeir J, Stark K, Rubin D, Wichmann HE, Hengstenberg C, Ouwehand W, Ziegler A, Tiret L, Thompson JR, Cambien F, Schunkert H, Samani NJ. Genome-wide haplotype association study identifies the SLC22A3-LPAL2-LPA gene cluster as a risk locus for coronary artery disease. Nat. Genet. 2009;41(3):283–285. doi: 10.1038/ng.314. [DOI] [PubMed] [Google Scholar]
  • 123.Naukkarinen J, Surakka I, Pietilainen KH, Rissanen A, Salomaa V, Ripatti S, Yki-Jarvinen H, van Duijn CM, Wichmann HE, Kaprio J, Taskinen MR, Peltonen L. Use of genome-wide expression data to mine the "Gray Zone" of GWA studies leads to novel candidate obesity genes. PLoS Genet. 2010;6(6):e1000976. doi: 10.1371/journal.pgen.1000976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Gamazon ER, Zhang W, Konkashbaev A, Duan S, Kistner EO, Nicolae DL, Dolan ME, Cox NJ. SCAN: SNP and copy number annotation. Bioinformatics. 2010;26(2):259–262. doi: 10.1093/bioinformatics/btp644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Durbin RM, Abecasis GR, Altshuler DL, Auton A, Brooks LD, Durbin RM, Gibbs RA, Hurles ME, McVean GA. A map of human genome variation from population-scale sequencing. Nature. 2010;467(7319):1061–1073. doi: 10.1038/nature09534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.De La Vega FM, Bustamante CD, Leal SM. Genome-wide association mapping and rare alleles: From population genomics to personalized medicine - Session Introduction. Pac. Symp. Biocomput. 2011:74–75. doi: 10.1142/9789814335058_0008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Hoffmann TJ, Marini NJ, Witte JS. Comprehensive approach to analyzing rare genetic variants. PLoS One. 2010;5(11):e13584. doi: 10.1371/journal.pone.0013584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Harville HM, Held S, Diaz-Font A, Davis EE, Diplas BH, Lewis RA, Borochowitz ZU, Zhou W, Chaki M, Macdonald J, Kayserili H, Beales PL, Katsanis N, Otto E, Hildebrandt F. Identification of 11 Novel Mutations in 8 BBS Genes by High-Resolution Homozygosity Mapping. J. Med. Genet. 2010;47(4):262–267. doi: 10.1136/jmg.2009.071365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Ng SB, Bigham AW, Buckingham KJ, Hannibal MC, McMillin MJ, Gildersleeve HI, Beck AE, Tabor HK, Cooper GM, Mefford HC, Lee C, Turner EH, Smith JD, Rieder MJ, Yoshiura KI, Matsumoto N, Ohta T, Niikawa N, Nickerson DA, Bamshad MJ, Shendure J. Exome sequencing identifies MLL2 mutations as a cause of Kabuki syndrome. Nat. Genet. 2010;42(9):790–793. doi: 10.1038/ng.646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Loos RJ, Lindgren CM, Li S, Wheeler E, Zhao JH, Prokopenko I, Inouye M, Freathy RM, Attwood AP, Beckmann JS, Berndt SI, Jacobs KB, Chanock SJ, Hayes RB, 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 AS, Elliott KS, Elliott P, Evans DM, Sadaf Farooqi I, Froguel P, Ghori J, Groves CJ, Gwilliam R, Hadley D, Hall AS, Hattersley AT, Hebebrand J, Heid IM, Lamina C, Gieger C, Illig T, Meitinger T, Wichmann HE, Herrera B, Hinney A, Hunt SE, Jarvelin MR, Johnson T, Jolley JD, Karpe F, Keniry A, Khaw KT, Luben RN, 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 CN, Papadakis K, Potter S, Pouta A, Qi L, Randall JC, Rayner NW, Ring SM, Sandhu MS, Scherag A, Sims MA, Song K, Soranzo N, Speliotes EK, Syddall HE, Teichmann SA, Timpson NJ, Tobias JH, Uda M, Vogel CI, Wallace C, Waterworth DM, Weedon MN, Willer CJ, Wraight; 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. Common variants near MC4R are associated with fat mass, weight and risk of obesity. Nat. Genet. 2008;40(6):768–775. doi: 10.1038/ng.140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Harris RB. Role of set-point theory in regulation of body weight. FASEB J. 1990;4(15):3310–3318. doi: 10.1096/fasebj.4.15.2253845. [DOI] [PubMed] [Google Scholar]
  • 132.Muller MJ, Bosy-Westphal A, Krawczak M. Genetic studies of common types of obesity: a critique of the current use of phenotypes. Obes. Rev. 2010;11(8):612–618. doi: 10.1111/j.1467-789X.2010.00734.x. [DOI] [PubMed] [Google Scholar]
  • 133.Pare G, Cook NR, Ridker PM, Chasman DI. On the use of variance per genotype as a tool to identify quantitative trait interaction effects: a report from the Women's Genome Health Study. PLoS Genet. 2010;6(6):e1000981. doi: 10.1371/journal.pgen.1000981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Dulloo AG, Jacquet J, Solinas G, Montani JP, Schutz Y. Body composition phenotypes in pathways to obesity and the metabolic syndrome. Int. J. Obes. (Lond) 2010;34(Suppl 2):S4–17. doi: 10.1038/ijo.2010.234. [DOI] [PubMed] [Google Scholar]
  • 135.Tracy RP. 'Deep phenotyping': characterizing populations in the era of genomics and systems biology. Curr. Opin. Lipidol. 2008;19(2):151–157. doi: 10.1097/MOL.0b013e3282f73893. [DOI] [PubMed] [Google Scholar]
  • 136.Fisher JO, Cai G, Jaramillo SJ, Cole SA, Comuzzie AG, Butte NF. Heritability of hyperphagic eating behavior and appetite-related hormones among Hispanic children. Obesity (Silver Spring) 2007;15(6):1484–1495. doi: 10.1038/oby.2007.177. [DOI] [PubMed] [Google Scholar]
  • 137.Muller MJ, Bosy-Westphal A, Later W, Haas V, Heller M. Functional body composition: insights into the regulation of energy metabolism and some clinical applications. Eur. J. Clin. Nutr. 2009;63(9):1045–1056. doi: 10.1038/ejcn.2009.55. [DOI] [PubMed] [Google Scholar]
  • 138.Contopoulos-Ioannidis DG, Alexiou GA, Gouvias TC, Ioannidis JP. Medicine. Life cycle of translational research for medical interventions. Science. 2008;321(5894):1298–1299. doi: 10.1126/science.1160622. [DOI] [PubMed] [Google Scholar]
  • 139.Liu YJ, Liu WG, Wang L, Dina C, Yan H, Liu JF, Levy S, Papasian CJ, Drees BM, Meyre D, Delplanque J, Pei YF, Zhang L, Recker RR, Froquel P, Deng HW. Genome-wide association scan indentified CTNNBL1 as a novel gene for obesity. Hum. Mol. Genet. 2008;17(12):1803–1813. doi: 10.1093/hmg/ddn072. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Current Genomics are provided here courtesy of Bentham Science Publishers

RESOURCES