Genetics and Epigenetics of Obesity

Embed Size (px)

Citation preview

  • 7/22/2019 Genetics and Epigenetics of Obesity

    1/22

    Maturitas. 2011 May; 69(1): 4149.

    doi: 10.1016/j.maturitas.2011.02.018

    PMCID: PMC3213306

    Genetics and epigenetics of obesity

    Blanca M. Herrera, Sarah Keildson, and Cecilia M. Lindgren

    Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom

    Oxford Centre for Diabetes, Endocrinology and Metabolism University of Oxford, United Kingdom

    Cecilia M. Lindgren: [email protected]

    Corresponding author. Tel.: +44 1865 287598; fax: +44 1865 287664. Email: [email protected]

    Received June 14, 2010; Revised February 14, 2011; Accepted February 14, 2011.

    Copyright 2011 Elsevier Ireland Ltd.

    This document may be redistributed and reused, subject to certain conditions.

    This document was posted here by permission of the publisher. At the time of the deposit, it included all changes made during peer review,

    copy editing, and publishing. The U. S. National Library of Medicine is responsible for all links within the document and for incorporating any

    publisher-supplied amendments or retractions issued subsequently. The published journal article, guaranteedto be such by Elsevier, is available

    for free, on ScienceDirect, at: http://dx.doi.org/10.1016/j.maturitas.2011.02.018

    Abstract

    Obesity results from interactions between environmental and genetic factors. Despite a relatively high

    heritability of common, non-syndromic obesity (4070%), the search for genetic variants contributing

    to susceptibility has been a challenging task. Genome wide association (GWA) studies have

    dramatically changed the pace of detection of common genetic susceptibility variants. To date, more

    than 40 genetic variants have been associated with obesity and fat distribution. However, since these

    variants do not fully explain the heritability of obesity, other forms of variation, such as epigeneticsmarks, must be considered.

    Epigenetic marks, or imprinting, affect gene expression without actually changing the DNA

    sequence. Failures in imprinting are known to cause extreme forms of obesity (e.g. PraderWilli

    syndrome), but have also been convincingly associated with susceptibility to obesity. Furthermore,

    environmental exposures during critical developmental periods can affect the profile of epigenetic

    marks and result in obesity.

    We review the most recent evidence for genetic and epigenetic mechanisms involved in the

    susceptibility and development of obesity. Only a comprehensive understanding of the underlying

    genetic and epigenetic mechanisms, and the metabolic processes they govern, will allow us to

    manage, and eventually prevent, obesity.

    Abbreviations: BMI, body mass index; WC, waist circumference; WHR, waist:hip ratio; T2D,

    type-2-diabetes; LD, linkage disequilibrium; CNV, copy number variants; PWS, PraderWilli

    syndrome; QTL, quantitative trait loci; SNP, single nucleotide polymorphism

    Keywords: Genetic, Epigenetic, Fat distribution, Obesity, Methylation, Imprinting

    1. Introduction

    Overweight and obesity are becoming more widespread with global projections of more than 2.16

    billion overweight and 1.12 billion obese individuals by 2030 [1]. This clearly presents a worldwide

    Sponsored Document from

    MATURITAS

    a a a,b,

    a

    b

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    2 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    2/22

    clinical and public health burden, associated with social and personal criticism. It is also correlated

    with an increased risk of type-2-diabetes (T2D), cardiovascular disease, cancer and mortality [2,3].

    Despite intensive research, current efforts to reduce obesity by diet, exercise, education, surgery and

    drug therapies are failing to provide effective long-term solutions to this epidemic.

    At an individual level, obesity occurs when abnormal amounts of triglycerides are stored in adipose

    tissue and released from adipose tissue as free fatty acids (FFA) with detrimental effects [4]. Excess

    fat accumulation occurs when energy intake exceeds energy expenditure, although individuals

    respond differently to this imbalance owing to genetic predisposition. Twin studies estimateheritability of body mass index (BMI) to be 4070% in children and adults [57], and other

    anthropometric measures of obesity and regional fat distribution [skinfold thickness, waist

    circumference (WC) and waist:hip ratio (WHR)] show similar heritability [512]. Furthermore, there

    are ethnic differences in obesity; admixture mapping studies demonstrate that obesity correlates

    closely with the percentage of ancestry derived from ethnic groups with elevated prevalence [13,14].

    The goal of obesity research is to elucidate pathways and mechanisms that control obesity and to

    improve prevention, management and therapy.

    Here we review recent advances in identifying factors contributing to obesity susceptibility. We focus

    on:

    We discuss the impact of recent findings in these two areas and their joint potential to enhance

    understanding of obesity susceptibility mechanisms and aetiology.

    2. The identification of susceptibi lity loci for obesity

    Until 2006, the main approaches used to track down common variants influencing obesity, involved

    either hypothesis-free genome-wide linkage mapping in families with multiple obese subjects or

    association studies within candidate genes using casecontrol samples or parentoffspring trios. The

    former suffered from being underpowered for any sensible susceptibility models, as linkage is best

    placed to detect variants with high penetrance. As far as we can tell, common variants with high

    penetrance do not contribute substantially to risk of common forms of obesity and few, if any, robust

    signals have emerged from such efforts [15,16]. The latter candidate-gene association approach has

    historically been compromised by difficulties in selecting credible candidates. Selection was typically

    based on hypotheses about biological mechanisms putatively involved in obesity pathogenesis but, as

    the function of much of the genome is poorly characterized, it remains almost impossible to make fully

    informed decisions. In addition, all too often these candidate-gene studies were conducted in sample

    sets far too small to offer confident detection of variants with the range of effect sizes that are now

    known to be realistic. With hindsight, it is easy to appreciate why these approaches yielded fewexamples of genuine obesitysusceptibility variants.

    Consequently, over the last two decades, efforts in identifying and replicating genetic variants

    predisposing individuals to common forms of obesity were largely characterized by slow progress and

    limited success, in sharp contrast to the successful gene identification in monogenic and syndromic

    forms of obesity [15]. The most recent edition of the Human Obesity Gene Map gives an excellent

    overview of this; it lists 11 single gene mutations, 50 loci related to Mendelian syndromes relevant to

    human obesity, 244 knockout or transgenic animal models and 127 candidate genes, of which slightly

    less than 20% are replicated by 5 or more studies [15]. A total of 253 quantitative trait loci (QTL), for

    recent successes in identification of genetic variation affecting obesity trait susceptibility;(a)

    emerging evidence connecting epigenetic (heritable changes which affect gene function but do

    not modify DNA sequence) events with obesity.

    (b)

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    2 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    3/22

    different obesity-related phenotypes, have been reported from 61 genome-wide linkage scans and of

    these, only 20% are supported by more than one study [15].

    Over the past three years it has become possible, from technical and economic perspectives, to

    undertake hypothesis-free GWA testing in samples of sufficient size to generate convincing

    association results. The advent of the GWA approach was the result of three components. The first

    was the human genome sequence which subsequently enabled cataloguing genome-sequence

    variation. Secondly, the International HapMap Consortium (http://www.hapmap.org) [17]taught us

    that, in non-African-descent populations, extensive correlations (linkage disequilibrium, LD) betweenneighbouring single nucleotide polymorphisms (SNPs) constrain the number of independent genetic

    tests required to survey the genome, such that 80% of all common variation can be sampled using

    500 000 carefully selected SNPs [18,19]. Lastly, novel genotyping methods address the challenges of

    massively parallel SNP-typing at high accuracy and low cost [20]. The GWA approach has been

    hugely successful in identifying loci harbouring common forms of obesity (as defined by

    anthropometric measures: BMI, WC and/or WHR) susceptibility genes and hereto results from a total

    of 15 high-density GWAs (i.e. 300 000 SNPs, offering genome-wide coverage >65%) have been

    published (Table 1). These studies combined, have yielded over 50 loci associated with obesity

    (p-values

  • 7/22/2019 Genetics and Epigenetics of Obesity

    4/22

    association for 7 of the 13 loci previously reported to influence BMI [3234]. This suggests that

    variants influencing BMI might also contribute to more severe forms of obesity, which represents the

    extreme, of the phenotypic spectrum rather than a distinct condition, although, this needs

    confirmation in appropriately powered studies.

    Recently, a large-scale meta-analysis of 249 769 individuals confirmed 14 of these previously

    identified obesity susceptibility loci and identified 18 novel loci associated with BMI and overall

    adiposity (Table 2) [37]. As a consequence of the increased power of this study, new signals with

    lower minor allele frequencies and smaller effect sizes, compared to previously identified variants,were discovered. These results suggest that genome-wide association studies are only the first step in

    the identification of the causal variants that play a role in common, overall obesity and weight

    regulation and further insights into the underlying biological mechanisms and pathways will be

    needed for the effective treatment and management of these traits [37].

    While no consistent association has been shown between height and BMI-associated variants as a

    group, three of the loci (MC4R,RBJADCY3POMCand MTCH2NDUFS3) show individual

    associations with height [37]. The BMI-increasing alleles of the variants near POMCand MC4Rwere

    associated with decreased and increased height respectively, and this is analogous to the effects of

    severe mutations in POMCand MC4Ron height and weight [31,38].

    2.1. Genetics of fat distribution

    As described above, efforts to identify common and rare variants influencing BMI and risk of obesity

    have emphasized a key role for neuronal regulation of overall obesity [2124,2630,3235], but until

    recently provided few clues to processes responsible for variation in central obesity and fat

    distribution [29,39,40]. Measures of central and general adiposity are highly correlated: BMI has an

    r 0.9 with WC and 0.6 WHR. Also, WC and WHR are correlated with measures of intra-

    abdominal fat, measured by magnetic resonance imaging in obese women (r 0.6 and 0.5,

    respectively). Clinically, central obesity is associated with susceptibility to T2D, cardiovascular disease

    and increased mortality [3]. Evidence indicates that individual variability in patterns of fat

    distribution involve local, depot-specific and body-shape processes, which are probably independentof the mechanisms that control overall energy balance and general obesity. First, anthropometric

    measures of central adiposity are highly heritable [41]and, after BMI correction, heritability

    estimates remain high (0.6 for WC and 0.45 for WHR) [10,11,42]. Second, substantial gender-

    specific differences in fat distribution, reflect specific genetic influences [43,44]. Third, inherited

    lipodystrophies, which are monogenic syndromes, demonstrate that DNA variants can have specific

    effects on the development and/or maintenance of specific regional fat-depots and body shape [45].

    Hereto, five GWA studies of central obesity (WC and WHR) have been published [29,39,40,44,46](

    Table 1) and have identified 17 novel common obesity loci. The associations of three of these loci

    (MSRA; TFAP2Band NRXN3, Table 2) to WC appear to be mediated through BMI [36,37,39,44],

    implying their involvement in overall adiposity. The remaining 14 loci, however, showed significant

    associations with WHR, after correction for BMI, suggesting their role in body fat distribution,

    independent of overall obesity. Interestingly, some of these loci showed strong sex-specific effects in

    women [39,44]and in a study by Heid et al. [44], the WHR-increasing allele was shown to be

    associated with an increase in WC for 14 loci in women, but only six men. This evident heterogeneity

    between men and women across many of these WHR loci is a reflection of the sex-specific genetic

    effects driving individual patterns of body fat.

    3. Epigenetics and obesity

    Epigenetics is loosely defined as the study of heritable changes which affect gene function without

    2

    2

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    2 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    5/22

    3.1.1. DNA methylation

    3.1.2. Histone modif ications

    modifying the DNA sequence [47]. The maintenance of epigenetic marks through generations is

    poorly understood and the notion of their transmission is contentious [48]. Epigenetic marks are

    tissue specific and include DNA methylation and histone modifications which mediate biological

    processes such as imprinting. As many imprinted genes are growth factors, or regulators of gene

    expression controlling growth, imprinting disorders often feature obesity as one of their clinical

    characteristics.

    Genomic imprinting determines expression of alleles according to their maternal or paternal origin

    [49]and establishes a balance between the expression of the parental alleles influencing growth [50],resulting in counteracting growth effects of paternal and maternal genomes [51]. In addition to

    growth, imprinted genes are also involved in differentiation, development, viability and metabolic

    functions [52]. Two main clusters of genomic imprinting are known in humans: a region at 11p15

    containing several imprinted genes including IGF2,INS,KCNQ10T1(LIT1) (paternally expressed)

    and H19,KCNQ1, CDKN1C,PHLDA2,KVLQT1(maternally expressed) [53]. The second cluster at

    15q11q12 contains at least 7 imprinted genes, including; MKRN3,MAGEL2,NDN,SNURFSNRPN

    (paternally expressed) and UBE3A,ATP10A(maternally expressed) [54].

    Failures in imprinting which result in obesity by altering expression of growth and cellular

    differentiation factors can arise due to numerous genetic events: translocation, inversion, duplication,

    paternal disomy and hyper/hypo-methylation. For instance; paternal deletion or uniparental disomy

    at 15q11q13, results in PraderWilli syndrome (PWS). A syndrome characterized by severe

    (sometimes life threatening), early onset obesity caused by hyperphagia (due to dysfunction in the

    satiety centre [55]). Moderate obesity appears in Albright hereditary osteodystrophy (AHO), due to

    disruption of imprinting at the GNASgene (20q13.11 [50]).

    3.1. Mediators of genomic imprinting

    Genomic imprinting is mediated by DNA methylation as exemplified in the H19

    and IGF2loci [56]. Methylation is a widespread feature of the genome, and is obtained through the

    addition of a methyl group (CH3) to a cytosine positioned next to a guanine nucleotide (CpGs),

    usually in regions with a high presence of CpG dinucleotides (>60%). Methylation in a promoterregion results in the repression (silencing) of gene expression [57], this effect may be achieved by a

    number of mechanisms including: obstructing access to transcription factors/activators and

    recruitment of co-repressors (like histone deacetylases) which alter chromatin structure [58]resulting

    in failure to initiate transcription (Fig. 1).

    DNA in cells is packaged as chromatin in a beads on a string

    configuration. A length of 147 DNA base pairs wraps around a core histone octamer (a tetramer of

    histones H3 and H4, flanked by two H2AH2B dimmers) and these nucleosome beads are

    separated by DNA strings between 20 and 60 base pairs. Linker histones (H1/H5) occupy the exit

    and entry of the DNA into the histone, these structures are in turn coiled into a compact helical

    closed configuration [59]. Packaging DNA in this manner allows its efficient storage and isparamount for regulation of gene expression, as the closed configuration does not allow access to

    transcriptional enzymes. Post translational modification of the histones by: H3 and H4

    hyperacetylation in the promoter [60], methylation of the lys4 and lys36 of histone H3 open the

    structure and allow gene expression. In contrast, methylation of lys9, lys20 and lys27 on H3 [60,61])

    and ubiquitination of H2A present at high density CpG promoters [62]result in gene silencing

    closed configuration. Further complexity is achieved by the level of methylation, so mono-, bi- or

    tri-methylation may also effect the control of gene expression [63].

    A feature of both methylation and histone modifications is that they are both tissue specific and can

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    2 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    6/22

    vary with age (and developmental stage). Therefore, in order to place findings in an appropriate

    context, it is of paramount importance that evaluation of epigenetic factors be carried out on suitable

    tissues extracted at specified times [64,65](Fig. 2).

    3.2. Epigenetic changes introduced during early development may increase the risk of obesity

    Although birth weight (BW) is an imperfect summary index of growth, however, it has been widely

    used as a proxy for foetal nutrition and intrauterine growth. Although better measurements of foetal

    growth exist, BW measurements are used because they are non-intrusive, easily obtained and oftenfound in birth records, which enabled its use as in retrospective studies linking BW with adult onset

    diseases [66]. Many studies focus on the hypothesis that early environmental influences induce

    epigenetic variation, thereby permanently affecting metabolism and chronic disease risk. Specifically,

    for obesity, it has been shown that obese mothers tend to have obese children [67], it has been shown

    that clinical intervention to cause maternal weight loss can have a positive effect on reducing risk of

    obesity in the offspring [68]. The mechanisms by which nutritional challenges affect the risk of

    disease in later life are poorly understood. However, evidence indicates that the establishment of the

    epigenome can be affected by environmental factors during critical developmental periods [69].

    Possible disturbances of methylation may arise during foetal development due to lack of availability of

    dietary methyl donors [7072]. Potential interactions between the environment and epigenetic

    mechanisms mediating the expression of genes associated with increased BMI and adiposity, may

    also be possible as suggested for; the FTOlocus is a DNA-demethylase enzyme [23], the MC4Rgene

    which has reduced methylation following long-term exposure to a high fat diet [73], the PPAR

    protein which interacts with histone acetyltransferases [58]during adipogenesis and on the effect of

    diet on methylation ofPOMC[74]and Leptin[75].

    With this in mind, it is important to consider the effect of assisted reproduction technology on

    epigenetics and subsequently on obesity susceptibility. Despite small numbers, increased incidence of

    PWS, when compared to background population prevalence, has been shown [76]and subtler adverse

    effects may only become apparent later on in life. We are currently gaining more insight through the

    use of high-throughput sequencing methods for the detection of DNA methylation patterns, and

    chromatin immunoprecipitation (ChIP) used to detect histone modifications and new integrated

    genomics-based technologies which are currently being developed by the human epigenome project

    (http://www.epigenome.org) initiative.

    4. Remaining challenges

    4.1. Identifying novel loci

    Despite successes in susceptibility loci identification for obesity through the first wave of GWAs, the

    combined effect of the loci explains only 23% of the inherited contribution to obesity risk.

    4.2. Collaborativ e stud ies to fo r larger GWA meta-analysis

    The GIANT consortium is finalising the next wave of GWA meta-analysis for obesity related traits

    incorporating >100 000 samples. This increase in sample size should attain sufficient power to allow

    identification of common variants with even smaller effect sizes than hereto. Also, conditional

    analysis using the previously identified variants will reveal whether some of the already identified loci

    contain more than one independent association with obesity, which would be an additional source of

    unexplored variation contributing to the missing heritability.

    4.3. Obesity susceptibility associated to rare-low-frequency variants

    The obesity risk alleles that have been identified so far by GWAs are quite common (2791%) in

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    2 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    7/22

    European populations but the current approaches are limited in the range of minor allele frequencies

    (MAF) that are detected due to the range of allele frequencies (>5% MAF) present on current

    genotype arrays. The 1000 genomes project (http://www.1000genomes.org/) is designed to catalogue

    low and rare frequency variants and this will lead to the design of new genotype arrays including

    wider spectra of allele frequencies.

    4.4. Copy number variations

    Copy number variants (CNVs) are hard to detect due to technical constrains but recently [74], twocomplementary approaches have delivered the first examples of CNVs associated to obesity. The SNPs

    associated with obesity at theNEGR1locus are in strong LD with a nearby CNV [32], tagging a 45-kb

    deletion polymorphism which is a candidate causal-variant at this locus. Recently by analysing SNP

    genotype arrays that are enriched in CNV information and applying novel CNV detecting algorithms

    on these arrays deletions on chromosome 16p11.2 have been reported to be associated with extreme,

    highly penetrant obesity [77,78]. Further applications of these approaches in larger sample sets

    should allow a better understanding of total CNVs contribution to overall variation in obesity

    susceptibility.

    4.5. Characterization of associated loci and causal variants

    It is not clear how much of the remaining variation will be uncovered by the aforementioned steps.

    Beyond identifying more susceptibility loci, finding the causal gene(s) and variant(s) in each locus

    will be necessary to allow the detailed molecular and physiological investigations that are necessary

    to determine the mechanisms and pathways through which these contribute to obesity susceptibility

    and ultimately allow this knowledge to be translated into better prediction and treatment options.

    4.6. Integration of genetic and epigenetic information

    Genetic and epigenetic factors are intimately intertwined, as epigenetic marks and DNA modifications

    are the direct consequence of sequence-specific interactions between proteins and DNA [79]. The

    completion of the human epigenome project will increase our understanding of the genetic and

    epigenetics underlying cellular homeostasis and the integration of this knowledge with the known

    environmental risks to obesity, must be applied so that it may eventually be exploited and

    manipulated to appease the obesity epidemic.

    5. Conclusions

    The current obesity epidemic is clearly not of genetic origin per se, but due to unfavourable changes in

    lifestyle and environment (the obesogenic environment). The obesogenic environment has different

    effects on different individuals in the same environment, highlighting an underlying, inherited

    susceptibility to obesity and fat-distribution. For more than a decade, the genetics underlying

    common forms of obesity have remained elusive although the advent of the GWA approach has

    started to deliver robust associations to obesity. The epigenetic contribution to common forms ofobesity are still largely unknown but, from rare syndromes and animal models we conclude that it is

    likely that both genetic and environmental effects on epigenetics will in turn be associated with

    obesity. We have started to identify an emerging pattern of effects acting through CNS, suggesting

    that a component of an individual's response to the obesogenic environment is partly

    neurobehaviourally driven. There is also some evidence of effects acting more peripherally in the

    adipose tissue. Despite the success of GWAs in obesity loci identification, we still only explain a low

    fraction of the inter-individual variation of obesity. Extensive work including identification of more

    obesity susceptibility loci, a better understanding of the gene(s) through which the effect is executed,

    as well as further molecular and physiological characterization of the associated genes, is now

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    2 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    8/22

    necessary before any of these findings will lead to any useful therapeutic interventions.

    Contribu tors and their role

    All the authors were involved in the drafting, editing and approval of this manuscript.

    Blanca M. Herrera and Sarah L. Keildson were involved in the construction of the tables and figures.

    Competing interests

    None.

    Provenance and peer review

    Commissioned and externally peer reviewed.

    Acknow ledgement

    C.M.L. is a Wellcome Trust Research Career Development Fellow (086596/Z/08/Z).

    References

    1. Kelly T., Yang W., Chen C.S., Reynolds K., He J. Global burden of obesity in 2005 and projections to

    2030. Int J Obes (Lond) 2008;32(September (9)):14311437. [PubMed: 18607383]

    2. Flegal K.M., Graubard B.I., Williamson D.F., Gail M.H. Cause-specific excess deaths associated

    with underweight, overweight, and obesity. JAMA. 2007;298(November (17)):20282037.

    [PubMed: 17986696]

    3. Pischon T., Boeing H., Hoffmann K. General and abdominal adiposity and risk of death in Europe.

    N Engl J Med. 2008;359(November (20)):21052120. [PubMed: 19005195]

    4. ORahilly S. Human genetics illuminates the paths to metabolic disease. Nature.

    2009;462(November (7271)):307314. [PubMed: 19924209]

    5. Stunkard A.J., Foch T.T., Hrubec Z. A twin study of human obesity. JAMA. 1986;256(July(1)):5154. [PubMed: 3712713]

    6. Turula M., Kaprio J., Rissanen A., Koskenvuo M. Body weight in the finnish twin cohort. Diabetes

    Res Clin Pract. 1990;10 (Suppl. 1):S33S36. [PubMed: 2286148]

    7. Wardle J., Carnell S., Haworth C.M., Plomin R. Evidence for a strong genetic influence on

    childhood adiposity despite the force of the obesogenic environment. Am J Clin Nutr.

    2008;87(February (2)):398404. [PubMed: 18258631]

    8. Malis C., Rasmussen E.L., Poulsen P. Total and regional fat distribution is strongly influenced by

    genetic factors in young and elderly twins. Obes Res. 2005;13(December (12)):21392145.

    [PubMed: 16421348]

    9. Moll P.P., Burns T.L., Lauer R.M. The genetic and environmental sources of body mass index

    variability: the muscatine ponderosity family study. Am J Hum Genet. 1991;49(December

    (6)):12431255. [PMCID: PMC1686453] [PubMed: 1746554]

    10. Rose K.M., Newman B., Mayer-Davis E.J., Selby J.V. Genetic and behavioral determinants of

    waisthip ratio and waist circumference in women twins. Obes Res. 1998;6(November (6)):383392.

    [PubMed: 9845227]

    11. Selby J.V., Newman B., Quesenberry C.P., Jr., Fabsitz R.R., King M.C., Meaney F.J. Evidence of

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    2 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    9/22

    genetic influence on central body fat in middle-aged twins. Hum Biol. 1989;61(April (2)):179194.

    [PubMed: 2767669]

    12. Stunkard A.J., Sorensen T.I., Hanis C. An adoption study of human obesity. N Engl J Med.

    1986;314(January (4)):193198. [PubMed: 3941707]

    13. Redden D.T., Divers J., Vaughan L.K. Regional admixture mapping and structured association

    testing: conceptual unification and an extensible general linear model. PLoS Genet. 2006;2(August

    (8)):e137. [PMCID: PMC1557785] [PubMed: 16934005]

    14. Williams R.C., Long J.C., Hanson R.L., Sievers M.L., Knowler W.C. Individual estimates of

    European genetic admixture associated with lower body-mass index, plasma glucose, and prevalence

    of type 2 diabetes in Pima Indians. Am J Hum Genet. 2000;66(February (2)):527538.

    [PMCID: PMC1288106] [PubMed: 10677313]

    15. Rankinen T., Zuberi A., Chagnon Y.C. The human obesity gene map: the 2005 update. Obesity

    (Silver Spring) 2006;14(April (4)):529644. [PubMed: 16741264]

    16. Saunders C.L., Chiodini B.D., Sham P. Meta-analysis of genome-wide linkage studies in BMI and

    obesity. Obesity (Silver Spring) 2007;15(September (9)):22632275. [PubMed: 17890495]

    17. A haplotype map of the human genome. Nature. 2005;437(October (7063)):12991320.[PMCID: PMC1880871] [PubMed: 16255080]

    18. Barrett J.C., Cardon L.R. Evaluating coverage of genome-wide association studies. Nat Genet.

    2006;38(June (6)):659662. [PubMed: 16715099]

    19. Peer I., de Bakker P.I., Maller J., Yelensky R., Altshuler D., Daly M.J. Evaluating and improving

    power in whole-genome association studies using fixed marker sets. Nat Genet. 2006;38(June

    (6)):663667. [PubMed: 16715096]

    20. Fan J.B., Chee M.S., Gunderson K.L. Highly parallel genomic assays. Nat Rev Genet.

    2006;7(August (8)):632644. [PubMed: 16847463]

    21. Frayling T.M., Timpson N.J., Weedon M.N. A common variant in the FTO gene is associated with

    body mass index and predisposes to childhood and adult obesity. Science. 2007;316(May

    (5826)):889894. [PMCID: PMC2646098] [PubMed: 17434869]

    22. Zeggini E., Weedon M.N., Lindgren C.M. Replication of genome-wide association signals in UK

    samples reveals risk loci for type 2 diabetes. Science. 2007;316(June (5829)):13361341.

    [PMCID: PMC3772310] [PubMed: 17463249]

    23. Gerken T., Girard C.A., Tung Y.C. The obesity-associated FTO gene encodes a 2-oxoglutarate-

    dependent nucleic acid demethylase. Science. 2007;318(November (5855)):14691472.

    [PMCID: PMC2668859] [PubMed: 17991826]

    24. Wahlen K., Sjolin E., Hoffstedt J. The common rs9939609 gene variant of the fat mass- and

    obesity-associated gene FTO is related to fat cell lipolysis. J Lipid Res. 2008;49(March (3)):607611.

    [PubMed: 18048838]

    25. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared

    controls. Nature. 2007;447(June (7145)):661678. [PMCID: PMC2719288] [PubMed: 17554300]

    26. Dina C., Meyre D., Gallina S. Variation in FTO contributes to childhood obesity and severe adult

    obesity. Nat Genet. 2007;39(June (6)):724726. [PubMed: 17496892]

    27. Scuteri A., Sanna S., Chen W.M. Genome-wide association scan shows genetic variants in the FTO

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    2 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    10/22

    gene are associated with obesity-related traits. PLoS Genet. 2007;3(July (7)):e115.

    [PMCID: PMC1934391] [PubMed: 17658951]

    28. Loos R.J., Lindgren C.M., Li S. Common variants near MC4R are associated with fat mass, weight

    and risk of obesity. Nat Genet. 2008;40(June (6)):768775. [PMCID: PMC2669167]

    [PubMed: 18454148]

    29. Chambers J.C., Elliott P., Zabaneh D. Common genetic variation near MC4R is associated with

    waist circumference and insulin resistance. Nat Genet. 2008;40(June (6)):716718.

    [PubMed: 18454146]

    30. Qi L., Kraft P., Hunter D.J., Hu F.B. The common obesity variant near MC4R gene is associated

    with higher intakes of total energy and dietary fat, weight change and diabetes risk in women. Hum

    Mol Genet. 2008;17(November (22)):35023508. [PMCID: PMC2572696] [PubMed: 18697794]

    31. Farooqi I.S., Keogh J.M., Yeo G.S., Lank E.J., Cheetham T., ORahilly S. Clinical spectrum of

    obesity and mutations in the melanocortin 4 receptor gene. N Engl J Med. 2003;348(March

    (12)):10851095. [PubMed: 12646665]

    32. Willer C.J., Speliotes E.K., Loos R.J. Six new loci associated with body mass index highlight a

    neuronal influence on body weight regulation. Nat Genet. 2009;41(January (1)):2534.

    [PMCID: PMC2695662] [PubMed: 19079261]

    33. Thorleifsson G., Walters G.B., Gudbjartsson D.F. Genome-wide association yields new sequence

    variants at seven loci that associate with measures of obesity. Nat Genet. 2009;41(January

    (1)):1824. [PubMed: 19079260]

    34. Meyre D., Delplanque J., Chevre J.C. Genome-wide association study for early-onset and morbid

    adult obesity identifies three new risk loci in European populations. Nat Genet. 2009;41(February

    (2)):157159. [PubMed: 19151714]

    35. Cotsapas C., Speliotes E.K., Hatoum I.J. Common body mass index-associated variants confer risk

    of extreme obesity. Hum Mol Genet. 2009;18(September (18)):35023507. [PMCID: PMC2729668][PubMed: 19553259]

    36. Scherag A., Dina C., Hinney A. 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. [PMCID: PMC2858696] [PubMed: 20421936]

    37. Speliotes E.K., Willer C.J., Berndt S.I. Association analyses of 249,796 individuals reveal 18 new

    loci associated with body mass index. Nat Genet. 2010;42(November (11)):937948.

    [PMCID: PMC3014648] [PubMed: 20935630]

    38. Farooqi I.S., Drop S., Clements A. Heterozygosity for a POMC-null mutation and increased obesity

    risk in humans. Diabetes. 2006;55(September (9)):25492553. [PubMed: 16936203]

    39. Lindgren C.M., Heid I.M., Randall J.C. Genome-wide association scan meta-analysis identifies

    three loci influencing adiposity and fat distribution. PLoS Genet. 2009;5(June (6)):e1000508.

    [PMCID: PMC2695778] [PubMed: 19557161]

    40. Heard-Costa N.L., Zillikens M.C., Monda K.L. NRXN3 is a novel locus for waist circumference: a

    genome-wide association study from the CHARGE consortium. PLoS Genet. 2009;5(June

    (6)):e1000539. [PMCID: PMC2695005] [PubMed: 19557197]

    41. Kamel E.G., McNeill G., Van Wijk M.C. Usefulness of anthropometry and DXA in predicting intra-

    abdominal fat in obese men and women. Obes Res. 2000;8(January (1)):3642. [PubMed: 10678257]

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    22 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    11/22

    42. Selby J.V., Newman B., Quesenberry C.P., Jr. Genetic and behavioral influences on body fat

    distribution. Int J Obes. 1990;14(July (7)):593602. [PubMed: 2228394]

    43. Zillikens M.C., Yazdanpanah M., Pardo L.M. Sex-specific genetic effects influence variation in

    body composition. Diabetologia. 2008;51(December (12)):22332241. [PubMed: 18839131]

    44. Heid I.M., Jackson A.U., Randall J.C. Meta-analysis identifies 13 new loci associated with

    waisthip ratio and reveals sexual dimorphism in the genetic basis of fat distribution. Nat Genet.

    2010;42(November (11)):949960. [PMCID: PMC3000924] [PubMed: 20935629]

    45. Garg A. Acquired and inherited lipodystrophies. N Engl J Med. 2004;350(March (12)):12201234.

    [PubMed: 15028826]

    46. Cho Y.S., Go M.J., Kim Y.J. A large-scale genome-wide association study of Asian populations

    uncovers genetic factors influencing eight quantitative traits. Nat Genet. 2009;41(May (5)):527534.

    [PubMed: 19396169]

    47. Bird A. Perceptions of epigenetics. Nature. 2007;447(May (7143)):396398. [PubMed: 17522671]

    48. Chong S., Youngson N.A., Whitelaw E. Heritable germline epimutation is not the same as

    transgenerational epigenetic inheritance. Nat Genet. 2007;39(May (5)):574575. author reply 56.

    [PubMed: 17460682]

    49. Reik W., Walter J. Imprinting mechanisms in mammals. Curr Opin Genet Dev. 1998;8(April

    (2)):154164. [PubMed: 9610405]

    50. Butler M.G. Genomic imprinting disorders in humans: a mini-review. J Assist Reprod Genet.

    2009;26(SeptemberOctober (910)):477486. [PMCID: PMC2788689] [PubMed: 19844787]

    51. Haig D., Graham C. Genomic imprinting and the strange case of the insulin-like growth factor II

    receptor. Cell. 1991;64(March (6)):10451046. [PubMed: 1848481]

    52. Smith F.M., Garfield A.S., Ward A. Regulation of growth and metabolism by imprinted genes.

    Cytogenet Genome Res. 2006;113(14):279291. [PubMed: 16575191]

    53. Lee M.P., Brandenburg S., Landes G.M., Adams M., Miller G., Feinberg A.P. Two novel genes in

    the center of the 11p15 imprinted domain escape genomic imprinting. Hum Mol Genet. 1999;8(April

    (4)):683690. [PubMed: 10072438]

    54. Schweizer J., Zynger D., Francke U. In vivo nuclease hypersensitivity studies reveal multiple sites

    of parental origin-dependent differential chromatin conformation in the 150 kb SNRPN transcription

    unit. Hum Mol Genet. 1999;8(April (4)):555566. [PubMed: 10072422]

    55. Shapira N.A., Lessig M.C., He A.G., James G.A., Driscoll D.J., Liu Y. Satiety dysfunction in

    PraderWilli syndrome demonstrated by fMRI. J Neurol Neurosurg Psychiatry. 2005;76(February

    (2)):260262. [PMCID: PMC1739487] [PubMed: 15654046]56. Ferguson-Smith A.C., Sasaki H., Cattanach B.M., Surani M.A. Parental-origin-specific epigenetic

    modification of the mouse H19 gene. Nature. 1993;362(April (6422)):751755. [PubMed: 8469285]

    57. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002;16(January

    (1)):621. [PubMed: 11782440]

    58. Choy J.S., Wei S., Lee J.Y., Tan S., Chu S., Lee T.H. DNA methylation increases nucleosome

    compaction and rigidity. J Am Chem Soc. 2010;132(February (6)):17821783. [PubMed: 20095602]

    59. Zhu Q., Wani A.A. Histone modifications: crucial elements for damage response and chromatin

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    22 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    12/22

    restoration. J Cell Physiol. 2010;223(May (2)):283288. [PMCID: PMC2930755]

    [PubMed: 20112283]

    60. Litt M.D., Simpson M., Gaszner M., Allis C.D., Felsenfeld G. Correlation between histone lysine

    methylation and developmental changes at the chicken beta-globin locus. Science.

    2001;293(September (5539)):24532455. [PubMed: 11498546]

    61. Musri M.M., Gomis R., Parrizas M. Chromatin and chromatin-modifying proteins in adipogenesis.

    Biochem Cell Biol. 2007;85(August (4)):397410. [PubMed: 17713575]

    62. Kallin E.M., Cao R., Jothi R. Genome-wide uH2A localization analysis highlights Bmi1-dependent

    deposition of the mark at repressed genes. PLoS Genet. 2009;5(June (6)):e1000506.

    [PMCID: PMC2683938] [PubMed: 19503595]

    63. Lachner M., O'Sullivan R.J., Jenuwein T. An epigenetic road map for histone lysine methylation. J

    Cell Sci. 2003;116(June (Pt 11)):21172124. [PubMed: 12730288]

    64. Feinberg A.P. Epigenetics at the epicenter of modern medicine. JAMA. 2008;299(March

    (11)):13451350. [PubMed: 18349095]

    65. Feinberg A.P. Genome-scale approaches to the epigenetics of common human disease. Virchows

    Arch. 2010;456(January (1)):1321. [PMCID: PMC3107986] [PubMed: 19844740]

    66. Stein A.D., Zybert P.A., van de Bor M., Lumey L.H. Intrauterine famine exposure and body

    proportions at birth: the Dutch hunger winter. Int J Epidemiol. 2004;33(August (4)):831836.

    [PubMed: 15166208]

    67. Dabelea D., Mayer-Davis E.J., Lamichhane A.P. Association of intrauterine exposure to maternal

    diabetes and obesity with type 2 diabetes in youth: the SEARCH casecontrol study. Diabetes Care.

    2008;31(July (7)):14221426. [PMCID: PMC2453655] [PubMed: 18375420]

    68. Smith J., Cianflone K., Biron S. Effects of maternal surgical weight loss in mothers on

    intergenerational transmission of obesity. J Clin Endocrinol Metab. 2009;94(November

    (11)):42754283. [PubMed: 19820018]

    69. Waterland R.A., Jirtle R.L. Early nutrition, epigenetic changes at transposons and imprinted

    genes, and enhanced susceptibility to adult chronic diseases. Nutrition. 2004;20(January (1)):6368.

    [PubMed: 14698016]

    70. Waterland R.A., Michels K.B. Epigenetic epidemiology of the developmental origins hypothesis.

    Annu Rev Nutr. 2007;27:363388. [PubMed: 17465856]

    71. Poirier L.A. The effects of diet, genetics and chemicals on toxicity and aberrant DNA methylation:

    an introduction. J Nutr. 2002;132(August (8 Suppl.)):S2336S2339.

    72. Biniszkiewicz D., Gribnau J., Ramsahoye B. Dnmt1 overexpression causes genomichypermethylation, loss of imprinting, and embryonic lethality. Mol Cell Biol. 2002;22(April

    (7)):21242135. [PMCID: PMC133685] [PubMed: 11884600]

    73. Widiker S., Karst S., Wagener A., Brockmann G.A. High-fat diet leads to a decreased methylation

    of the Mc4r gene in the obese BFMI and the lean B6 mouse lines. J Appl Genet. 2010;51(2):193197.

    [PubMed: 20453306]

    74. Plagemann A., Harder T., Brunn M. Hypothalamic proopiomelanocortin promoter methylation

    becomes altered by early overfeeding: an epigenetic model of obesity and the metabolic syndrome. J

    Physiol. 2009;587(October (Pt 20)):49634976. [PMCID: PMC2770159] [PubMed: 19723777]

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    22 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    13/22

    75. Milagro F.I., Campion J., Garcia-Diaz D.F., Goyenechea E., Paternain L., Martinez J.A. High fat

    diet-induced obesity modifies the methylation pattern of leptin promoter in rats. J Physiol Biochem.

    2009;65(March (1)):19. [PubMed: 19588726]

    76. Manipalviratn S., DeCherney A., Segars J. Imprinting disorders and assisted reproductive

    technology. Fertil Steril. 2009;91(February (2)):305315. [PMCID: PMC3081604]

    [PubMed: 19201275]

    77. Walters R.G., Jacquemont S., Valsesia A. A new highly penetrant form of obesity due to deletions

    on chromosome 16p11.2. Nature. 2010;463(February (7281)):671675. [PMCID: PMC2880448]

    [PubMed: 20130649]

    78. Bochukova E.G., Huang N., Keogh J. Large, rare chromosomal deletions associated with severe

    early-onset obesity. Nature. 2010;463(February (7281)):666670. [PMCID: PMC3108883]

    [PubMed: 19966786]

    79. Ptashne M., Hobert O., Davidson E. Questions over the scientific basis of epigenome project.

    Nature. 2010;464(March (7288)):487. [PubMed: 20336114]

    Figures and Tables

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    22 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    14/22

    Fig. 1

    CpG methylation and regulation of gene expression. Unmethylated or hypomethylated DNA (usually in the

    promoter region) allows binding of the transcription factors (TF) and other regulatory mechanisms which results in

    transcription and mRNA production. Methylated DNA (bottom panel) obstructs binding of the TF, and in some cases

    might recruit methyl-CpG binding proteins and other transcription co-repressors, blocking access of the

    transcription enzymes and resulting in gene silencing.

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    22 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    15/22

    Fig. 2

    Histone modification. This simplified diagram of a nucleosome shows a histone octamer bead surrounded by a

    DNA strand and try-methylation at lysine-9, this kind of modification exemplifies modifications found at promoter

    regions of silenced genes.

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    22 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    16/22

    Table 1

    Overview of GWA scans or meta-analysis thereof for obesity phenotypes.

    Reference Study name

    (if any)

    Number of samples in

    discovery cohort

    Ancestry of discovery

    cohort

    Phenotype

    Frayling et al.

    [20]

    WTCCC 1924 Europeans BMI quantitative analysis

    Scuteri et al.

    [26]

    Sardinia 4741 Europeans BMI waist circumference

    (WC) quantitative analysis

    Chambers et al.

    [28]

    LOLIPOP 2684 Indian Asians Insulin resistance and

    related quantitative

    phenotypes

    Loos et al. [27] 16 876 Northern European BMI quantitative analysis

    Heard-Costa et

    al. [36]

    The CHARGE

    consortium

    31 373 Europeans WC quantitative analysis

    Lindgren et al.

    [35]

    The GIANT

    consortium

    38 580 Europeans WC and waist:hip-ratio

    (WHR) quantitative

    analysis

    Cotsapas et al.

    [33]

    775 cases and 3197

    unascertained controls

    Europeans Extreme obesity/BMI

    Meyre et al.

    [32]

    1380 and 1416 age-matched

    normal-weight control

    Europeans Early onset and morbid

    adult obesity

    Thorleifsson et

    al. [31]

    DeCODE 37 347 Europeans + African

    Americans

    BMI quantitative analysis

    Willer et al.

    [30]

    The GIANT

    consortium

    32 387 Europeans BMI quantitative analysis

    Hinney et al.

    [79]

    487 extremely obese young

    cases and 442 healthy lean

    controls

    Europeans Extreme obesity/BMI

    Scherag et al.

    [34]

    453 extremely obese young

    cases and 435 healthy lean

    controls

    Europeans Extreme obesity/BMI

    Cho et al. [41] KARE 8842 Asian BMI, WHR quantitative

    analysis

    Heid et al. [43] MAGIC 77 167 European WHR quantitative analysis

    Speliotes et al.

    [37]

    123 865 European BMI quantitative analysis

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    22 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    17/22

    Table 2

    The 54 loci associated to anthropometric obesity phenotypes.

    Closest gene(s) Chromosomal

    location

    Phenotype Associated

    lead

    SNP(s)

    Proposed

    molecular or

    cellular function

    Additional

    phenotypes

    References

    TBX15WARS2 1p12 WHR rs984222 Transcription

    factor involved inadipocyte and

    specific adipose

    depot development

    Implicated in

    Cousin syndrome

    Heid et al.

    [43]

    PTBP2 1p21.3 BMI rs1555543 Speliotes et al.

    [37]

    NEGR1 1p31 BMI rs2815752,

    rs3101336,

    rs2568958

    Neuronal

    outgrowth

    Thorleifsson et

    al. [31], Willer

    et al. [30],

    Speliotes et al.

    [37]

    TNNI3K 1p31.1 BMI rs1514175 Speliotes et al.

    [37]

    DNM3PIGC 1q24.3 WHR rs1011731 Dominant, negative

    mutations in DNM

    enzymes promote

    GLUT6 and GLUT8

    transporters to

    adipocyte cell

    surface in rats.

    Heid et al.

    [43]

    SEC16B,RASAL2 1q25 BMI rs10913469 Thorleifsson et

    al. [31],

    Speliotes et al.

    [37]

    LYPLAL1; ZC3H11B 1q41 WHR rs2605100 Encodes protein

    thought to act as

    triglyceride lipase

    and is upregulated

    in subcutaneous

    adipose tissue in

    obese patients

    Lindgren et al.

    [35], Heid et

    al. [43]

    SDCCAG8 1q43q44 BMI rs12145833 Scherag et al.[34]

    FANCL 2p16.1 BMI rs887912 Speliotes et al.

    [37]

    RBJADCY3POMC 2p23.3 BMI rs713586 Rare POMC

    mutations cause

    human obesity

    Speliotes et al.

    [37]

    TMEM18 2p25 BMI rs6548238,

    rs2867125,

    rs4854344,

    Neural

    development

    Associated with

    T2D

    Willer et al.

    [30],

    Thorleifsson et

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    22 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    18/22

    Closest gene(s) Chromosomal

    location

    Phenotype Associated

    lead

    SNP(s)

    Proposed

    molecular or

    cellular function

    Additional

    phenotypes

    References

    rs7561317,

    rs11127485

    al. [31],

    Scherag et al.

    [34], Speliotes

    et al. [37]

    ZNRF3KREMEN1 2q12.1 WHR rs4823006 Kremen1 proteinforms a complex

    with LDL

    receptor-related

    protein 6

    Heid et al.[43]

    LRP1B 2q22.2 BMI rs2890652 LRP1Bdeletions

    seen in several

    types of human

    cancers

    Speliotes et al.

    [37]

    GRB14 2q24.3 WHR rs10195252 Associated with

    triglyceride and

    insulin levels.

    GRB14-deficient

    mice exhibit

    increased body

    weight

    Heid et al.

    [43]

    ADAMTS9 3p14.1 WHR rs6795735 Important for

    spatial distribution

    of cells in

    embryonic

    development

    Associated with

    T2D

    Heid et al.

    [43]

    NISCHSTAB1 3p21.1 WHR rs6784615 Interacts withinsulin receptor

    substrate

    Heid et al.[43]

    CADM2 3p21.1 BMI rs13078807 Speliotes et al.

    [37]

    ETV5(locus with

    three genes,

    strongest association

    in ETV5)

    3q27 BMI rs7647305 Thorleifsson et

    al. [31],

    Speliotes et al.

    [37]

    Gene desert;

    GNPDA2is one of

    three genes nearby

    4p13 BMI rs10938397 Associated with

    T2D

    Willer et al.

    [30], Speliotes

    et al. [37]

    SLC39A8 4q24 BMI rs13107325 Speliotes et al.

    [37]

    FLJ35779 5q13.3 BMI rs2112347 Speliotes et al.

    [37]

    ZNF608 5q23.2 BMI rs4836133 Speliotes et al.

    [37]

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    22 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    19/22

    Closest gene(s) Chromosomal

    location

    Phenotype Associated

    lead

    SNP(s)

    Proposed

    molecular or

    cellular function

    Additional

    phenotypes

    References

    CPEB4 5q35.2 WHR rs6861681 Regulates

    polyadenylation

    elongation

    Heid et al.

    [43]

    TFAP2B 6p12 WC, BMI rs987237 Lindgren et al.

    [35], Spelioteset al. [37]

    Locus containing

    NCR3, AIF1and

    BAT2

    6p21 BMI rs2844479,

    rs2260000,

    rs1077393

    Associated with

    weight, not BMI

    Thorleifsson et

    al. [31]

    VEGFA 6p21.1 WHR rs6905288 Involved in vascular

    development. Key

    mediator of

    adipogenesis

    VEGFAvariants

    nominally

    associated with

    T2D

    Heid et al.

    [43]

    NUDT3HMGA1 6p21.31 BMI rs206936 Speliotes et al.

    [37]

    PRL 6p22.2p21.3 BMI rs4712652 Meyre et al.

    [32]

    LY86 6p25.1 WHR rs1294421 Plays a role in

    recognition of

    lipopolysaccharide

    Associated with

    asthma

    Heid et al.

    [43]

    RSPOS 6q22.33 WHR rs9491696 Promotes

    angiogenesis and

    vascular

    development

    Oncogene in

    mouse mammary

    epithelial cells

    Heid et al.

    [43]

    NFE2L3 7p15.2 WHR rs1055144 Heid et al.

    [43]

    MSRA 8p23.1 WC, BMI rs7826222,

    rs17150703

    Lindgren et al.

    [35], Scherag

    et al. [34]

    LRRN6C 9p21.3 BMI rs10968576 Speliotes et al.

    [37]

    PTER 10p12 BMI rs10508503 Meyre et al.

    [32]

    MTCH2(locus with

    14 genes)

    11p11.2 BMI rs10838738 Cellular apoptosis Willer et al.

    [30], Speliotes

    et al. [37]

    BDNF(locus with

    four genes, strongest

    association near

    BDNF)

    11p14 BMI rs4074134,

    rs4923461,

    rs925946,

    rs10501087,

    rs6265

    BDNFexpression is

    regulated by

    nutritional state

    and MC4R

    signalling

    Associated with

    T2D. Individuals

    with WAGR

    syndrome with

    BDNFdeletion

    have BMI >95th

    percentile.BDNF

    knockdown in

    Thorleifsson et

    al. [31],

    Speliotes et al.

    [37]

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    22 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    20/22

    Closest gene(s) Chromosomal

    location

    Phenotype Associated

    lead

    SNP(s)

    Proposed

    molecular or

    cellular function

    Additional

    phenotypes

    References

    mouse

    hypothalamus

    causes

    hyperphagia and

    obesity

    RPL27A 11p15.4 BMI rs4929949 Speliotes et al.

    [37]

    ITPR2SSPN 12p21.1 WHR rs718314 Mice lacking

    ITPR2and ITPR3

    exhibited

    hypoglycaemia

    and lean body

    type

    Heid et al.

    [43]

    HOXC13 12q13.13 WHR rs1443512 Transcription

    factor important in

    cell spatial

    distribution in

    embryonic

    development

    Heid et al.

    [43]

    FAIM2(locus also

    containsBCDIN3D)

    12q13 BMI rs7138803 Adipocyte

    apoptosis

    Thorleifsson et

    al. [31],

    Speliotes et al.

    [37]

    C12orf51 12q24 WHR rs2074356 Cho et al. [41]

    MTIF3GTF3A 13q12.2 BMI rs4771122 Speliotes et al.

    [37]

    PRKD1 14q12 BMI rs11847697 Speliotes et al.

    [37]

    NRXN3 14q31 WC, BMI rs10146997 Heard-Costa

    et al. [36],

    Speliotes et al.

    [37]

    MAP2K5 15q23 BMI rs2241423 Speliotes et al.

    [37]

    SH2B1(locus with

    1925 genes)

    16p11.2 BMI rs7498665,

    rs8049439,

    rs4788102,

    rs7498665

    Neuronal role in

    energy homeostasis

    Sh2b1-null mice

    are obese and

    diabetic

    Willer et al.

    [30],

    Thorleifsson et

    al. [31],

    Speliotes et al.

    [37]

    GPRC5B 16p12.3 BMI rs12444979 Speliotes et al.

    [37]

    MAF 16q22q23 BMI rs1424233 Transcription

    factor involved in

    adipogenesis and

    Meyre et al.

    [32]

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    22 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    21/22

    Closest gene(s) Chromosomal

    location

    Phenotype Associated

    lead

    SNP(s)

    Proposed

    molecular or

    cellular function

    Additional

    phenotypes

    References

    insulinglucagon

    regulation

    FTO 16q22.2 BMI rs9939609,

    rs6499640,

    rs8050136,rs3751812,

    rs7190492,

    rs8044769,

    rs1558902

    Neuronal function

    associated with

    control of appetite

    Associated with

    T2D

    Frayling et al.

    [20], Scuteri

    et al. [26],Chambers et

    al. [28], Willer

    et al. [30],

    Thorleifsson et

    al. [31], Meyre

    et al. [32],

    Scherag et al.

    [34], Speliotes

    et al. [37]

    NPC1 18q11.2 BMI rs1805081 Intracellular lipid

    transport

    NPC1-null mice

    show late-onset

    weight loss and

    poor food intake.

    NPC1interferes

    with function of

    raft-associated

    insulin receptor

    signalling

    Meyre et al.

    [32]

    MC4R 18q22 BMI rs17782313,

    rs12970134,

    rs17700144

    Hypothalamic

    signalling

    Haplo-

    insufficiency in

    humans is

    associated with

    morbid obesity.

    MC4R-deficient

    mice show

    hyperphagia and

    obesity

    Willer et al.

    [30],

    Thorleifsson et

    al. [31], Meyre

    et al. [32],

    Loos et al.

    [27],

    Chambers et

    al. [28],

    Scherag et al.

    [34], Speliotes

    et al. [37]

    KCTD15 19q13.11 BMI rs11084753,

    rs29941

    Willer et al.

    [30],

    Thorleifsson et

    al. [31],

    Speliotes et al.

    [37]

    QPTCL-GIPR 19q13.32 BMI rs2287019 Encodes incretin

    receptor

    Associated with

    fasting and 2-h

    glucose

    Speliotes et al.

    [37]

    TMEM160 19q13.32 BMI rs3810291 Speliotes et al.

    [37]

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=

    22 5.1.20

  • 7/22/2019 Genetics and Epigenetics of Obesity

    22/22

    tics and epigenetics of obesity http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3213306/?report=