Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Apr 1.
Published in final edited form as: Nature. 2015 Feb 12;518(7538):197–206. doi: 10.1038/nature14177

Genetic studies of body mass index yield new insights for obesity biology

Adam E Locke 1,#, Bratati Kahali 2,#, Sonja I Berndt 3,#, Anne E Justice 4,#, Tune H Pers 5,6,7,8,#, Felix R Day 9, Corey Powell 2, Sailaja Vedantam 5,6, Martin L Buchkovich 10, Jian Yang 11,12, Damien C Croteau-Chonka 10,13, Tonu Esko 5,6,7,14, Tove Fall 15,16,17, Teresa Ferreira 18, Stefan Gustafsson 16,17, Zoltán Kutalik 19,20,21, Jian’an Luan 9, Reedik Mägi 14,18, Joshua C Randall 18,22, Thomas W Winkler 23, Andrew R Wood 24, Tsegaselassie Workalemahu 25, Jessica D Faul 26, Jennifer A Smith 27, Jing Hua Zhao 9, Wei Zhao 27, Jin Chen 28, Rudolf Fehrmann 29, Åsa K Hedman 16,17,18, Juha Karjalainen 29, Ellen M Schmidt 30, Devin Absher 31, Najaf Amin 32, Denise Anderson 33, Marian Beekman 34,35, Jennifer L Bolton 36, Jennifer L Bragg-Gresham 1,37, Steven Buyske 38,39, Ayse Demirkan 32,40, Guohong Deng 41,42,43, Georg B Ehret 44,45, Bjarke Feenstra 46, Mary F Feitosa 47, Krista Fischer 14, Anuj Goel 18,48, Jian Gong 49, Anne U Jackson 1, Stavroula Kanoni 50, Marcus E Kleber 51,52, Kati Kristiansson 53, Unhee Lim 54, Vaneet Lotay 55, Massimo Mangino 56, Irene Mateo Leach 57, Carolina Medina-Gomez 58,59,60, Sarah E Medland 61, Michael A Nalls 62, Cameron D Palmer 5,6, Dorota Pasko 24, Sonali Pechlivanis 63, Marjolein J Peters 58,60, Inga Prokopenko 18,64,65, Dmitry Shungin 66,67,68, Alena Stančáková 69, Rona J Strawbridge 70, Yun Ju Sung 71, Toshiko Tanaka 72, Alexander Teumer 73, Stella Trompet 74,75, Sander W van der Laan 76, Jessica van Setten 77, Jana V Van Vliet-Ostaptchouk 78, Zhaoming Wang 3,79, Loïc Yengo 80,81,82, Weihua Zhang 41,83, Aaron Isaacs 32,84, Eva Albrecht 85, Johan Ärnlöv 16,17,86, Gillian M Arscott 87, Antony P Attwood 88,89, Stefania Bandinelli 90, Amy Barrett 64, Isabelita N Bas 91, Claire Bellis 92,93, Amanda J Bennett 64, Christian Berne 94, Roza Blagieva 95, Matthias Blüher 96,97, Stefan Böhringer 34,98, Lori L Bonnycastle 99, Yvonne Böttcher 96, Heather A Boyd 46, Marcel Bruinenberg 100, Ida H Caspersen 101, Yii-Der Ida Chen 102,103, Robert Clarke 104, E Warwick Daw 47, Anton J M de Craen 75, Graciela Delgado 51, Maria Dimitriou 105, Alex S F Doney 106, Niina Eklund 53,107, Karol Estrada 6,60,108, Elodie Eury 80,81,82, Lasse Folkersen 70, Ross M Fraser 36, Melissa E Garcia 109, Frank Geller 46, Vilmantas Giedraitis 110, Bruna Gigante 111, Alan S Go 112, Alain Golay 113, Alison H Goodall 114,115, Scott D Gordon 61, Mathias Gorski 23,116, Hans-Jörgen Grabe 117,118, Harald Grallert 85,119,120, Tanja B Grammer 51, Jürgen Gräßler 121, Henrik Grönberg 15, Christopher J Groves 64, Gaëlle Gusto 122, Jeffrey Haessler 49, Per Hall 15, Toomas Haller 14, Goran Hallmans 123, Catharina A Hartman 124, Maija Hassinen 125, Caroline Hayward 126, Nancy L Heard-Costa 127,128, Quinta Helmer 34,98,129, Christian Hengstenberg 130,131, Oddgeir Holmen 132, Jouke-Jan Hottenga 133, Alan L James 134,135, Janina M Jeff 55, Åsa Johansson 136, Jennifer Jolley 88,89, Thorhildur Juliusdottir 18, Leena Kinnunen 53, Wolfgang Koenig 52, Markku Koskenvuo 137, Wolfgang Kratzer 138, Jaana Laitinen 139, Claudia Lamina 140, Karin Leander 111, Nanette R Lee 91, Peter Lichtner 141, Lars Lind 142, Jaana Lindström 53, Ken Sin Lo 143, Stéphane Lobbens 80,81,82, Roberto Lorbeer 144, Yingchang Lu 55,145, François Mach 45, Patrik K E Magnusson 15, Anubha Mahajan 18, Wendy L McArdle 146, Stela McLachlan 36, Cristina Menni 56, Sigrun Merger 95, Evelin Mihailov 14,147, Lili Milani 14, Alireza Moayyeri 56,148, Keri L Monda 4,149, Mario A Morken 99, Antonella Mulas 150, Gabriele Müller 151, Martina Müller-Nurasyid 85,130,152,153, Arthur W Musk 154, Ramaiah Nagaraja 155, Markus M Nöthen 156,157, Ilja M Nolte 158, Stefan Pilz 159,160, Nigel W Rayner 18,22,64, Frida Renstrom 66, Rainer Rettig 161, Janina S Ried 85, Stephan Ripke 108,162, Neil R Robertson 18,64, Lynda M Rose 163, Serena Sanna 150, Hubert Scharnagl 164, Salome Scholtens 100, Fredrick R Schumacher 165, William R Scott 41,83, Thomas Seufferlein 138, Jianxin Shi 166, Albert Vernon Smith 167,168, Joanna Smolonska 29,169, Alice V Stanton 170, Valgerdur Steinthorsdottir 171, Kathleen Stirrups 22,50, Heather M Stringham 1, Johan Sundström 142, Morris A Swertz 29, Amy J Swift 99, Ann-Christine Syvänen 16,172, Sian-Tsung Tan 41,173, Bamidele O Tayo 174, Barbara Thorand 120,175, Gudmar Thorleifsson 171, Jonathan P Tyrer 176, Hae-Won Uh 34,98, Liesbeth Vandenput 177, Frank C Verhulst 178, Sita H Vermeulen 179,180, Niek Verweij 57, Judith M Vonk 169, Lindsay L Waite 31, Helen R Warren 181, Dawn Waterworth 182, Michael N Weedon 24, Lynne R Wilkens 54, Christina Willenborg 183,184, Tom Wilsgaard 185, Mary K Wojczynski 47, Andrew Wong 186, Alan F Wright 126, Qunyuan Zhang 47; The LifeLines Cohort Study262,, Eoin P Brennan 187, Murim Choi 188, Zari Dastani 189, Alexander W Drong 18, Per Eriksson 70, Anders Franco-Cereceda 190, Jesper R Gådin 70, Ali G Gharavi 191, Michael E Goddard 192,193, Robert E Handsaker 6,7, Jinyan Huang 194,195, Fredrik Karpe 64,196, Sekar Kathiresan 6,197, Sarah Keildson 18, Krzysztof Kiryluk 191, Michiaki Kubo 198, Jong-Young Lee 199,, Liming Liang 194,200, Richard P Lifton 201, Baoshan Ma 194,202, Steven A McCarroll 6,7,162, Amy J McKnight 203, Josine L Min 146, Miriam F Moffatt 173, Grant W Montgomery 61, Joanne M Murabito 127,204, George Nicholson 205,206, Dale R Nyholt 61,207, Yukinori Okada 208,209, John R B Perry 18,24,56, Rajkumar Dorajoo 210, Eva Reinmaa 14, Rany M Salem 5,6,7, Niina Sandholm 211,212,213, Robert A Scott 9, Lisette Stolk 34,60, Atsushi Takahashi 208, Toshihiro Tanaka 209,214,215, Ferdinand M van ’t Hooft 70, Anna A E Vinkhuyzen 11, Harm-Jan Westra 29, Wei Zheng 216, Krina T Zondervan 18,217; The ADIPOGen Consortium262,; The AGEN-BMI Working Group262,; The CARDIOGRAMplusC4D Consortium262,; The CKDGen Consortium262,; The GLGC262,; The ICBP262,; The MAGIC Investigators262,; The MuTHER Consortium262,; The MIGen Consortium262,; The PAGE Consortium262,; The ReproGen Consortium262,; The GENIE Consortium262,; The International Endogene Consortium262,, Andrew C Heath 218, Dominique Arveiler 219, Stephan J L Bakker 220, John Beilby 87,221, Richard N Bergman 222, John Blangero 92, Pascal Bovet 223,224, Harry Campbell 36, Mark J Caulfield 181, Giancarlo Cesana 225, Aravinda Chakravarti 44, Daniel I Chasman 163,226, Peter S Chines 99, Francis S Collins 99, Dana C Crawford 227,228, L Adrienne Cupples 127,229, Daniele Cusi 230,231, John Danesh 232, Ulf de Faire 111, Hester M den Ruijter 76,233, Anna F Dominiczak 234, Raimund Erbel 235, Jeanette Erdmann 183,184, Johan G Eriksson 53,236,237, Martin Farrall 18,48, Stephan B Felix 238,239, Ele Ferrannini 240,241, Jean Ferrières 242, Ian Ford 243, Nita G Forouhi 9, Terrence Forrester 244, Oscar H Franco 58,59, Ron T Gansevoort 220, Pablo V Gejman 245, Christian Gieger 85, Omri Gottesman 55, Vilmundur Gudnason 167,168, Ulf Gyllensten 136, Alistair S Hall 246, Tamara B Harris 109, Andrew T Hattersley 247, Andrew A Hicks 248, Lucia A Hindorff 249, Aroon D Hingorani 250, Albert Hofman 58,59, Georg Homuth 73, G Kees Hovingh 251, Steve E Humphries 252, Steven C Hunt 253, Elina Hyppönen 254,255,256,257, Thomas Illig 119,258, Kevin B Jacobs 3,79, Marjo-Riitta Jarvelin 83,259,260,261,263,263, Karl-Heinz Jöckel 63, Berit Johansen 101, Pekka Jousilahti 53, J Wouter Jukema 74,264,265, Antti M Jula 53, Jaakko Kaprio 53,107,137, John J P Kastelein 251, Sirkka M Keinanen-Kiukaanniemi 263,266, Lambertus A Kiemeney 179,267, Paul Knekt 53, Jaspal S Kooner 41,173,268, Charles Kooperberg 49, Peter Kovacs 96,97, Aldi T Kraja 47, Meena Kumari 269,270, Johanna Kuusisto 271, Timo A Lakka 125,272,273, Claudia Langenberg 9,269, Loic Le Marchand 54, Terho Lehtimäki 274, Valeriya Lyssenko 275,276, Satu Männistö 53, André Marette 277,278, Tara C Matise 39, Colin A McKenzie 244, Barbara McKnight 279, Frans L Moll 280, Andrew D Morris 106, Andrew P Morris 14,18,281, Jeffrey C Murray 282, Mari Nelis 14, Claes Ohlsson 177, Albertine J Oldehinkel 124, Ken K Ong 9,186, Pamela A F Madden 218, Gerard Pasterkamp 76, John F Peden 283, Annette Peters 119,130,175, Dirkje S Postma 284, Peter P Pramstaller 248,285, Jackie F Price 36, Lu Qi 13,25, Olli T Raitakari 286,287, Tuomo Rankinen 288, D C Rao 47,71,218, Treva K Rice 71,218, Paul M Ridker 163,226, John D Rioux 143,289, Marylyn D Ritchie 290, Igor Rudan 36,291, Veikko Salomaa 53, Nilesh J Samani 114,115, Jouko Saramies 292, Mark A Sarzynski 288, Heribert Schunkert 130,131, Peter E H Schwarz 121,293, Peter Sever 294, Alan R Shuldiner 295,296,297, Juha Sinisalo 298, Ronald P Stolk 169, Konstantin Strauch 85,153, Anke Tönjes 96,97, David-Alexandre Trégouët 299,300,301, Angelo Tremblay 302, Elena Tremoli 303, Jarmo Virtamo 53, Marie-Claude Vohl 278,304, Uwe Völker 73,239, Gérard Waeber 305, Gonneke Willemsen 133, Jacqueline C Witteman 59, M Carola Zillikens 58,60, Linda S Adair 306, Philippe Amouyel 307, Folkert W Asselbergs 250,264,308, Themistocles L Assimes 309, Murielle Bochud 223,224, Bernhard O Boehm 310,311, Eric Boerwinkle 312, Stefan R Bornstein 121, Erwin P Bottinger 55, Claude Bouchard 288, Stéphane Cauchi 80,81,82, John C Chambers 41,83,268, Stephen J Chanock 3, Richard S Cooper 174, Paul I W de Bakker 77,313,314, George Dedoussis 105, Luigi Ferrucci 72, Paul W Franks 25,66,67, Philippe Froguel 65,80,81,82, Leif C Groop 107,276, Christopher A Haiman 165, Anders Hamsten 70, Jennie Hui 87,221,315, David J Hunter 13,25,194, Kristian Hveem 132, Robert C Kaplan 316, Mika Kivimaki 269, Diana Kuh 186, Markku Laakso 271, Yongmei Liu 317, Nicholas G Martin 61, Winfried März 51,164,318, Mads Melbye 309,319, Andres Metspalu 14,147, Susanne Moebus 63, Patricia B Munroe 181, Inger Njølstad 185, Ben A Oostra 32,84,320, Colin N A Palmer 106, Nancy L Pedersen 15, Markus Perola 14,53,107, Louis Pérusse 278,302, Ulrike Peters 49, Chris Power 257, Thomas Quertermous 309, Rainer Rauramaa 125,273, Fernando Rivadeneira 58,59,60, Timo E Saaristo 321,322, Danish Saleheen 232,323,324, Naveed Sattar 325, Eric E Schadt 326, David Schlessinger 155, P Eline Slagboom 34,35, Harold Snieder 169, Tim D Spector 56, Unnur Thorsteinsdottir 171,327, Michael Stumvoll 96,97, Jaakko Tuomilehto 53,328,329,330, André G Uitterlinden 58,59,60, Matti Uusitupa 331,332, Pim van der Harst 29,57,264, Mark Walker 333, Henri Wallaschofski 239,334, Nicholas J Wareham 9, Hugh Watkins 18,48, David R Weir 26, H-Erich Wichmann 335,336,337, James F Wilson 36, Pieter Zanen 338, Ingrid B Borecki 47, Panos Deloukas 22,50,339, Caroline S Fox 127, Iris M Heid 23,85, Jeffrey R O’Connell 295,296, David P Strachan 340, Kari Stefansson 171,327, Cornelia M van Duijn 32,58,59,84, Gonçalo R Abecasis 1, Lude Franke 29, Timothy M Frayling 24, Mark I McCarthy 18,64,341, Peter M Visscher 11,12, André Scherag 63,342, Cristen J Willer 28,30,343, Michael Boehnke 1, Karen L Mohlke 10, Cecilia M Lindgren 6,18, Jacques S Beckmann 20,21,344, Inês Barroso 22,345,346, Kari E North 4,347,§, Erik Ingelsson 16,17,18,§, Joel N Hirschhorn 5,6,7,§, Ruth J F Loos 9,55,145,348,§, Elizabeth K Speliotes 2,§
PMCID: PMC4382211  NIHMSID: NIHMS668049  PMID: 25673413

Abstract

Obesity is heritable and predisposes to many diseases. To understand the genetic basis of obesity better, here we conduct a genome-wide association study and Metabochip meta-analysis of body mass index (BMI), a measure commonly used to define obesity and assess adiposity, in up to 339,224 individuals. This analysis identifies 97 BMI-associated loci (P < 5 × 10−8), 56 of which are novel. Five loci demonstrate clear evidence of several independent association signals, and many loci have significant effects on other metabolic phenotypes. The 97 loci account for ~2.7% of BMI variation, and genome-wide estimates suggest that common variation accounts for >20% of BMI variation. Pathway analyses provide strong support for a role of the central nervous system in obesity susceptibility and implicate new genes and pathways, including those related to synaptic function, glutamate signalling, insulin secretion/action, energy metabolism, lipid biology and adipogenesis.


Obesity is a worldwide epidemic associated with increased morbidity and mortality that imposes an enormous burden on individual and public health. Around 40–70% of inter-individual variability in BMI, commonly used to assess obesity, has been attributed to genetic factors13. At least 77 loci have previously been associated with an obesity measure4, 32 loci from our previous meta-analysis of BMI genome-wide association studies (GWAS)5. Nevertheless, most of the genetic variability in BMI remains unexplained. Moreover, although analyses of previous genetic association results have suggested intriguing biological processes underlying obesity susceptibility, few specific genes supported these pathways5,6. For the vast majority of loci, the probable causal gene(s) and pathways remain unknown.

To expand the catalogue of BMI susceptibility loci and gain a better understanding of the genes and biological pathways influencing obesity, we performed the largest GWAS meta-analysis for BMI so far. This work doubles the number of individuals contributing GWAS results, incorporates results from >100,000 individuals genotyped with Metabochip7, and nearly doubles the number of BMI-associated loci. Comprehensive assessment of meta-analysis results provides several lines of evidence supporting candidate genes at many loci and highlights pathways that reinforce and expand our understanding of biological processes underlying obesity.

Identification of 97 genome-wide significant loci

This BMI meta-analysis included association results for up to 339,224 individuals from 125 studies, 82 with GWAS results (n = 236,231) and 43 with results from Metabochip (n = 103,047; Extended Data Table 1 and Supplementary Tables 13). After regression on age and sex and inverse normal transformation of the residuals, we carried out association analyses with genotypes or imputed genotype dosages. GWAS were meta-analysed together, as were Metabochip studies, followed by a combined GWAS plus Metabochip meta-analysis. In total, we analysed data from 322,154 individuals of European descent and 17,072 individuals of non-European descent (Extended Data Fig. 1).

Our primary meta-analysis of European-descent individuals from GWAS and Metabochip studies (n = 322,154) identified 77 loci reaching genome-wide significance (GWS) and separated by at least 500 kilo-bases (kb) (Table 1, Extended Data Table 2 and Supplementary Figs 1 and 2). We carried out additional analyses to explore the effects of power and heterogeneity. The inclusion of 17,072 non-European-descent individuals (total n = 339,224) identified ten more loci, while secondary analyses identified another ten GWS loci (Table 2, Supplementary Tables 48 and Supplementary Figs 39). Of the 97 BMI-associated loci, 41 have previously been associated with one or more obesity measure5,812. Thus, our current analyses identified 56 novel loci associated with BMI (Tables 1 and 2 and Extended Data Table 2).

Table 1.

Novel GWS BMI loci In European meta-analysis

SNP Chr:position Notable gene(s)* Alleles EAF β s.e.m. P value
rs657452 1:49,362,434 AGBL4(N) A/G 0.394 0.023 0.003 5.48 × 10−13
rs12286929 11:114,527,614 CADM1(N) G/A 0.523 0.022 0.003 1.31 × 10−12
rs7903146 10:114,748,339 TCF7L2(B,N) C/T 0.713 0.023 0.003 1.11 × 10−11
rs10132280 14:24,998,019 STXBP6(N) C/A 0.682 0.023 0.003 1.14 × 10−11
rs17094222 10:102,385,430 HIF1AN(N) C/T 0.211 0.025 0.004 5.94 × 10−11
rs7599312 2:213,121,476 ERBB4(D,N) G/A 0.724 0.022 0.003 1.17 × 10−10
rs2365389 3:61,211,502 FHIT(N) C/T 0.582 0.020 0.003 1.63 × 10−10
rs2820292 1:200,050,910 NAV1(N) C/A 0.555 0.020 0.003 1.83 × 10−10
rs12885454 14:28,806,589 PRKD1(N) C/A 0.642 0.021 0.003 1.94 × 10−10
rs16851483 3:142,758,126 RASA2(N) T/G 0.066 0.048 0.008 3.55 × 10−10
rs1167827 7:75,001,105 HIP1(B,N); PMS2L3(B,Q); PMS2P5(Q);
WBSCR16(Q)
G/A 0.553 0.020 0.003 6.33 × 10−10
rs758747 16:3,567,359 NLRC3(N) T/C 0.265 0.023 0.004 7.47 × 10−10
rs1928295 9:119,418,304 TLR4(B,N) T/C 0.548 0.019 0.003 7.91 × 10−10
rs9925964 16:31,037,396 KAT8(N);ZNF646(M,Q); VKORC1(Q);
ZNF668(Q); STX1B(D); FBXL19(D)
A/G 0.620 0.019 0.003 8.11 × 10−10
rs11126666 2:26,782,315 KCNK3(D,N) A/G 0.283 0.021 0.003 1.33 × 10−9
rs2650492 16:28,240,912 SBK1(D,N); APOBR(B) A/G 0.303 0.021 0.004 1.92 × 10−9
rs6804842 3:25,081,441 RARB(B) G/A 0.575 0.019 0.003 2.48 × 10−9
rs4740619 9:15,624,326 C9orf93(C,M,N) T/C 0.542 0.018 0.003 4.56 × 10−9
rs13191362 6:162,953,340 PARK2(B,D,N) A/G 0.879 0.028 0.005 7.34 × 10−9
rs3736485 15:49,535,902 SCG3(B,D); DMXL2(M,N) A/G 0.454 0.018 0.003 7.41 × 10−9
rs17001654 4:77,348,592 NUP54(M); SCARB2(Q,N) G/C 0.153 0.031 0.005 7.76 × 10−9
rs11191560 10:104,859,028 NT5C2(N); CYP17A1(B); SFXN2(Q) C/T 0.089 0.031 0.005 8.45 × 10−9
rs1528435 2:181,259,207 UBE2E3(N) T/C 0.631 0.018 0.003 1.20 × 10−8
rs1000940 17:5,223,976 RABEP1(N) G/A 0.320 0.019 0.003 1.28 × 10−8
rs2033529 6:40,456,631 TDRG1(N); LRFN2(D) G/A 0.293 0.019 0.003 1.39 × 10−8
rs11583200 1:50,332,407 ELAVL4(B,D,N,Q) C/T 0.396 0.018 0.003 1.48 × 10−8
rs9400239 6:109,084,356 FOXO3(B,N); HSS00296402(Q) C/T 0.688 0.019 0.003 1.61 × 10−8
rs10733682 9:128,500,735 LMX1B(B,N) A/G 0.478 0.017 0.003 1.83 × 10−8
rs11688816 2:62,906,552 EHBP1(B,N) G/A 0.525 0.017 0.003 1.89 × 10−8
rs11057405 12:121,347,850 CLIP1(N) G/A 0.901 0.031 0.006 2.02 × 10−8
rs11727676 4:145,878,514 HHIP(B,N) T/C 0.910 0.036 0.006 2.55 × 10−8
rs3849570 3:81,874,802 GBE1(B,M,N) A/C 0.359 0.019 0.003 2.60 × 10−8
rs6477694 9:110,972,163 EPB41L4B(N); C9orf4(D) C/T 0.365 0.017 0.003 2.67 × 10−8
rs7899106 10:87,400,884 GRID1(B,N) G/A 0.052 0.040 0.007 2.96 × 10−8
rs2176598 11:43,820,854 HSD17B12(B,M,N) T/C 0.251 0.020 0.004 2.97 × 10−8
rs2245368 7:76,446,079 PMS2L11(N) C/T 0.180 0.032 0.006 3.19 × 10−8
rs17724992 19:18,315,825 GDF15(B); PGPEP1(Q,N) A/G 0.746 0.019 0.004 3.42 × 10−8
rs7243357 18:55,034,299 GRP(B,G,N) T/G 0.812 0.022 0.004 3.86 × 10−8
rs2033732 8:85,242,264 RALYL(D,N) C/T 0.747 0.019 0.004 4.89 × 10−8

GWS isdefinedas P < 5 × 10−8. SNP positions are reported according to Build 36 and their alleles are coded based on the positive strand. Alleles (effect/other), effect allele frequency (EAF), beta (β), standard error of the mean (s.e.m.) and P values are based on the meta-analysis of GWAS I + II + Metabochip association data from the European sex-combined data set.

*

Notable genes from biological relevance to obesity (B); copy number variation (C); DEPICT analyses (D); GRAIL results (G); BMI-associated variantis in strong LD (r2 ≥ 0.7) with a missens evariant in the indicated gene (M); gene nearest to index SNP (N); association and eQTL data converge to affect gene expression (Q).

Table 2.

GWS BMI loci from secondary analyses

SNP Chr:position Notable gene(s)* Alleles EAF β s.e.m. P value Analysis
Novel loci
rs9641123 7:93,035,668 CALCR(B,N); hsa-miR-653(Q) C/G 0.430 0.029 0.005 2.08 × 10−10 EPB
rs7164727 15:70,881,044 LOC100287559(N), BBS4(B,M,Q) T/C 0.671 0.019 0.003 3.92 × 10−9 All
rs492400 2:219,057,996 PLCD4(B,Q); CYP27A1(B); USP37(N);
TTLL4(M,Q); STK36(B,M); ZNF142(M);
RQCD1(Q)
C/T 0.424 0.024 0.004 6.78 × 10−9 Men
rs2080454 16:47,620,091 CBLN1(N) C/A 0.413 0.017 0.003 8.60 × 10−9 All
rs7239883 18:38,401,669 LOC284260(N); RIT2(B,D) G/A 0.391 0.023 0.004 1.51 × 10−8 Women
rs2836754 21:39,213,610 ETS2(N) C/T 0.599 0.017 0.003 1.61 × 10−8 All
rs9914578 17:1,951,886 SMG6(D,N); N29617(Q) G/C 0.229 0.020 0.004 2.07 × 10−8 All
rs977747 1:47,457,264 TAL1(N) T/G 0.403 0.017 0.003 2.18 × 10−8 All
rs9374842 6:120,227,364 LOC285762(N); T/C 0.744 0.023 0.004 2.67 × 10−8 EPB
rs4787491 16:29,922,838 MAPK3(D); KCTD13(D); INO80E(N);
TAOK2(D); YPEL3(D); DOC2A(D);
FAM57B(D)
G/A 0.510 0.022 0.004 2.70 × 10−8 EPB
rs1441264 13:78,478,920 MIR548A2(N) A/G 0.613 0.017 0.003 2.96 × 10−8 All
rs17203016 2:207,963,763 CREB1(B,N); KLF7(B) G/A 0.195 0.021 0.004 3.41 × 10−8 All
rs16907751 8:81,538,012 ZBTB10(N) C/T 0.913 0.047 0.009 3.89 × 10−8 Men
rs13201877 6:137,717,234 IFNGR1(N); OLIG3(G) G/A 0.140 0.024 0.004 4.29 × 10−8 All
rs9540493 13:65,103,705 MIR548X2(N); PCDH9(D) A/G 0.452 0.021 0.004 4.97 × 10−8 EPB
rs1460676 2:164,275,935 FIGN(N) C/T 0.179 0.021 0.004 4.98 × 10−8 All
rs6465468 7:95,007,450 ASB4(B,N) T/G 0.306 0.025 0.005 4.98 × 10−8 Women

Previously identified loci

rs6091540 20:50,521,269 ZFP64(N) C/T 0.721 0.030 0.004 2.15 × 10−11 Women
rs7715256 5:153,518,086 GALNT10(N) G/T 0.422 0.017 0.003 8.85 × 10−9 All
rs2176040 2:226,801,046 LOC646736(N); IRS1(B,Q) A/G 0.365 0.024 0.004 9.99 × 10−9 Men

SNP positions are reported according to Build 36 and their alleles are coded based on the positive strand. Alleles (effect/other), EAF, beta (β), s.e.m. and P values are based on the meta-analysis of GWAS I + II+ Metabochip association data from the data set shown in the ‘Analysis’ column. EPB denotes European population-based studies, ‘All’ denotes all ancestries.

*

Notable genes from biological relevance to obesity (B); copy number variation (C); DEPICT analyses (D); GRAIL results (G); BMI-associated variant is in strong LD (r2 ≥ 0.7) with a missense variant in the indicated gene (M); gene nearest to the index SNP (N); association and eQTL data converge to affect gene expression (Q).

Effects of associated loci on BMI

Newly identified loci generally have lower minor allele frequency and/or smaller effect size estimates than previously known loci (Extended Data Fig. 2a, b). On the basis of effect estimates in the discovery data set, which may be inflated owing to winner’s curse, the 97 loci account for 2.7% of BMI phenotypic variance (Supplementary Table 4 and Extended Data Fig. 2a, b). We conservatively used only GWS single nucleotide polymorphisms (SNPs) after strict double genomic control correction, which probably over-corrects association statistics given the lack of evidence for population stratification in family-based analyses13 (Extended Data Fig. 3 and Extended Data Table 1). Polygene analyses suggest that SNPs with P values well below GWS add significantly to the phenotypic variance explained. For example, 2,346 SNPs selected from conditional and joint multiple-SNP analysis with P < 5 × 10−3 explained 6.6 ± 1.1% (mean ± s.e.m.) of variance, compared to 21.6 ± 2.2% explained by all HapMap3 SNPs (31–54% of heritability; Fig. 1a). Furthermore, of 1,909 independent SNPs (pairwise distance >500 kb and r2 < 0.1) included on Metabochip for replication of suggestive BMI associations, 1,458 (76.4%) have directionally consistent effects with our previous GWAS meta-analysis5 and the non-overlapping samples in the current meta-analysis (Extended Data Fig. 2c). On the basis of the significant excess of these directionally consistent observations (sign test P = 2.5 × 10−123), we estimate ~1,007 of the 1,909 SNPs represent true BMI associations.

Figure 1. Cumulative variance explained and example of secondary signals.

Figure 1

a, The estimated variance in BMI explained by SNPs selected at a range of P values using unrelated individuals from the QIMR (n = 3,924; purple) and TwinGene (n = 5,668; gold), their weighted average (cyan), inferred from within-family prediction (red; Extended Data Fig. 2), and by all HapMap phase III SNPs in 16,275 unrelated individuals from the QIMR, TwinGene and ARIC studies (orange). b, Plot of the region surrounding MC4R (ref. 36). SNP associations from the European sex-combined meta-analysis are plotted with joint conditional P values (Pj) indicated for the three conditionally significant signals. SNPs are shaded and shaped based on the index SNP with which they are in strongest LD (rs6567160 in blue, rs994545 in yellow and rs17066842 in green).

We compared the effects of our 97 BMI-associated SNPs between the sexes, between ethnicities, and across several cross-sections of our data (Supplementary Tables 411 and Extended Data Fig. 4). Two previously identified loci, near SEC16B (P = 5.2 × 10−5) and ZFP64 (P = 9.1 × 10−5), showed evidence of heterogeneity between men and women. Both have stronger effects in women (Supplementary Table 10). Two SNPs, near NEGR1 (P = 9.1 × 10−5) and PRKD1 (P = 1.9 × 10−5), exhibited significant evidence for heterogeneity of effect between European- and African-descent samples, and one SNP, near GBE1 (P = 1.3 × 10−4), exhibited evidence for heterogeneity between European and east Asian individuals (Supplementary Table 9). These findings may reflect true heterogeneity at these loci, but are most likely due to linkage disequilibrium (LD) differences across ancestries. Effect estimates for 79% of BMI-associated SNPs in African-descent samples (P = 9.2 × 10−9) and 91% in east Asian samples (P = 1.8 × 10−15) showed directional consistency with our European-only analyses. These results suggest that common BMI-associated SNPs have comparable effects across ancestries and between sexes. In additional heterogeneity analyses, we detected an influence of ascertainment at TCF7L2 (stronger effects in type 2 diabetes case/control studies than in population-based studies); however, we saw no evidence of systematic ascertainment bias at other loci owing to inclusion of case/control studies (Supplementary Tables 10 and 11).

We also took advantage of LD differences across populations to fine-map association signals using Bayesian methods14,15. At 10 of 27 loci fine-mapped for BMI on Metabochip, the addition of non-European individuals into the meta-analysis either narrowed the genomic region containing the 99% credible set, or decreased the number of SNPs in the credible set (Supplementary Table 12 and Supplementary Fig. 10). At the SEC16B and FTO loci, the all ancestries credible set includes a single SNP, although the SNP we highlight at FTO (rs1558902) differs from that identified by a recent fine-mapping effort in African-American cohorts16. Fine-mapping efforts using larger, more diverse study samples and more complete catalogues of variants will help to further narrow association signals.

We examined the combined effects of lead SNPs at the 97 loci in an independent sample of 8,164 European-descent individuals from the Health and Retirement Study17. We observed an average increase of 0.1 BMI units (kg per m2) per BMI-increasing allele, equivalent to 260–320 g for an individual 160–180 cm in height. There was a 1.8 kg per m2 difference in mean BMI between the 145 individuals (1.78%) carrying the most BMI-increasing alleles (>104) and those carrying the mean number of BMI-increasing alleles in the sample (91; Extended Data Fig. 2d), corresponding to a difference of 4.6–5.8 kg for an individual 160–180 cm in height, and a 1.5 kg m−2 difference (3.8–4.9 kg difference) in mean BMI between the 95 individuals (1.16%) carrying the least BMI-increasing alleles (<78) and those carrying the mean number. Such differences are medically significant in predisposing to development of metabolic disease18. For predicting obesity (BMI ≥ 30 kg per m2), adding genetic risk score to a model including age, age squared, sex and four genotype-based principal components slightly, but significantly increases the area under the receiver-operating characteristic curve from 0.576 to 0.601.

Additional associated variants at BMI loci

To identify additional SNPs with independent BMI associations at the 97 established loci, we used genome-wide complex trait analysis (GCTA)19 to perform approximate joint and conditional association analysis20 using summary statistics from European sex-combined meta-analysis after removing family-based validation studies (TwinGene and QIMR). GCTA confirmed two signals at MC4R previously identified using exact conditional analyses5, and identified five loci with evidence of independent associations (Table 3): second signals near LINC01122, NLRC3-ADCY9, GPRC5B-GP2 and BDNF, and a third signal near MC4R (rs9944545, Fig. 1b). Joint conditional analyses at two genomic regions separated by >500 kb (the AGBL4-ELAVL4 regions on chr. 1, and the ATP2A1-SBK1 regions on chr. 16), indicate that these pairs of signals may not be independent owing to extended LD.

Table 3.

Secondary signals reaching GWS by conditional analysis

SNP Chr: position Nearest gene Alleles EAF β s.e.m. Variance explained P value
rs1016287 2:59159129 LINC01122 T/C 0.294 0.023 0.003 0.021% 2.62 × 10−11
rs4671328 2:58788786 LINC01122 T/G 0.457 0.021 0.004 0.021% 2.73 × 10−8

rs758747 16:3567359 NLRC3 T/C 0.241 0.022 0.004 0.018% 2.00 × 10−9
rs879620 16:3955730 ADCY9 T/C 0.620 0.024 0.004 0.027% 2.17 × 10−9

rs12446632 16:19842890 GPRC5B G/A 0.860 0.036 0.005 0.031% 1.06 × 10−14
rs11074446 16:20162624 GP2 T/C 0.867 0.029 0.005 0.019% 1.71 × 10−10

rs6567160 18:55980115 MC4R C/T 0.233 0.048 0.004 0.084% 3.52 × 10−38
rs17066842 18:56191604 MC4R G/A 0.960 0.051 0.008 0.020% 6.99 × 10−10
rs9944545 18:56109224 MC4R T/C 0.296 0.020 0.004 0.017% 1.01 × 10−8

rs11030104 11:27641093 BDNF A/G 0.791 0.051 0.004 0.087% 1.26 × 10−34
rs10835210 11:27652486 BDNF C/A 0.570 0.020 0.004 0.020% 1.25 × 10−8

SNP positions are reported according to Build 36 and their alleles are coded based on the positive strand. Alleles (effect/other), EAF, estimated beta (β), s.e.m., explained variance, and P values from GCTA. First row at each locus represents lead signal, other row(s) represent secondary signals.

Effects of BMI variants on other traits

We tested for associations between our 97 BMI-associated index SNPs and other metabolic phenotypes (Supplementary Tables 1315 and Extended Data Figs 5 and 6). Thirteen of the twenty-three phenotypes tested had significantly more SNPs with effects in the anticipated direction than expected by chance (Supplementary Table 16). These results corroborate the epidemiological relationships of BMI with metabolic traits. Whether this reflects a common genetic aetiology or a causal relationship of BMI on these traits requires further investigation.

Interestingly, some loci showed significant association with traits in the opposite direction than expected based on their phenotypic correlation with BMI (Extended Data Fig. 5). For example, at HHIP, the BMI-increasing allele is associated with decreased type 2 diabetes risk and higher high-density lipoprotein cholesterol (HDL). At LOC646736 and IRS1, the BMI-increasing allele is associated with reduced risk of coronary artery disease (CAD) and diabetic nephropathy, decreased triglyceride levels, increased HDL, higher adiponectin, and lower fasting insulin. This may be due to increased subcutaneous fat and possible production of metabolic mediators protective against the development of metabolic disease despite increased adiposity8. These unexpected associations may help us to understand better the complex pathophysiology underlying these traits, and may indicate benefits or side effects if these regions contain targets of therapeutic intervention. Furthermore, of our 97 GWS loci, 35 (binomial P = 0.0019) were in high LD (r2 > 0.7) with one or more GWS SNPs in the National Human Genome Research Institute (NHGRI) GWAS catalogue (P < 5 × 10−8), even after removing anthropometric trait-associated SNPs. These SNPs were associated not only with cardiometabolic traits, but also with schizophrenia, smoking behaviour, irritable bowel syndrome, and Alzheimer’s disease (Supplementary Table 17a, b).

BMI tissues, biological pathways and gene sets

We anticipated the expanded sample size would not only identify additional BMI-associated variants, but also more clearly highlight the biology implicated by genetic studies of BMI. By applying multiple complementary methods, we identified biologically relevant tissues, pathways and gene sets, and highlighted potentially causal genes at associated loci. These approaches included systematic methods incorporating diverse data types, including the novel approach, Data-driven Expression Prioritized Integration for Complex Traits (DEPICT)21, and extensive manual review of the literature.

DEPICT used 37,427 human gene expression microarray samples to identify tissues and cell types in which genes near BMI-associated SNPs are highly expressed, and then tested for enrichment of specific tissues by comparing results with randomly selected loci matched for gene density. In total, 27 out of 31 significantly enriched tissues were in the central nervous system (CNS) (out of 209 tested; Fig. 2a and Supplementary Table 18). Current results are not sufficient to isolate specific brain regions important in regulating BMI. However, we observe enrichment not only in the hypothalamus and pituitary gland—key sites of central appetite regulation—but even more strongly in the hippocampus and limbic system, tissues that have a role in learning, cognition, emotion and memory.

Figure 2. Tissues and reconstituted gene sets significantly enriched for genes within BMI-associated loci.

Figure 2

a, DEPICT predicts genes within BMI-associated loci (P < 5 × 10−4) are enriched for expression in the brain and central nervous system. Tissues are sorted by physiological system and significantly enriched tissues are in black; the dotted line represents statistically significant enrichment. b, The gene sets most significantly enriched for BMI-associated loci by DEPICT (P < 10−6, FDR < 4 × 10−4). Nodes represent reconstituted gene sets and are colour-coded by P value. Edge thickness between nodes is proportional to degree of gene overlap as measured by the Jaccard index. Nodes with gene overlap greater than 25% were collapsed into a single ‘meta-node’ (blue border). c, The nodes contained within the most enriched meta-node, ‘clathrin-coated vesicle’, which shares genes with other gene sets relevant to glutamate signalling and synapse biology. d, The ‘generation of a signal involved in cell–cell signalling’ meta-node represents several overlapping gene sets relevant to obesity and energy metabolism (gene sets with P < 4 × 10−3, FDR < 0.05 shown). For the complete list of enriched gene sets refer to Supplementary Table 21a.

As a complementary approach, we examined overlap of associated variants at the 97 loci (r2 > 0.7 with the lead SNP) with five regulatory marks found in most of the 14 selected cell types from brain, blood, liver, pancreatic islet and adipose tissue from the ENCODE Consortium22 and Roadmap Epigenomics Project23 (Supplementary Table 19a–c). We found evidence of enrichment (P < 1.2 × 10−3) in 24 out of 41 data sets examined. The strongest enrichment was observed with promoter (histone 3 Lys 4 trimethylation (H3K4me3), histone 3 Lys 9 acetylation (H3K9ac)) and enhancer (H3K4me1, HeK27ac) marks detected in mid-frontal lobe, anterior caudate, astrocytes and substantia nigra, supporting neuronal tissues in BMI regulation.

To identify pathways or gene sets implicated by the BMI-associated loci, we first used Meta-Analysis Gene-set Enrichment of varia NT Associations (MAGENTA)24, which takes as input pre-annotated gene sets, and then tests for overrepresentation of gene set genes at BMI-associated loci. We found enrichment (false discovery rate (FDR) < 0.05) of seven gene sets, including neurotrophin signalling. Other highlighted gene sets related to general growth and patterning: basal cell carcinoma, acute myeloid leukaemia, and hedgehog signalling (Supplementary Table 20a, b).

Second, we used DEPICT, that uses predefined gene sets reconstituted using coexpression data, to perform gene set enrichment analysis. After merging highly correlated gene sets, nearly 500 gene sets were significantly enriched (FDR < 0.05) for genes in BMI-associated loci (Fig. 2b and Supplementary Table 21a, b). The most strongly enriched gene sets highlight potentially novel pathways in the CNS. These include gene sets related to synaptic function, long-term potentiation and neurotransmitter signalling (glutamate signalling in particular, but also noradrenaline, dopamine and serotonin release cycles, and GABA (γ-aminobutyric acid) receptor activity; Fig. 2c). Potentially relevant mouse behavioural phenotypes, such as physical activity and impaired coordination were also highly enriched (Fig. 2b and Supplementary Table 21a). Several gene sets previously linked to obesity, such as integration of energy metabolism, polyphagia, secretion and action of insulin and related hormones (for example, ‘regulation of insulin secretion by glucagon-like peptide 1′ and ‘glucagon signalling in metabolic regulation’), mTOR signalling (which affects cell growth in response to nutrient intake via insulin and growth factors25), and gene sets overlapping the neurotrophin signalling pathway identified by MAGENTA were also enriched, although not as significantly as other CNS processes (Fig. 2d). DEPICT also identified significant enrichment for additional cellular components and processes: calcium channels, MAP kinase activity, chromatin organization and modification, and ubiquitin ligases.

Third, we manually reviewed literature related to all 405 genes within 500 kb and r2 > 0.2 of the 97 index SNPs. We classified these genes into one or more biological categories, and observed 25 categories containing three or more genes (Supplementary Table 22). The largest category comprised genes involved in neuronal processes, including monogenic obesity genes involved in hypothalamic function and energy homeostasis, and genes involved in neuronal transmission and development. Other processes highlighted by the manual literature review included glucose and lipid homeostasis and limb development, which were less notable in the above methods, but may still be related to the underlying biology of BMI.

To identify specific genes that may account for BMI association, we considered each of the following to represent supportive evidence for a gene within a locus: (1) the gene nearest the index SNP26; (2) genes containing missense, nonsense or copy number variants, or a cis-expression quantitative trait locus (eQTL) in LD with the index SNP; (3) genes prioritized by integrative methods implemented in DEPICT; (4) genes prioritized by connections in published abstracts by GRAIL (Gene Relationships Across Implicated Loci)27; or (5) genes biologically related to obesity, related metabolic disease, or energy expenditure based on manual literature review (Tables 1 and 2, Extended Data Tables 24 and Supplementary Tables 2325). We first focused on the 64 genes in associated loci with more than one consistent line of supporting evidence. As expected, many of these genes overlap with CNS processes, including synaptic function, cell–cell adhesion, and glutamate signalling (ELAVL4, GRID1, CADM2, NRXN3, NEGR1 and SCG3), cause monogenic obesity syndromes (MC4R, BDNF, BBS4 and POMC), or function in extreme/early onset obesity in humans and mouse models (SH2B1 and NEGR1)6,28,29. Other genes with several lines of supporting evidence are related to insulin secretion and action, energy metabolism, lipid biology, and/or adipogenesis (TCF7L2, GIPR, IRS1, FOXO3, ASB4, RPTOR, NPC1, CREB1, FAM57B, APOBR and HSD17B12), encode RNA binding/processing proteins (PTBP2, ELAVL4, CELF1 and possibly RALYL), are in the MAP kinase signalling pathway (MAP2K5 and MAPK3), or regulate cell proliferation or cell survival (FAIM2, PARK2 and OLFM4). Although we cannot be certain that any individual gene is related to the association at a given locus, the strong enrichment of pathways among genes within associated loci argues for a causal role for these pathways, prioritizes specific genes for follow-up experiments, and provides the strongest genetic evidence so far for a role of particular biological and CNS processes in the regulation of human body mass.

Discussion

Our meta-analysis of nearly 340,000 individuals identified 97 GWS loci associated with BMI, 56 of which are novel. These loci account for 2.7% of the variation in BMI, and suggest that as much as 21% of BMI variation can be accounted for by common genetic variation. Our analyses provide robust evidence to implicate particular genes and pathways affecting BMI, including synaptic plasticity and glutamate receptor activity—pathways that respond to changes in feeding and fasting, are regulated by key obesity-related molecules such as BDNF and MC4R, and impinge on key hypothalamic circuits3032. These pathways also overlap with one of the several proposed mechanisms of action of topiramate, a component of one of two weight-loss drugs approved by the US Food and Drug Administration33,34. This observation suggests that the relevant site of action for this drug may be glutamate receptor activity, supporting the idea that these genes and pathways could reveal more targets for weight-loss therapies. BMI-associated loci also overlap with genes and pathways implicated in neurodevelopment (Supplementary Tables 21 and 22). Finally, consistent with previous work and findings from monogenic obesity syndromes, we confirm a role for the CNS—particularly genes expressed in the hypothalamus—in the regulation of body mass.

Examining the genes at BMI-associated loci in the context of gene expression, molecular pathways, eQTL results, mutational evidence and genomic location provides several complementary avenues through which to prioritize genes for relevance in BMI biology. Genes such as NPC1 and ELAVL4 are implicated by many lines of evidence (literature, mutational, eQTL and DEPICT) and become strong candidate genes in their respective locations. It is important to recognize that pathway methods and literature reviews are limited by current data sets and knowledge, and thus provide only a working model of obesity biology. For example, little is known about host genetic factors that regulate the microbiome. Variation in immune-related genes such as TLR4 could presumably exert an influence on obesity through the microbiome35. Together, our results underscore the heterogeneous aetiology of obesity and its links with several related metabolic diseases and processes.

BMI variants are generally associated with related cardiometabolic traits in accord with established epidemiological relationships. This could be due to shared genetic effects or to other causes of cross-phenotypic correlations. However, some BMI-associated variants have effects on related traits counter to epidemiological expectations. Once better understood, these mechanisms may not only help to explain why not all obese individuals develop related metabolic diseases, but also suggest possible mechanisms to prevent development of metabolic disease in those who are already obese.

Larger studies of common genetic variation, studies of rare variation (including those based on imputation, exome chips and sequencing), and improved computational tools will continue to identify genetic variants associated with BMI and help to further refine the biology of obesity. The 97 loci identified here represent an important step in understanding the physiological mechanisms leading to obesity. These findings strengthen the connection between obesity and other metabolic diseases, enhance our appreciation of the tissues, physiological processes, and molecular pathways that contribute to obesity, and will guide future research aimed at unravelling the complex biology of obesity.

METHODS

Study design

We conducted a two-stage meta-analysis to identify BMI-associated loci in European adults (Extended Data Fig. 1 and Extended Data Table 1). In stage 1 we performed meta-analysis of 80 GWAS (n = 234,069); and stage 2 incorporated data from 34 additional studies (n = 88,137) genotyped using Metabochip7 (Supplementary Tables 13). Secondary meta-analyses were also conducted for: (1) all ancestries, (2) European men, (3) European women, and (4) European population-based studies. The total number of subjects and SNPs included in each stage for all analyses is shown in Extended Data Table 1. No statistical methods were used to predetermine sample size.

Phenotype

BMI, measured or self-reported weight in kg per height in metres squared (Supplementary Tables 1 and 3) was adjusted for age, age squared, and any necessary study-specific covariates (for example, genotype-derived principal components) in a linear regression model. The resulting residuals were transformed to approximate normality using inverse normal scores. For studies with no known related individuals, residuals were calculated separately by sex and case/control status. For family-based studies, residuals were calculated with men and women together, adding sex as an additional covariate in the linear regression model. Relatedness was accounted for in a study-specific manner (Supplementary Table 2).

Sample quality control, imputation and association

Following study-specific quality control measures (Supplementary Table 2), all contributing GWAS common SNPs were imputed using the HapMap phase II CEU reference panel for European-descent studies37, and CEU+YRI+CHB+JPT HapMap release 22 for the African-American and Hispanic GWAS. Directly genotyped (GWAS and Metabochip) and imputed variants (GWAS only) were then tested for association with the inverse normally transformed BMI residuals using linear regression assuming an additive genetic model. Quality control following study level analyses was conducted following procedures outlined elsewhere38.

Meta-analysis

Fixed effects meta-analyses were conducted using the inverse variance-weighted method implemented in METAL39. Study-specific GWAS results as well as GWAS meta-analysis results were corrected for genomic control using all SNPs40. Study-specific Metabochip results as well as Metabochip meta-analysis results were genomic-control-corrected using 4,425 SNPs included on Metabochip for replication of associations with QT-interval, a phenotype not correlated with BMI, after pruning of SNPs within 500 kb of an anthropometry replication SNP. The final meta-analysis combined the genomic-control-corrected GWAS and Metabochip meta-analysis results.

Identification of novel loci

We used a distance criterion of ±500 kb surrounding each GWS peak (P < 5 × 10−8) to define independent loci and to place our results in the context of previous studies, including our previous GIANT meta-analyses. Of several locus models tested, this definition most closely reflected the loci defined by approximate conditional analysis using GCTA (Tables 1 and 2, respectively). Current index SNPs falling within 500 kb of a SNP previously associated with BMI, weight, extreme obesity or body fat percentage5,811 were considered previously identified.

Characterization of BMI-associated SNP effects

To investigate potential sources of heterogeneity between groups we compared the effect estimates of our 97 GWS SNPs for men versus women of European ancestry and Europeans versus non-Europeans. To address the effects of studies ascertained on a specific disease or phenotype on our results we also compare the effect estimates of European ancestry studies of population-based studies with the following European-descent subsets of studies: (1) non-population-based studies (that is, those ascertained on a specific disease or phenotype); (2) type 2 diabetes cases; (3) type 2 diabetes controls; (4) combined type 2 diabetes cases and controls; (5) CAD cases; (6) CAD controls; and (7) combined CAD cases and controls (Supplementary Tables 10 and 11). We also tested for heterogeneity of effect estimates between our European sex-combined meta-analysis and results from recent GWAS meta-analyses for BMI in individuals of African or east Asian ancestry10,41 (Supplementary Table 9). Heterogeneity was assessed as described previously42. A Bonferroni-corrected P < 5 × 10−4 (corrected for 97 tests) was used to assess significance. For heterogeneity tests assessing effects of ascertainment, we also used a 5% FDR threshold to assess significance of heterogeneity statistics (Supplementary Table 11).

Fine-mapping

We compared the meta-analysis results and credible sets of SNPs likely to contain the causal variant, based on the method described previously14, across the European-only, non-European, and all ancestries sex-combined meta-analyses. For each index SNP falling within a Metabochip fine-mapping region (27 for BMI), all SNPs available within 500 kb on either side of the index SNP were selected. Effect size estimates and standard errors for each SNP were converted to approximate Bayes’ factors according to the method described previously15. All approximate Bayes’ factors were then summed across the 1-megabase (Mb) region and the proportion of the posterior odds of being the causal variant was calculated for each variant (approximate Bayes’ factor for SNPi/sum of approximate Bayes’ factors for the region). The set of SNPs that accounts for 99% of posterior odds of association in the region denotes the set most likely to contain the causal variant for that association region (Supplementary Table 12).

Cumulative effects, risk prediction and variance explained

We assessed the cumulative effects of the 97 GWS loci on mean BMI and on their ability to predict obesity (BMI ≥ 30 kg m−2) using the c statistic from logistic regression models in the Health and Retirement Study17, a longitudinal study of 26,000 European Americans 50 years or older. The variance explained (VarExp) by each SNP was calculated using the effect allele frequency (f) and beta (β) from the meta-analyses using the formula VarExp = β2(1 − f)2f.

For polygene analyses, the approximate conditional analysis from GCTA19,20, was used to select SNPs using a range of P value thresholds (that is, 5 × 10−8, 5 × 10−7, …, 5 × 10−3) based on summary data from the European sex-combined meta-analysis excluding TwinGene and QIMR studies. We performed a within-family prediction analysis using full-sib pairs selected from independent families (1,622 pairs from the QIMR cohort and 2,758 pairs from the TwinGene cohort) and then SNPs at each threshold were used to calculate the percentage of phenotypic variance explained and predict risk (Extended Data Figs 2 and 3). We then confirmed the results from population-based prediction and estimation analyses in an independent sample of unrelated individuals from the TwinGene (n = 5,668) and QIMR (n = 3,953) studies (Extended Data Fig. 3 and Fig. 1c). The SNP-derived predictor was calculated using the profile scoring approach implemented in PLINK and estimation analyses were performed using the all-SNP estimation approach implemented in GCTA.

Enrichment analysis of Metabochip SNPs selected for replication

The 5,055 SNPs that were included for BMI replication on Metabochip included 1,909 independent SNPs (r2 < 0.1 and > 500 kb apart), of which 1,458 displayed directionally consistent effect estimates with those reported previously5. To estimate the number of Metabochip SNPs truly associated with BMI, we counted the number of SNPs with directional consistency (DC) between ref. 5 and a meta-analysis of non-overlapping samples for these 1,909 SNPs. We then calculated DC in the presence of a mixture of associated and non-associated SNPs assuming P(DC ∣ associated) = 1 and P(DC ∣ not associated) = 0.5. In this formulation, DC = R/2 + S, meaning that S = 2DC – T, in which T equals the total number of SNPs, R equals the number of SNPs not associated with BMI, and S equals the number of SNPs associated with BMI. With DC = 1,458 and T = 1,909, we estimate S to be 2DC – T = 2 × 1,458 – 1,909 = 1,007.

Joint and conditional multiple SNP association analysis

To identify additional signals in regions of association, we used GCTA19, an approach that uses meta-analysis summary statistics and an LD matrix derived from a reference sample, to perform approximate joint and conditional SNP association analysis. We used 6,654 unrelated individuals of European ancestry from the ARIC cohort as the reference sample to approximate conditional P values.

Manual gene annotation and biological description

All genes within 500 kb of an index SNP were annotated for molecular function, cellular function, and for evidence of association with BMI-related traits in human or animal model experiments (Supplementary Table 22). We used several avenues for annotation, including Spotter (http://csg.sph.umich.edu/boehnke/spotter/), SNIPPER (http://csg.sph.umich.edu/boehnke/snipper/), PubMed (http://www.ncbi.nlm.nih.gov/pubmed/), OMIM (http:/www.omim.org) and UNIPROT (http://www.uniprot.org/). When no genes mapped to this interval the nearest gene on each side of the index SNP was annotated. In examining possible functions of genes in the region, we excluded any references to GWAS or other genetic association studies. We analysed 405 genes in the 97 GWS loci and manually curated them into 25 biological categories containing more than three genes.

Functional variants

All variants within 500 kb (HapMap release 22/1000 Genomes CEU) and in LD (r2 > 0.7) with an index SNP were annotated for functional effects based on RefSeq transcripts using Annovar43 (http://www.openbioinformatics.org/annovar/). PhastCon, Grantham, GERP, and PolyPhen44 predictions were accessed via the Exome Variant Server45 (http://evs.gs.washington.edu/EVS), and from SIFT46 (http://sift.jcvi.org/) (Extended Data Table 4).

Copy number variations correlated with BMI index SNPs

To study common copy number variations, we used a list of copy number variations well-tagged by SNPs in high LD (r2 > 0.8) with deletions in European populations from phase 1 release of the 1000 Genomes Project47 (Supplementary Table 25).

eQTLs

We examined the cis associations between the 97 GWS SNPs and expression of nearby genes in whole blood, lymphocytes, skin, liver, omental fat, subcutaneous fat and brain tissue4855 (Supplementary Table 23). Conditional analyses were performed by including both the BMI-associated SNP and the most significant cis-associated SNP for the given transcript. Conditional analyses were conducted for all data sets, except the brain tissue data set due to limited power. To minimize the potential for false-positives, only cis associations below a study-specific FDR of 5% (or 1% for some data sets), in LD with the peak SNP (r2 > 0.7) for the transcript, and with conditional P > 0.05 for the peak SNP, are reported (Extended Data Table 2).

MAGENTA

We used the MAGENTA method to test predefined gene sets for enrichment at BMI-associated loci24. We used the GWAS + Metabochip data as input and applied default settings.

GRAIL

We used GRAIL27 to identify genes near BMI-associated loci having similarities in the published scientific text using PubMed abstracts as of December 2006. The BMI loci were queried against HapMap release 22 for the European panel, and we controlled for gene size.

DEPICT

We used DEPICT to identify the most likely causal gene at a given associated locus, reconstituted gene sets enriched for BMI associations, and tissues and cell types in which genes from associated loci are highly expressed21. To accomplish this, the method relies on publicly available gene sets (including molecular pathways) and uses gene expression data from 77,840 gene expression arrays75 to predict which other genes are likely to be part of these gene sets, thus combining known annotations with predicted annotations. For details and negative control analyses please see Supplementary Methods.

We first clumped the European-only GWAS-based meta-analysis summary statistics using 500 kb flanking regions, LD r2 > 0.1 and excluded SNPs with P ≥ 5 × 10−4; which resulted in a list of 590 independent SNPs. HapMap phase II CEU genotype data37 was used to compute LD and genomic coordinates were defined by genome build GRCh38. Because the GWAS meta-analysis was based on both GWAS and Metabochip studies, there were discrepancies in the index SNPs that are referenced in Table 1 of the paper and the ones used in DEPICT, which was run on the GWAS data only. Therefore we forced in GWS index SNPs from the GWAS plus Metabochip GWA meta-analysis into the DEPICT GWAS-only based analysis. This enabled a more straightforward comparison of genes in DEPICT loci and genes in GWS loci highlighted by manual lookups, and did not lead to any significant bias towards SNPs on Metabochip (data not shown). We forced in 62 of the GWS loci in Table 1, so all of the 97 SNPs were among the 590 SNPs. The 590 SNPs were further merged into 511 non-overlapping regions (FDR < 0.05) used in DEPICT analysis. For additional information on the analysis please refer to Supplementary Methods.

Cross-trait analyses

To explore the relationship between BMI and an array of cardiometabolic traits and diseases, association results for the 97 BMI index SNPs were requested from 13 GWAS meta-analysis consortia: DIAGRAM (type 2 diabetes)56, CARDIoGRAM-C4D (CAD)57, ICBP (systolic and diastolic blood pressure (SBP, DBP))58, GIANT (waist-to-hip ratio, hip circumference, and waist circumference, each unadjusted and adjusted for BMI)13,59, GLGC (HDL, low density lipoprotein cholesterol, triglycerides, and total cholesterol)60, MAGIC (fasting glucose, fasting insulin, fasting insulin adjusted for BMI, and two-hour glucose)6163, ADIPOGen (BMI-adjusted adiponectin)64, CKDgen (urine albumin-to-creatinine ratio (UACR), estimated glomerular filtration rate, and overall CKD)65,66, ReproGen (age at menarche, age at menopause)67,68, GENIE (diabetic nephropathy)69,70. Proxies (r2 > 0.8 in CEU) were used when an index SNP was unavailable.

Enrichment of concordant effects

We compared the effects for the 97 BMI index SNP across these related traits using a one-sided binomial test of the number of concordant effects versus a null expectation of P = 0.5. Concordant and nominally significant (P < 0.05) SNP effects were similarly tested using a one-sided binomial test with a null expectation of P = 0.05. We evaluated significance in either test with a Bonferroni-corrected threshold of P = 0.002 (0.05/23 traits tested).

Joint effects of cross-trait associations

To determine the joint effect of all 97 BMI loci on other cardiometabolic phenotypes, we used the meta-regression technique from ref. 64 to correlate the effect estimates of the BMI-increasing alleles with effect estimates from meta-analyses for each of the metabolic traits from other consortia (DIAGRAM, MAGIC, ICBP, GLGC, ADIPOGen, ReproGen and CARDIoGRAM).

Cross-traits heatmap

To explore observed concordance in effects of BMI loci on other cardiometabolic and anthropometric traits, we converted the effect estimates and standard errors (or P values) from meta-analysis to Z-scores oriented with respect to the BMI-increasing allele, for each of the 97 BMI index SNPs in the twenty-three traits. We then classified each Z-score as follows to generate a vector of the Z-score of each trait at each locus: 0 (not significant) if −2 ≤ Z ≤ 2; 1 (significant positive) if Z > 2; −1 (significant negative) if Z < −2.

Extended Data Fig. 5 displays these locus-trait relationships in a heatmap using Euclidean distance and complete linkage clustering to order both loci and traits.

Cross-traits bubble plot

We also represent the genetic overlap between other cardiometabolic traits and BMI susceptibility loci with a bubble plot in which the size of each bubble is proportional to the fraction of BMI-associated loci for which there was a significant association (P < 5 × 10−4). Each pair of bubbles is connected by a line proportional to the number of significant BMI-increasing loci overlapping between the traits.

NHGRI GWAS catalogue lookups

We extracted previously reported GWAS association within 500 kb of and r2 > 0.7 with any BMI-index SNP from the NHGRI GWAS catalogue71 (http://www.genome.gov/gwastudies; Supplementary Table 17a, b). For studies reporting greater than 30 significant hits, additional SNP-trait associations were pulled from the literature and compared to BMI index SNPs the same as with other GWAS catalogue studies.

ENCODE/Roadmap

To identify global enrichment of data sets at the BMI-associated loci we performed permutation-based tests in a subset of 41 open chromatin (DNase-seq), histone modification (H3K27ac, H3K4me1, H3K4me3 and H3K9ac), and transcription factor binding data sets from the ENCODE Consortium22, Roadmap Epigenomics Project23 and when available the ENCODE Integrative Analysis60,72 (Supplementary Table 19). We processed Roadmap Epigenomics sequencing data with multiple biological replicates using MACS2 (ref. 73) and then applied same Irreproducible Discovery Rate pipeline used in the ENCODE Integrative Analysis60,72. Roadmap Epigenomics data with only a single replicate were analysed using MACS2 alone. We examined variants in LD with 97 BMI index SNPs based on r2 > 0.7 from the 1000 Genomes phase 1 version 2 EUR samples74. We matched the index SNP at each locus with 500 variants having no evidence of association (P > 0.5, ~1.2 million total variants) with a similar distance to the nearest gene (± 11,655 bp), number of variants in LD (±8 variants), and minor allele frequency. Using these pools, we created 10,000 sets of control variants for each of the 97 loci and identified variants in LD (r2 > 0.7) and within 1 Mb. For each SNP set, we calculated the number of loci with at least one variant located in a regulatory region under the assumption that one regulatory variant is responsible for each association signal. We estimated the P value assuming a sum of binomial distributions to represent the number of index SNPs (or their LD proxies; r2 > 0.7) that overlap a regulatory data set compared to the expectation observed in the 500 matched control sets. Data sets were considered significantly enriched if the P value was below a Bonferroni-corrected threshold of 1.2 × 10−3, adjusting for 41 tests.

Extended Data

Extended Data Figure 1. Study design.

Extended Data Figure 1

*The SNP counts reflect sample size filter of n ≥ 50,000. §Counts represent the primary European sex-combined analysis. Please see Extended Data Table 1 for counts for secondary analyses.

Extended Data Figure 2. Genetic characterization of BMI-associated variants.

Extended Data Figure 2

a, Plot of the cumulative phenotypic variance explained by each locus ordered by decreasing effect size. b, The relationship between effect size and allele frequency. Previously identified loci are blue circles and novel loci are red triangles. c, Quantile–quantile (Q–Q) plot of meta-analysis P values for all 1,909 BMI-replication SNPs (blue) and after removing SNPs near the 97 associated loci (green). d, Histogram of cumulative effect of BMI risk alleles. Mean BMI for each bin is shown by the black dots (with standard deviation) and corresponds to the right-hand y axis.

Extended Data Figure 3. Partitioning the variance in and risk prediction from SNP-derived predictor.

Extended Data Figure 3

a, b, The analyses were performed using 2,758 full sibling pairs from the TwinGene cohort (a) and 1,622 pairs from the QIMR cohort (b). The SNP-based predictor was adjusted for the first 20 principal components. The variance of the SNP-based predictor can be partitioned into four components (Vg, Ve, Cg and Ce) using the within-family prediction analysis, in which Vg is the variance explained by real SNP effects, Cg is the covariance between predictors attributed to the real effects of SNPs that are not in LD but correlated due to population stratification, Ve is the accumulated variance due to the errors in estimating SNP effects, and Ce is the covariance between predictors attributed to errors in estimating the effects of SNPs that are correlated due to population stratification. Error bars reflect s.e.m. of estimates. c, The prediction R2 shown on the y axis is the squared correlation between phenotype and SNP-based genetic predictor in unrelated individuals from the TwinGene (n = 5,668) and QIMR (n = 3,953) studies. The number shown in each column is the number of SNPs selected from the GCTA joint and conditional analysis at a range of P-value thresholds. In each case, the predictor was adjusted by the first 20 principal components. The column in orange is the average prediction R2 weighted by sample size over the two cohorts. The dashed grey line is the value inferred from the within-family prediction analyses using this equation R2 = (Vg + Cg)2/(Vg + Ve + Cg + Ce).

Extended Data Figure 4. Comparison of BMI-associated index SNPs across ethnicities.

Extended Data Figure 4

a, b, BMI effects observed in European ancestry individuals (x axes) compared to African ancestry (a) or Asian ancestry (b) individuals (y axes). c, d, Allele frequencies between ancestry groups, as in a and b. e, f, Comparison of the estimates of explained variance. In all plots, novel loci are in red and previously identified loci are in blue.

Extended Data Figure 5. Effects of BMI-associated loci on related metabolic traits.

Extended Data Figure 5

Unsupervised hierarchical clustering of the 97 BMI-associated loci (y axis) on 23 related metabolic traits (x axis). The top row shows the a priori expected relationship with BMI (green is concordant effect direction, purple is opposite). Loci with statistically significant concordant direction of effect are highlighted in green, and significant but opposing effects are in purple. Grey indicates a non-significant relationship and those with no information are in white. The key in the top left corner also shows the count of gene–phenotype pairs in each category (cyan bars).

Extended Data Figure 6. Bubble chart representing the genetic overlap across traits at BMI susceptibility loci.

Extended Data Figure 6

Each bubble represents a trait for which association results were requested for the 97 GWS BMI loci. The size of the bubble is proportional to the number of BMI-increasing loci with a significant association. A line connects each pair of bubbles with thickness proportional to the number of significant loci shared between the traits. Traits tested include the current study BMI SNPs, African-American BMI (AA BMI), hip circumference (HIP), HIP adjusted for BMI (HIPadjBMI), waist circumference (WC), waist circumference adjusted for BMI (WCadjBMI), waist-to-hip ratio (WHR), waist-to-hip ratio adjusted for BMI (WHRadjBMI), height, adiponectin, coronary artery disease (CAD), diastolic blood pressure (DBP), systolic blood pressure (SBP), high-density lipoprotein (HDL), low-density lipoprotein (LDL), total cholesterol (TC), triglycerides (TG), type 2 diabetes (T2D), fasting glucose (FG), fasting insulin (FI), fasting insulin adjusted for BMI (FIadjBMI), two-hour glucose (Glu2hr), diabetic nephropathy (Diab_Neph), age at menopause (AgeMenopause), and age at menarche (AgeMenarche).

Extended Data Table 1.

Descriptive characteristics of meta-analyses

Meta-analysis Total number
of studies
Maximum number of
subjects
Number of
SNPs*
λ GC
European sex-combined
 GWAS 80 234,069 2,550,021 1.526
 Metabochip 34 88,137 156,997 1.25
 Joint GWAS+Metabochip 114 322,154 2,554,623 1.084
European men
 GWAS 72 104,666 2,473,152 1.279
 Metabochip 34 48,274 152,326 1.121
 Joint GWAS+Metabochip 106 152,893 2,477,617 1.006
European women
 GWAS 74 132,115 2,491,697 1.336
 Metabochip 33 39,864 153,086 1.029
 Joint GWAS+Metabochip 107 171,977 2,494,571 1.002
European population-based
 GWAS 49 162,262 2,502,573 1.385
 Metabochip 20 46,263 155,617 1.034
 Joint GWAS+Metabochip 69 209,521 2,506,448 1.003
All ancestries
 GWAS 82 236,231 2,550,614 1.451
 Metabochip 43 103,047 181,718 1.25
 Joint GWAS+Metabochip 125 339,224 2,555,496 1.004
*

For the GWAS and joint GWAS+Metabochip analyses, SNP count reflects n ≥ 50,000.

Extended Data Table 2.

Previously known GWS BMI loci in European meta-analysis

SNP Chr:Position *Notable gene(s) Alleles EAF β SE P value
rs1558902 16:52,361,075 FTO(B,N) A/T 0.415 0.082 0.003 7.51E-153
rs6567160 18:55,980,115 MC4R(B,N) C/T 0.236 0.056 0.004 3.93E-53
rs13021737 2:622,348 TMEM18(N) G/A 0.828 0.06 0.004 1.11E-50
rs10938397 4:44,877,284 GNPDA2(N); GABRG1(B) G/A 0.434 0.04 0.003 3.21E-38
rs543874 1:176,156,103 SEC16B(N) G/A 0.193 0.048 0.004 2.62E-35
rs2207139 6:50,953,449 TFAP2B(B,N) G/A 0.177 0.045 0.004 4.13E-29
rs11030104 11:27,641,093 BDAF(B,M,N) A/G 0.792 0.041 0.004 5.56E-28
rs3101336 1:72,523,773 NEGR1(B,C,D,N) C/T 0.613 0.033 0.003 2.66E-26
rs7138803 12:48,533,735 BCDIN3D(N); FAIM2(D) A/G 0.384 0.032 0.003 8.15E-24
rs10182181 2:25,003,800 ADCY3(B,M,N,Q); POMC(B,G);
NCOA1(B)
SH2B1(B,M,Q); APOBR(M,Q);
G/A 0.462 0.031 0.003 8.78E-24
rs3888190 16:28,796,987 ATXN2L(Q); SBK1(Q,D); SULT1A2(Q);
TUFM(Q)
A/C 0.403 0.031 0.003 3.14E-23
rs1516725 3:187,306,698 E7V5(N) C/T 0.872 0.045 0.005 1.89E-22
rs12446632 16:19,842,890 GPRC5B(C,N); IQCK(Q) G/A 0.865 0.04 0.005 1.48E-18
rs2287019 19:50,894,012 QPCTL(N); GIPR(B,M) C/T 0.804 0.036 0.004 4.59E-18
rs16951275 15:65,864,222 M4P2K5(B,D,N); LBXCOR1(M) T/C 0.784 0.031 0.004 1.91E-17
rs3817334 11:47,607,569 MTCH2(M,Q); C1QTNF4(Q,I); SPI1(Q);
CELF1(D)
T/C 0.407 0.026 0.003 5.15E-17
rs2112347 5:75,050,998 POC5(M); HMGCR(B); COL4A3BP(B) T/G 0.629 0.026 0.003 6.19E-17
rs12566985 1:74,774,781 FPGT-TNNI3K(N) G/A 0.446 0.024 0.003 3.28E-15
rs3810291 19:52,260,843 ZC3H4(D,N,Q) A/G 0.666 0.028 0.004 4.81E-15
rs7141420 14:78,969,207 NRXN3(D,N) T/C 0.527 0.024 0.003 1.23E-14
rs13078960 3:85,890,280 CADM2(D,N) G/T 0.196 0.03 0.004 1.74E-14
rs10968576 9:28,404,339 LINGO2(D,N) G/A 0.32 0.025 0.003 6.61E-14
rs17024393 1:109,956,211 GNAT2(N); AMPD2(D) C/T 0.04 0.066 0.009 7.03E-14
rs12429545 13:53,000,207 OLFM4(B,N) A/G 0.133 0.033 0.005 1.09E-12
rs13107325 4:103,407,732 SLC39A8(M,N,Q) T/C 0.072 0.048 0.007 1.83E-12
rs11165643 1:96,696,685 PTBP2(D,N) T/C 0.583 0.022 0.003 2.07E-12
rs17405819 8:76,969,139 HNF4G(B,N) T/C 0.7 0.022 0.003 2.07E-11
rs1016287 2:59,159,129 LINC01122(N) T/C 0.287 0.023 0.003 2.25E-11
rs4256980 11:8,630,515 TRIM66(D,M,N); TUB(B) G/C 0.646 0.021 0.003 2.90E-11
rs12401738 1:78,219,349 FUBP1(N); USP33(D) A/G 0.352 0.021 0.003 1.15E-10
rs205262 6:34,671,142 C6orf106(N); SNRPC(Q) G/A 0.273 0.022 0.004 1.75E-10
rs12016871 13:26,915,782 MTIF3(N); GTF3A(Q) T/C 0.203 0.03 0.005 2.29E-10
rs12940622 17:76,230,166 RPTOR(B,N) G/A 0.575 0.018 0.003 2.49E-09
rs11847697 14:29,584,863 PRKD1(N) T/C 0.042 0.049 0.008 3.99E-09
rs2075650 19:50,087,459 TOMM40(B,N); APOE(B); APOC1(B) A/G 0.848 0.026 0.005 1.25E-08
rs2121279 2:142,759,755 LRP1B(N) T/C 0.152 0.025 0.004 2.31E-08
rs29941 19:39,001,372 KCTD15(N) G/A 0.669 0.018 0.003 2.41E-08
rs1808579 18:19,358,886 NPC1(B,G,M,Q); C18orf8(N,Q) C/T 0.534 0.017 0.003 4.17E-08

SNP positions are reported according to Build 36 and their alleles are coded based on the positive strand. Effect alleles, allele frequencies, betas (β), s.e.m., sample sizes (n), and P values are based on the meta-analysis of GWAS I + II + Metabochip association data from the European sex-combined data set.

*

Notable genes from biological relevance to obesity (B); GRAIL results (G); BMI-associated variant is in strong LD (r2 ≥ 0.7) with a missense variant in the indicated gene (M); gene nearest to Index SNP (N); association and eQTL data converge to affect gene expression (Q); DEPICT analyses (D); copy number variation (C).

Extended Data Table 3.

Association of the GWS SNPs for BMI with cis-gene expression (cis-eQTLs)

SNP Chr. BMI
increasing
allele
Tissue Gene β for
Giant
SNP
P for
GIANT SNP
Padj for
GIANT
SNP
PeakSNP r 2 P for peak
SNP
Padj for
peak SNP
Reference
Novel loci
rs11583200 1 c Subcutaneous ELAVL4 −0.066 1.90E-12 0.44 rs6588374 0.78 1.07E-12 0.36 Zhong et al.
rs492400 2 c Liver PLCD4 −0.054 4.64E-40 0.98 rs10187066 1.00 4.49E-40 0.98 Zhong et al.
rs492400 2 c Lymphocyte RQCD1 0.392 7.11E-22 0.94 rs526134 1.00 4.06E-22 0.21 Dixon et al.
rs492400 2 c PBMC RQCD1 −0.102 2.43E-06 0.98 rs526134 0.95 2.21E-06 0.96 PBMC meta-analysis
rs492400 2 c Omental TTLL4 0.018 1.33E-10 0.82 rs12987009 0.73 2.82E-13 0.07 Zhong et al.
rs492400 2 c Lymphocyte TTLL4 0.158 9.02E-06 1 rs492400 1.00 9.02E-06 1 Dixon et al.
rs17001654 4 G Lymphocyte SCARB2 0.248 5.57E-09 0.59 rs6835324 0.94 3.42E-09 0.25 Dixon et al.
rs9400239 6 C Subcutaneous HSS00296402 0.034 9.51E-22 0.97 rs2153960 0.94 1.93E-23 0.48 Zhong et al.
rs9400239 6 C Omental HSS00296402 0.015 1.34E-13 0.50 rs2153960 0.93 4.64E-17 0.22 Zhong et al.
rs1167827 7 G Blood PMS2P3 −0.595 4.20E-32 0.66 rs6963105 0.93 3.00E-32 0.39 Emilsson et al.
rs1167827 7 G Omental PMS2P3 −0.027 1.57E-11 0.95 rs6963105 0.98 6.94E-12 0.86 Zhong et al.
rs1167827 7 G Subcutaneous PMS2P3 −0.030 1.30E-10 0.71 rs1167796 0.73 1.04E-12 0.10 Zhong et al.
rs1167827 7 G Adipose PMS2P3 −0.346 3.40E-09 1 rs1167827 1.00 3.40E-09 1 Emilsson et al.
rs1167827 7 G Blood PMS2P5 −0.367 1.20E-11 0.47 rs6963105 0.93 5.00E-12 0.14 Emilsson et al.
rs1167827 7 G Subcutaneous WBSCR16 0.025 1.44E-10 1 rs1167827 1.00 1.44E-10 1 Zhong et al.
rs1167827 7 G Omental WBSCR16 0.017 1.75E-06 1 rs1167827 1.00 1.75E-06 1 Zhong et al.
rs9641123 7 C Abdominal SAT hsa-miR-653 −0.344 1.54E-04 0.23 rs16868443 0.71 1.38E-04 0.20 Parts et al.
rs11191560 10 C Gluteal SAT SFXN2 0.153 1.72E-05 0.20 rs71496550 NA 4.42E-06 0.41 Min et al.
rs11191560 10 C Abdominal SAT SFXN2 0.628 1.44E-04 0.02 rs71496550 NA 9.13E-05 0.94 Min et al.
rs7164727 15 T Lymphocyte BBS4 −0.163 3.14E-05 1 rs7164727 1.00 3.14E-05 1 Dixon et al.
rs9925964 16 A Liver VKORC1 0.122 4.41E-37 0.84 rs2303223 0.88 3.62E-44 0.05 Zhong et al.
rs9925964 16 A Subcutaneous ZNF646 0.017 2.55E-06 1 rs9925964 1.00 2.55E-06 1 Zhong et al.
rs9925964 16 A Blood ZNF668 −0.382 1.70E-12 0.48 rs10871454 0.93 1.10E-12 0.26 Emilsson et al.
rs9914578 17 G Subcutaneous C17orf13 −0.010 3.01E-06 0.99 rs7225843 0.99 2.86E-06 0.97 Zhong et al.
rs1808579 18 C SKIN C18orf8 −0.073 5.74E-10 0.86 rs1788781 0.90 1.67E-10 0.13 Grundberg et al.
rs1808579 18 C Subcutaneous C18orf8 −0.014 8.41E-08 1 rs1808579 1.00 8.41E-08 1 Zhong et al.
rs17724992 19 A Blood PGPEP1 −0.825 1.60E-40 1 rs17724992 1.00 1.60E-40 1 Emilsson et al.

Previously reported loci

rs10182181 2 G Subcutaneous ADCY3 0.022 7.57E-06 0.69 rs11684619 0.72 8.70E-09 0.05 Zhong et al.
rs2176040 2 A Omental IRS1 −0.036 3.74E-09 0.97 rs908252 0.87 3.98E-10 0.47 Zhong et al.
rs13107325 4 T Liver SLC39A8 −0.101 1.29E-17 1 rs13107325 1.00 1.29E-17 1 Zhong et al.
rs205262 6 G Blood SNRPC −0.462 9.60E-15 0.58 rs6457792 0.96 9.40E-15 0.55 Emilsson et al.
rs205262 6 G PBMC SNRPC −0.127 3.40E-09 0.03 rs2744943 0.73 3.15E-11 0.12 PBMC meta-analysis
rs205262 6 G Omental SNRPC −0.012 6.64E-06 0.81 rs2814984 0.75 8.03E-07 0.30 Zhong et al.
rs3817334 11 T SKIN C1QTNF4 −0.051 1.34E-09 0.82 rs7124681 1.00 9.42E-10 0.34 Grundberg et al.
rs3817334 11 T Subcutaneous MTCH2 0.044 7.64E-13 0.76 rs12794570 0.76 2.54E-15 0.10 Zhong et al.
rs3817334 11 T Brain MTCH2 28.255 7.51E-08 NA NA NA NA NA Myers et al.
rs3817334 11 T FAT SPI1 −0.090 9.90E-07 0.90 rs10769262 0.70 1.15E-08 1 Grundberg et al.
rs12016871 13 T PBMC GTF3A −0.258 6.68E-34 0.90 rs7988412 0.81 1.81E-36 0.29 PBMC meta-analysis
rs12016871 13 T Lymphocyte GTF3A −0.375 3.89E-15 0.32 rs7988412 0.86 1.32E-15 0.06 Dixon et al.
rs12446632 16 G Omental IQCK 0.028 2.27E-10 0.83 rs11865578 0.83 4.14E-13 0.14 Zhong et al.
rs12446632 16 G Liver IQCK 0.031 5.39E-06 0.74 rs9921401 0.70 3.82E-07 0.20 Zhong et al.
rs3888190 16 A Blood APOBR 0.303 2.10E-08 0.68 rs2411453 0.83 1.10E-08 0.25 Emilsson et al.
rs3888190 16 A PBMC ATXN2L 0.084 1.04E-04 0.99 rs8049439 0.99 8.59E-05 0.88 PBMC meta-analysis
rs3888190 16 A SKIN SBK1 −0.063 1.63E-06 0.41 rs4788084 0.82 2.87E-07 0.10 Grundberg et al.
rs3888190 16 A Adipose SH2B1 −0.407 4.10E-13 0.67 rs12928404 0.92 2.40E-13 0.30 Emilsson et al.
rs3888190 16 A Omental SH2B1 −0.014 5.29E-07 0.87 rs12928404 0.93 4.65 E-07 0.83 Zhong et al.
rs3888190 16 A Subcutaneous SULT1A2 0.067 3.36E-21 0.52 rs1074631 0.80 3.93E-23 0.14 Zhong et al.
rs3888190 16 A PBMC TUFM 0.694 9.81E-198 0.94 rs8049439 0.99 9.81E-198 0.12 PBMC meta-analysis
rs1808579 18 C Subcutaneous NPC1 −0.027 2.52E-10 0.83 rs1805081 0.78 7.86E-14 0.06 Zhong et al.
rs3888190 16 A SKIN TUFM 0.074 7.90E-10 0.46 rs2411453 0.76 1.91E-10 0.09 Grundberg et al.
rs3810291 19 A Adipose ZC3H4 −0.386 3.70E-09 1 rs3810291 1.00 3.70E-09 1 Emilsson et al.

Extended Data Table 4.

Putative coding variants in LD (r2 ≥ 0.7) with GWS BMI loci

BMI SNP Chr. Source Putative Coding
Variant
r 2 Gene Protein
Alteration
PhastCon
Score
GERP
Score
Grantham
Score
PolyPhen SIFT
Prediction
SIFT Score
Novel genome-wide significant loci
rs492400 2 1000G rs3770213 0.89 ZNF142 L956H 0 −1.6 99 possibly damaging Damaging 0
rs492400 2 1000G rs3770214 0.89 ZNF142 S751G 0.2 1.4 56 benign Tolerated 0.08
rs492400 2 1000G rs2230115 0.963 ZNF142 A541S 0.5 5.1 99 benign Tolerated 0.044
rs492400 2 1000G rs1344642 0.963 STK36 R583Q 0 2.4 43 possibly damaging Damaging 0
rs492400 2 1000G rs1863704 0.89 STK36 G1003D 0 2 94 possibly damaging Tolerated 0.41
rs492400 2 1000G rs1863704 0.89 STK36 G982D 0 2 94 possibly damaging - -
rs492400 2 1000G rs3731877 0.792 TLL4 E34Q 1 5.5 29 probably damaging Unknown Not scored
rs17001654 4 1000G rs61750814 1 NUP54 N250S 1 5.5 46 benign Damaging 0.05
rs4740619 9 1000G rs4741510 0.901 CCDC171 S121T 1 2 58 benign Damaging 0.05
rs4740619 9 1000G rs1539172 0.74 CCDC171 K1069R 1 4.1 26 benign Tolerated 1
rs2176598 11 1000G rs11555762 0.774 HSD17B12 S280L 0 0.4 145 benign Tolerated 0.74
rs3849570 3 1000G rs2229519 0.771 GBE1 R190G 1 4.8 125 benign Damaging 0.04
rs3736485 15 1000G rs12102203 0.966 DMXL2 S1288P 0.7 1.7 74 benign Tolerated 0.32
rs7164727 15 1000G rs2277598 0.839 BBS4 I182T 0 −4.4 89 benign Tolerated 0.47
rs9925964 16 1000G rs749670 0.869 ZNF646 E327G 1 4.2 98 benign Tolerated 0.44

Previously identified genome-wide significant loci

rs10182181 2 HapMap rs11676272 0.967 ADCY3 S107P 0 2.9 74 benign Tolerated 0.28
rs13107325 4 1000G rs13107325 1 SLC39A8 A324T 1 4.4 5.8 benign Tolerated 0.09
rs13107325 4 1000G rs13107325 1 SLC39A8 A391T 1 4.4 5.8 benign Tolerated 0.09
rs2112347 5 1000G rs2307111 0.862 POC5 H11R 0.9 5.8 29 benign Unknown Not scored
rs2112347 5 1000G rs2307111 0.862 POC5 H36R 0.9 5.8 29 benign Unknown Not scored
rs4256980 11 HapMap rs7935453 0.729 TRIM66 L630V - - - - Tolerated 1
rs4256980 11 1000G rs11042022 0.876 TRIM66 H466R - - - - Tolerated 0.38
rs4256980 11 1000G rs11042023 0.959 TRIM66 H324R 1 5.1 29 probably damaging Damaging 0.03
rs11030104 11 1000G rs6265 0.817 BDNF V148M 1 5.2 21 probably damaging Damaging 0
rs11030104 11 1000G rs6265 0.817 BDNF V66M 1 5.2 21 probably damaging Damaging 0
rs11030104 11 1000G rs6265 0.817 BDNF V74M 1 5.2 21 probably damaging Damaging 0
rs11030104 11 1000G rs6265 0.817 BDNF V81M 1 5.2 21 probably damaging Damaging 0
rs11030104 11 1000G rs6265 0.817 BDNF V95M 1 5.2 21 probably damaging Damaging 0
rs3817334 11 1000G rs1064608 0.809 MTCH2 P290A 1 5.1 27 probably damaging Tolerated 0.12
rs3888190 16 1000G rs180743 0.789 APOBR P428A 0.1 0.5 27 benign Unknown Not scored
rs3888190 16 1000G rs7498665 1 SH2B1 T484A 1 3.1 58 benign Tolerated 0.25
rs16951275 15 1000G rs7170185 1 LBXCOR1 W200R - - - - - -
rs1808579 18 1000G rs1805082 0.935 NPC1 I858V 1 6.1 29 benign Tolerated 0.24
rs1808579 18 1000G rs1805081 0.905 NPC1 H215R 0 −1.1 29 benign Tolerated 0.59
rs2287019 19 1000G rs1800437 0.714 GIPR E354Q 1 3.1 29 probably damaging Tolerated 0.09

r2 is the LD between the BMI index SNP and the putative coding variant.

Supplementary Material

1
2

Acknowledgements

A full list of acknowledgments can be found in the Supplementary Information.

Footnotes

Online Content Methods, along with any additional Extended Data display items and Source Data, are available in the online version of the paper; references unique to these sections appear only in the online paper.

Supplementary Information is available in the online version of the paper.

Author Contributions A full list of author contributions can be found in the Supplementary Information.

Author Information Reprints and permissions information is available at www.nature.com/reprints.

The authors declare competing financial interests: details are available in the online version of the paper. Readers are welcome to comment on the online version of the paper.

References

  • 1.Maes HH, Neale MC, Eaves LJ. Genetic and environmental factors in relative body weight and human adiposity. Behav. Genet. 1997;27:325–351. doi: 10.1023/a:1025635913927. [DOI] [PubMed] [Google Scholar]
  • 2.Visscher PM, Brown MA, McCarthy MI, Yang J. Five years of GWAS discovery. Am. J. Hum. Genet. 2012;90:7–24. doi: 10.1016/j.ajhg.2011.11.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Zaitlen N, et al. Using extended genealogy to estimate components of heritability for 23 quantitative and dichotomous traits. PLoS Genet. 2013;9:e1003520. doi: 10.1371/journal.pgen.1003520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Fall T, Ingelsson E. Genome-wide association studies of obesity and metabolic syndrome. Mol. Cell. Endocrinol. 2014;382:740–757. doi: 10.1016/j.mce.2012.08.018. [DOI] [PubMed] [Google Scholar]
  • 5.Speliotes EK, et al. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nature Genet. 2010;42:937–948. doi: 10.1038/ng.686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Willer CJ, et al. Six new loci associated with body mass index highlight a neuronal influence on body weight regulation. Nature Genet. 2009;41:25–34. doi: 10.1038/ng.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Voight BF, et al. The metabochip, a custom genotyping array for genetic studies of metabolic, cardiovascular, and anthropometric traits. PLoS Genet. 2012;8:e1002793. doi: 10.1371/journal.pgen.1002793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kilpeläinen TO, et al. Genetic variation near IRS1 associates with reduced adiposity and an impaired metabolic profile. Nature Genet. 2011;43:753–760. doi: 10.1038/ng.866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bradfield JP, et al. A genome-wide association meta-analysis identifies new childhood obesity loci. Nature Genet. 2012;44:526–531. doi: 10.1038/ng.2247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Monda KL, et al. A meta-analysis identifies new loci associated with body mass index in individuals of African ancestry. Nature Genet. 2013;45:690–696. doi: 10.1038/ng.2608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Berndt SI, et al. Genome-wide meta-analysis identifies 11 new loci for anthropometric traits and provides insights into genetic architecture. Nature Genet. 2013;45:501–512. doi: 10.1038/ng.2606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Guo Y, et al. Gene-centric meta-analyses of 108 912 individuals confirm known body mass index loci and reveal three novel signals. Hum. Mol. Genet. 2013;22:184–201. doi: 10.1093/hmg/dds396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wood AR, et al. Defining the role of common variation in the genomic and biological architecture of adult human height. Nature Genet. 2014;46:1173–1186. doi: 10.1038/ng.3097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Maller JB, et al. Bayesian refinement of association signals for 14 loci in 3 common diseases. Nature Genet. 2012;44:1294–1301. doi: 10.1038/ng.2435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wakefield J. A Bayesian measure of the probability of false discovery in genetic epidemiology studies. Am. J. Hum. Genet. 2007;81:208–227. doi: 10.1086/519024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Peters U, et al. A systematic mapping approach of 16q12.2/FTO and BMI in more than 20,000 African Americans narrows in on the underlying functional variation: results from the Population Architecture using Genomics and Epidemiology (PAGE) study. PLoS Genet. 2013;9:e1003171. doi: 10.1371/journal.pgen.1003171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Juster FT, Suzman R. An overview of the Health and Retirement Study. J. Hum. Resour. 1995;30:S7–S56. [Google Scholar]
  • 18.Bouchonville M, et al. Weight loss, exercise or both and cardiometabolic risk factors in obese older adults: results of a randomized controlled trial. Int. J. Obes. 2013;38:423–431. doi: 10.1038/ijo.2013.122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Yang J, Lee SH, Goddard ME, Visscher PM. GCTA: a tool for genome-wide complex trait analysis. Am. J. Hum. Genet. 2011;88:76–82. doi: 10.1016/j.ajhg.2010.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yang J, et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nature Genet. 2012;44:369–375. doi: 10.1038/ng.2213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Pers T, et al. Biological interpretation of genome-wide association studies using predicted gene functions. Nat. Commun. 2014;5:5890. doi: 10.1038/ncomms6890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.The ENCODE Project Consortium An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57–74. doi: 10.1038/nature11247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bernstein BE, et al. The NIH Roadmap Epigenomics Mapping Consortium. Nature Biotechnol. 2010;28:1045–1048. doi: 10.1038/nbt1010-1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Segrè AV, Groop L, Mootha VK, Daly MJ, Altshuler D. Common inherited variation in mitochondrial genes is not enriched for associations with type 2 diabetes or related glycemic traits. PLoS Genet. 2010;6:e1001058. doi: 10.1371/journal.pgen.1001058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell. 2006;124:471–484. doi: 10.1016/j.cell.2006.01.016. [DOI] [PubMed] [Google Scholar]
  • 26.Lango Allen H, et al. Hundreds of variants clustered in genomic loci and biological pathways affect human height. Nature. 2010;467:832–838. doi: 10.1038/nature09410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Raychaudhuri S, et al. Identifying relationships among genomic disease regions: predicting genes at pathogenic SNP associations and rare deletions. PLoS Genet. 2009;5:e1000534. doi: 10.1371/journal.pgen.1000534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Mägi R, et al. Contribution of 32 GWAS-identified common variants to severe obesity in European adults referred for bariatric surgery. PLoS ONE. 2013;8:e70735. doi: 10.1371/journal.pone.0070735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lee AW, et al. Functional inactivation of the genome-wide association study obesity gene neuronal growth regulator 1 in mice causes a body mass phenotype. PLoS ONE. 2012;7:e41537. doi: 10.1371/journal.pone.0041537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yang Y, Atasoy D, Su HH, Sternson SM. Hunger states switch a flip-flop memory circuit via a synaptic AMPK-dependent positive feedback loop. Cell. 2011;146:992–1003. doi: 10.1016/j.cell.2011.07.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wu Q, Clark MS, Palmiter RD. Deciphering a neuronal circuit that mediates appetite. Nature. 2012;483:594–597. doi: 10.1038/nature10899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Shen Y, Fu WY, Cheng EY, Fu AK, Ip NY. Melanocortin-4 receptor regulates hippocampal synaptic plasticity through a protein kinase A-dependent mechanism. J. Neurosci. 2013;33:464–472. doi: 10.1523/JNEUROSCI.3282-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Gibbs JW, III, Sombati S, DeLorenzo RJ, Coulter DA. Cellular actions of topiramate: blockade of kainate-evoked inward currents in cultured hippocampal neurons. Epilepsia. 2000;41(suppl. 1):S10–S16. doi: 10.1111/j.1528-1157.2000.tb02164.x. [DOI] [PubMed] [Google Scholar]
  • 34.Poulsen CF, et al. Modulation by topiramate of AMPA and kainate mediated calcium influx in cultured cerebral cortical, hippocampal and cerebellar neurons. Neurochem. Res. 2004;29:275–282. doi: 10.1023/b:nere.0000010456.92887.3b. [DOI] [PubMed] [Google Scholar]
  • 35.Henao-Mejia J, et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature. 2012;482:179–185. doi: 10.1038/nature10809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Pruim RJ, et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics. 2010;26:2336–2337. doi: 10.1093/bioinformatics/btq419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Frazer KA, et al. A second generation human haplotype map of over 3.1 million SNPs. Nature. 2007;449:851–861. doi: 10.1038/nature06258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Winkler TW, et al. Quality control and conduct of genome-wide association meta-analyses. Nature Protocols. 2014;9:1192–1212. doi: 10.1038/nprot.2014.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Willer CJ, Li Y, Abecasis GR. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics. 2010;26:2190–2191. doi: 10.1093/bioinformatics/btq340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Devlin B, Roeder K. Genomic control for association studies. Biometrics. 1999;55:997–1004. doi: 10.1111/j.0006-341x.1999.00997.x. [DOI] [PubMed] [Google Scholar]
  • 41.Wen W, et al. Meta-analysis identifies common variants associated with body mass index in east Asians. Nature Genet. 2012;44:307–311. doi: 10.1038/ng.1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Randall JC, et al. Sex-stratified genome-wide association studies including 270,000 individuals show sexual dimorphism in genetic loci for anthropometric traits. PLoS Genet. 2013;9:e1003500. doi: 10.1371/journal.pgen.1003500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38:e164. doi: 10.1093/nar/gkq603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Adzhubei IA, et al. A method and server for predicting damaging missense mutations. Nature Methods. 2010;7:248–249. doi: 10.1038/nmeth0410-248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.NHLBI Exome Sequencing Project (ESP) Exome Variant Server. http://evs.gs.washington.edu/EVS/ [Google Scholar]
  • 46.Ng PC. Henikoff, S. Predicting deleterious amino acid substitutions. Genome Res. 2001;11:863–874. doi: 10.1101/gr.176601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mills RE, et al. Mapping copy number variation by population-scale genome sequencing. Nature. 2011;470:59–65. doi: 10.1038/nature09708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Emilsson V, et al. Genetics of gene expression and its effect on disease. Nature. 2008;452:423–428. doi: 10.1038/nature06758. [DOI] [PubMed] [Google Scholar]
  • 49.Zhong H, Yang X, Kaplan LM, Molony C, Schadt EE. Integrating pathway analysis and genetics of gene expression for genome-wide association studies. Am. J. Hum. Genet. 2010;86:581–591. doi: 10.1016/j.ajhg.2010.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Grundberg E, et al. Mapping cis- and trans-regulatory effects across multiple tissues in twins. Nature Genet. 2012;44:1084–1089. doi: 10.1038/ng.2394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Dixon AL, et al. A genome-wide association study of global gene expression. Nature Genet. 2007;39:1202–1207. doi: 10.1038/ng2109. [DOI] [PubMed] [Google Scholar]
  • 52.Fehrmann RS, et al. Trans-eQTLs reveal that independent genetic variants associated with a complex phenotype converge on intermediate genes, with a major role for the HLA. PLoS Genet. 2011;7:e1002197. doi: 10.1371/journal.pgen.1002197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Nelis M, et al. Genetic structure of Europeans: a view from the North-East. PLoS ONE. 2009;4:e5472. doi: 10.1371/journal.pone.0005472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Myers AJ, et al. A survey of genetic human cortical gene expression. Nature Genet. 2007;39:1494–1499. doi: 10.1038/ng.2007.16. [DOI] [PubMed] [Google Scholar]
  • 55.Westra HJ, et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nature Genet. 2013;45:1238–1243. doi: 10.1038/ng.2756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Morris AP, et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nature Genet. 2012;44:981–990. doi: 10.1038/ng.2383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Deloukas P, et al. Large-scale association analysis identifies new risk loci for coronary artery disease. Nature Genet. 2013;45:25–33. doi: 10.1038/ng.2480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ehret GB, et al. Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature. 2011;478:103–109. doi: 10.1038/nature10405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Shungin D, et al. New genetic loci link adipose and insulin biology to body fat distribution. Nature. (this issue) doi: 10.1038/nature14132. http://dx.doi.org/nature14132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Willer C, et al. Discovery and refinement of loci associated with lipid levels. Nature Genet. 2013;45:1274–1283. doi: 10.1038/ng.2797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Scott RA, et al. Large-scale association analyses identify new loci influencing glycemic traits and provide insight into the underlying biological pathways. Nature Genet. 2012;44:991–1005. doi: 10.1038/ng.2385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Manning AK, et al. A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance. Nature Genet. 2012;44:659–669. doi: 10.1038/ng.2274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Saxena R, et al. Genetic variation in GIPR influences the glucose and insulin responses to an oral glucose challenge. Nature Genet. 2010;42:142–148. doi: 10.1038/ng.521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Dastani Z, et al. Novel loci for adiponectin levels and their influence on type 2 diabetes and metabolic traits: a multi-ethnic meta-analysis of 45,891 individuals. PLoS Genet. 2012;8:e1002607. doi: 10.1371/journal.pgen.1002607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Pattaro C, et al. Genome-wide association and functional follow-up reveals new loci for kidney function. PLoS Genet. 2012;8:e1002584. doi: 10.1371/journal.pgen.1002584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Böger CA, et al. CUBN is a gene locus for albuminuria. J. Am. Soc. Nephrol. 2011;22:555–570. doi: 10.1681/ASN.2010060598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Stolk L, et al. Meta-analyses identify 13 loci associated with age at menopause and highlight DNA repair and immune pathways. Nature Genet. 2012;44:260–268. doi: 10.1038/ng.1051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Elks CE, et al. Thirty new loci for age at menarche identified by a meta-analysis of genome-wide association studies. Nature Genet. 2010;42:1077–1085. doi: 10.1038/ng.714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Williams WW, et al. Association testing of previously reported variants in a large case-control meta-analysis of diabetic nephropathy. Diabetes. 2012;61:2187–2194. doi: 10.2337/db11-0751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sandholm N, et al. New susceptibility loci associated with kidney disease in type 1 diabetes. PLoS Genet. 2012;8:e1002921. doi: 10.1371/journal.pgen.1002921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hindorff LA, et al. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc. Natl Acad. Sci. USA. 2009;106:9362–9367. doi: 10.1073/pnas.0903103106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Li Q, Brown JB, Huang H, Bickel PJ. Measuring reproducibility of high-throughput experiments. Ann. Appl. Stat. 2011;5:1752–1779. [Google Scholar]
  • 73.Feng J, Liu T, Qin B, Zhang Y, Liu XS. Identifying ChIP-seq enrichment using MACS. Nature Protocols. 2012;7:1728–1740. doi: 10.1038/nprot.2012.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Abecasis GR, et al. An integrated map of genetic variation from 1,092 human genomes. Nature. 2012;491:56–65. doi: 10.1038/nature11632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Fehrmann RS, et al. Gene expression analysis identifies global gene dosage sensitivity in cancer. Nature Genet. 2015;47:115–125. doi: 10.1038/ng.3173. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2

RESOURCES