Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Protein-coding variants implicate novel genes related to lipid homeostasis contributing to body-fat distribution

This article has been updated

Abstract

Body-fat distribution is a risk factor for adverse cardiovascular health consequences. We analyzed the association of body-fat distribution, assessed by waist-to-hip ratio adjusted for body mass index, with 228,985 predicted coding and splice site variants available on exome arrays in up to 344,369 individuals from five major ancestries (discovery) and 132,177 European-ancestry individuals (validation). We identified 15 common (minor allele frequency, MAF ≥5%) and nine low-frequency or rare (MAF <5%) coding novel variants. Pathway/gene set enrichment analyses identified lipid particle, adiponectin, abnormal white adipose tissue physiology and bone development and morphology as important contributors to fat distribution, while cross-trait associations highlight cardiometabolic traits. In functional follow-up analyses, specifically in Drosophila RNAi-knockdowns, we observed a significant increase in the total body triglyceride levels for two genes (DNAH10 and PLXND1). We implicate novel genes in fat distribution, stressing the importance of interrogating low-frequency and protein-coding variants.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Summary of meta-analysis study design and workflow.
Fig. 2: Minor allele frequency compared to estimated effect.
Fig. 3: Regional association plots for known loci with novel coding signals identified by conditional analyses.
Fig. 4: Heat maps showing DEPICT gene set enrichment results from the stage 1 all ancestry sex-combined individuals (N = 344,369).

Similar content being viewed by others

Data availability

Summary statistics of all analyses are available at https://portals.broadinstitute.org/collaboration/giant/index.php/GIANT_consortium_data_files.

Change history

  • 07 March 2019

    In the HTML version of the article originally published, the link for Supplementary Data 5 returned the file for Supplementary Data 7. The error has been corrected in the HTML version of the article.

References

  1. Pischon, T. et al. General and abdominal adiposity and risk of death in Europe. N. Engl. J. Med. 359, 2105–2120 (2008).

    Article  CAS  PubMed  Google Scholar 

  2. Wang, Y., Rimm, E. B., Stampfer, M. J., Willett, W. C. & Hu, F. B. Comparison of abdominal adiposity and overall obesity in predicting risk of type 2 diabetes among men. Am. J. Clin. Nutr. 81, 555–563 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Canoy, D. Distribution of body fat and risk of coronary heart disease in men and women. Curr. Opin. Cardiol. 23, 591–598 (2008).

    Article  PubMed  Google Scholar 

  4. Snijder, M. B. et al. Associations of hip and thigh circumferences independent of waist circumference with the incidence of type 2 diabetes: the Hoorn Study. Am. J. Clin. Nutr. 77, 1192–1197 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Yusuf, S. et al. Obesity and the risk of myocardial infarction in 27,000 participants from 52 countries: a case-control study. Lancet 366, 1640–1649 (2005).

    Article  PubMed  Google Scholar 

  6. Mason, C., Craig, C. L. & Katzmarzyk, P. T. Influence of central and extremity circumferences on all-cause mortality in men and women. Obesity. 16, 2690–2695 (2008).

    Article  PubMed  Google Scholar 

  7. Karpe, F. & Pinnick, K. E. Biology of upper-body and lower-body adipose tissue—llink to whole-body phenotypes. Nat. Rev. Endocrinol. 11, 90–100 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Manolopoulos, K. N., Karpe, F. & Frayn, K. N. Gluteofemoral body fat as a determinant of metabolic health. Int. J. Obes. 34, 949–959 (2010).

    Article  CAS  Google Scholar 

  9. Emdin, C. A. et al. Genetic association of waist-to-hip ratio with cardiometabolic traits, type 2 diabetes, and coronary heart disease. JAMA 317, 626–634 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Shungin, D. et al. New genetic loci link adipose and insulin biology to body fat distribution. Nature 518, 187–196 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Winkler, T. W. et al. The influence of age and sex on genetic associations with adult body size and shape: a large-scale genome-wide interaction study. PLoS Genet. 11, e1005378 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Wen, W. et al. Genome-wide association studies in east asians identify new loci for waist-hip ratio and waist circumference. Sci. Rep. 6, 17958 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gao, C. et al. A comprehensive analysis of common and rare variants to identify adiposity loci in hispanic Americans: the iras family study (IRASFS). PLoS ONE 10, e0134649 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Graff, M. et al. Genome-wide physical activity interactions in adiposity—meta-analysis of 200,452 adults. PLoS Genet. 13, e1006528 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Justice, A. E. et al. Genome-wide meta-analysis of 241,258 adults accounting for smoking behaviour identifies novel loci for obesity traits. Nat. Commun. 8, 14977 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Ng, M. C. Y. et al. Discovery and fine-mapping of adiposity loci using high density imputation of genome-wide association studies in individuals of African ancestry: African Ancestry Anthropometry Genetics Consortium. PLoS Genet. 13, e1006719 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Aschard, H., Vilhjalmsson, B. J., Joshi, A. D., Price, A. L. & Kraft, P. Adjusting for heritable covariates can bias effect estimates in genome-wide association studies. Am. J. Hum. Genet. 96, 329–339 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Day, F. R., Loh, P. R., Scott, R. A., Ong, K. K. & Perry, J. R. A robust example of collider bias in a genetic association study. Am. J. Hum. Genet. 98, 392–393 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Feng, S., Liu, D., Zhan, X., Wing, M. K. & Abecasis, G. R. RAREMETAL: fast and powerful meta-analysis for rare variants. Bioinformatics 30, 2828–2829 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Pers, T. H. et al. Biological interpretation of genome-wide association studies using predicted gene functions. Nat. Commun. 6, 5890 (2015).

    Article  CAS  PubMed  Google Scholar 

  21. Marouli, E. et al. Rare and low-frequency coding variants alter human adult height. Nature 542, 186–190 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lamparter, D., Marbach, D., Rueedi, R., Kutalik, Z. & Bergmann, S. Fast and rigorous computation of gene and pathway scores from SNP-based summary statistics. PLoS Comput. Biol. 12, e1004714 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Locke, A. E. et al. Genetic studies of body mass index yield new insights for obesity biology. Nature 518, 197–206 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kawai, M., de Paula, F. J. & Rosen, C. J. New insights into osteoporosis: the bone-fat connection. J. Intern. Med. 272, 317–329 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Turcot, V. et al. Protein-altering variants associated with body mass index implicate pathways that control energy intake and expenditure in obesity. Nat. Genet. 50, 26–41 (2018).

    Article  CAS  PubMed  Google Scholar 

  26. Liu, D. J. et al. Exome-wide association study of plasma lipids in >300,000 individuals. Nat. Genet. 49, 1758–1766 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kraja, A. T. et al. New blood pressure-associated loci identified in meta-analyses of 475 000 individuals. Circ. Cardiovasc. Genet. 10, e001778 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Mahajan, A. et al. Identification and functional characterization of G6PC2 coding variants influencing glycemic traits define an effector transcript at the G6PC2-ABCB11 locus. PLoS Genet. 11, e1004876 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Manning, A. et al. A low-frequency inactivating akt2 variant enriched in the finnish population is associated with fasting insulin levels and type 2 diabetes risk. Diabetes 66, 2019–2032 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhao, W. et al. Identification of new susceptibility loci for type 2 diabetes and shared etiological pathways with coronary heart disease. Nat. Genet. 49, 1450–1457 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Morris, A. P. et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat. Genet. 44, 981–990 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ng, M. C. et al. Meta-analysis of genome-wide association studies in African Americans provides insights into the genetic architecture of type 2 diabetes. PLoS Genet. 10, e1004517 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Mahajan, A. et al. Genome-wide trans-ancestry meta-analysis provides insight into the genetic architecture of type 2 diabetes susceptibility. Nat. Genet. 46, 234–244 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Saxena, R. et al. Genome-wide association study identifies a novel locus contributing to type 2 diabetes susceptibility in Sikhs of Punjabi origin from India. Diabetes 62, 1746–1755 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Cook, J. P. & Morris, A. P. Multi-ethnic genome-wide association study identifies novel locus for type 2 diabetes susceptibility. Eur. J. Hum. Genet. 24, 1175–1180 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Voight, B. F. et al. Twelve type 2 diabetes susceptibility loci identified through large-scale association analysis. Nat. Genet. 42, 579–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Burdett, T. et al. The NHGRI-EBI Catalog of published genome-wide association studies. V.1.0 edn Vol. 2015 (NHGRI-EBI, 2015).

  38. Hindorff, L. A. et al. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc. Natl Acad. Sci. USA 106, 9362–9367 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lutoslawska, G. et al. Relationship between the percentage of body fat and surrogate indices of fatness in male and female Polish active and sedentary students. J. Physiol. Anthropol. 33, 10 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Verma, M., Rajput, M., Sahoo, S. S., Kaur, N. & Rohilla, R. Correlation between the percentage of body fat and surrogate indices of obesity among adult population in rural block of Haryana. J. Family Med. Prim. Care 5, 154–159 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Pereira, P. F. et al. Measurements of location of body fat distribution: an assessment of colinearity with body mass, adiposity and stature in female adolescents. Rev. Paul. Pediatr. 33, 63–71 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Lu, Y. et al. New loci for body fat percentage reveal link between adiposity and cardiometabolic disease risk. Nat. Commun. 7, 10495 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Chambers, J. C. et al. Common genetic variation near MC4R is associated with waist circumference and insulin resistance. Nat. Genet. 40, 716–718 (2008).

    Article  CAS  PubMed  Google Scholar 

  44. Nead, K. T. et al. Contribution of common non-synonymous variants in PCSK1 to body mass index variation and risk of obesity: a systematic review and meta-analysis with evidence from up to 331 175 individuals. Hum. Mol. Genet. 24, 3582–3594 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pospisilik, J. A. et al. Drosophila genome-wide obesity screen reveals hedgehog as a determinant of brown versus white adipose cell fate. Cell 140, 148–160 (2010).

    Article  CAS  PubMed  Google Scholar 

  46. Consortium, G. T. Human genomics. The genotype-tissue expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).

    Article  CAS  Google Scholar 

  47. Baraille, F., Planchais, J., Dentin, R., Guilmeau, S. & Postic, C. Integration of chrebp-mediated glucose sensing into whole body metabolism. Physiology 30, 428–437 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Kursawe, R. et al. Decreased transcription of ChREBP-alpha/beta isoforms in abdominal subcutaneous adipose tissue of obese adolescents with prediabetes or early type 2 diabetes: associations with insulin resistance and hyperglycemia. Diabetes 62, 837–844 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lotta, L. A. et al. Integrative genomic analysis implicates limited peripheral adipose storage capacity in the pathogenesis of human insulin resistance. Nat. Genet. 49, 17–26 (2017).

    Article  CAS  PubMed  Google Scholar 

  50. Cargill, M. et al. A large-scale genetic association study confirms IL12B and leads to the identification of IL23R as psoriasis-risk genes. Am. J. Hum. Genet. 80, 273–290 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Hazlett, J. et al. IL-23R rs11209026 polymorphism modulates IL-17A expression in patients with rheumatoid arthritis. Genes Immun. 13, 282–287 (2012).

    Article  CAS  Google Scholar 

  52. Karaderi, T. et al. Association between the interleukin 23 receptor and ankylosing spondylitis is confirmed by a new UK case-control study and meta-analysis of published series. Rheumatology 48, 386–389 (2009).

    Article  CAS  PubMed  Google Scholar 

  53. Duerr, R. H. et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 314, 1461–1463 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Abdollahi, E., Tavasolian, F., Momtazi-Borojeni, A. A., Samadi, M. & Rafatpanah, H. Protective role of R381Q (rs11209026) polymorphism in IL-23R gene in immune-mediated diseases: comprehensive review. J. Immunotoxicol. 13, 286–300 (2016).

    Article  CAS  PubMed  Google Scholar 

  55. Abraham, C., Dulai, P. S., Vermeire, S. & Sandborn, W. J. Lessons learned from trials targeting cytokine pathways in patients with inflammatory bowel diseases. Gastroenterology 152, 374–388 e4 (2017).

    Article  CAS  PubMed  Google Scholar 

  56. Molinelli, E., Campanati, A., Ganzetti, G. & Offidani, A. Biologic therapy in immune mediated inflammatory disease: basic science and clinical concepts. Curr. Drug Saf. 11, 35–43 (2016).

    Article  CAS  PubMed  Google Scholar 

  57. Fuchsberger, C. et al. The genetic architecture of type 2 diabetes. Nature 536, 41–47 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Wells, J. C. Sexual dimorphism of body composition. Best. Pract. Res. Clin. Endocrinol. Metab. 21, 415–430 (2007).

    Article  PubMed  Google Scholar 

  59. Loomba-Albrecht, L. A. & Styne, D. M. Effect of puberty on body composition. Curr. Opin. Endocrinol. Diabetes. Obes. 16, 10–15 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Rogol, A. D., Roemmich, J. N. & Clark, P. A. Growth at puberty. J. Adolesc. Health 31, 192–200 (2002).

    Article  PubMed  Google Scholar 

  61. Gibson, G. Rare and common variants: twenty arguments. Nat. Rev. Genet. 13, 135–145 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Stern, J. H., Rutkowski, J. M. & Scherer, P. E. Adiponectin, leptin, and fatty acids in the maintenance of metabolic homeostasis through adipose tissue crosstalk. Cell. Metab. 23, 770–784 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Dewey, F. E. et al. Inactivating variants in angptl4 and risk of coronary artery disease. N. Engl. J. Med. 374, 1123–1133 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Bondestam, J. et al. cDNA cloning, expression studies and chromosome mapping of human type I serine/threonine kinase receptor ALK7 (ACVR1C). Cytogenet. Cell Genet. 95, 157–162 (2001).

    Article  CAS  PubMed  Google Scholar 

  65. Jornvall, H., Blokzijl, A., ten Dijke, P. & Ibanez, C. F. The orphan receptor serine/threonine kinase ALK7 signals arrest of proliferation and morphological differentiation in a neuronal cell line. J. Biol. Chem. 276, 5140–5146 (2001).

    Article  CAS  PubMed  Google Scholar 

  66. Kim, B. C. et al. Activin receptor-like kinase-7 induces apoptosis through activation of MAPKs in a Smad3-dependent mechanism in hepatoma cells. J. Biol. Chem. 279, 28458–28465 (2004).

    Article  CAS  PubMed  Google Scholar 

  67. Watanabe, R. et al. The MH1 domains of smad2 and smad3 are involved in the regulation of the ALK7 signals. Biochem. Biophys. Res. Commun. 254, 707–712 (1999).

    Article  CAS  PubMed  Google Scholar 

  68. Kogame, M. et al. ALK7 is a novel marker for adipocyte differentiation. J. Med. Invest. 53, 238–245 (2006).

    Article  PubMed  Google Scholar 

  69. Murakami, M. et al. Expression of activin receptor-like kinase 7 in adipose tissues. Biochem. Genet. 51, 202–210 (2013).

    Article  CAS  PubMed  Google Scholar 

  70. Carlsson, L. M. et al. ALK7 expression is specific for adipose tissue, reduced in obesity and correlates to factors implicated in metabolic disease. Biochem. Biophys. Res. Commun. 382, 309–314 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Carithers, L. J. & Moore, H. M. The genotype-tissue expression (GTEx) project. Biopreserv. Biobank. 13, 307–308 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Yogosawa, S., Mizutani, S., Ogawa, Y. & Izumi, T. Activin receptor-like kinase 7 suppresses lipolysis to accumulate fat in obesity through downregulation of peroxisome proliferator-activated receptor gamma and C/EBPalpha. Diabetes 62, 115–123 (2013).

    Article  CAS  PubMed  Google Scholar 

  73. Yogosawa, S. & Izumi, T. Roles of activin receptor-like kinase 7 signaling and its target, peroxisome proliferator-activated receptor gamma, in lean and obese adipocytes. Adipocyte 2, 246–250 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Seifi, M., Ghasemi, A., Namipashaki, A. & Samadikuchaksaraei, A. Is C771G polymorphism of MLX interacting protein-like (MLXIPL) gene a novel genetic risk factor for non-alcoholic fatty liver disease? Cell Mol. Biol. 60, 37–42 (2014).

    CAS  PubMed  Google Scholar 

  75. Cairo, S., Merla, G., Urbinati, F., Ballabio, A. & Reymond, A. WBSCR14, a gene mapping to the Williams–Beuren syndrome deleted region, is a new member of the Mlx transcription factor network. Hum. Mol. Genet. 10, 617–627 (2001).

    Article  CAS  PubMed  Google Scholar 

  76. Ambele, M. A., Dessels, C., Durandt, C. & Pepper, M. S. Genome-wide analysis of gene expression during adipogenesis in human adipose-derived stromal cells reveals novel patterns of gene expression during adipocyte differentiation. Stem Cell Res. 16, 725–734 (2016).

    Article  CAS  PubMed  Google Scholar 

  77. Marchler-Bauer, A. et al. CDD: NCBI’s conserved domain database. Nucleic Acids Res. 43, D222–D226 (2015).

    Article  CAS  PubMed  Google Scholar 

  78. Toyofuku, T. et al. Semaphorin-4A, an activator for T-cell-mediated immunity, suppresses angiogenesis via Plexin-D1. EMBO J. 26, 1373–1384 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Gitler, A. D., Lu, M. M. & Epstein, J. A. PlexinD1 and semaphorin signaling are required in endothelial cells for cardiovascular development. Dev. Cell. 7, 107–116 (2004).

    Article  CAS  PubMed  Google Scholar 

  80. Luchino, J. et al. Semaphorin 3E suppresses tumor cell death triggered by the plexin D1 dependence receptor in metastatic breast cancers. Cancer Cell. 24, 673–685 (2013).

    Article  CAS  PubMed  Google Scholar 

  81. Shimizu, I. et al. Semaphorin3E-induced inflammation contributes to insulin resistance in dietary obesity. Cell. Metab. 18, 491–504 (2013).

    Article  CAS  PubMed  Google Scholar 

  82. Verzijl, H. T., van der Zwaag, B., Cruysberg, J. R. & Padberg, G. W. Mobius syndrome redefined: a syndrome of rhombencephalic maldevelopment. Neurology 61, 327–333 (2003).

    Article  PubMed  Google Scholar 

  83. Verzijl, H. T., van der Zwaag, B., Lammens, M., ten Donkelaar, H. J. & Padberg, G. W. The neuropathology of hereditary congenital facial palsy vs Mobius syndrome. Neurology 64, 649–653 (2005).

    Article  CAS  PubMed  Google Scholar 

  84. Fujita, M., Reinhart, F. & Neutra, M. Convergence of apical and basolateral endocytic pathways at apical late endosomes in absorptive cells of suckling rat ileum in vivo. J. Cell Sci. 97(Pt 2), 385–394 (1990).

    Article  PubMed  Google Scholar 

  85. Briegel, W. Neuropsychiatric findings of mobius sequence—a review. Clin. Genet. 70, 91–97 (2006).

    Article  CAS  PubMed  Google Scholar 

  86. Ta-Shma, A. et al. Isolated truncus arteriosus associated with a mutation in the plexin-D1 gene. Am. J. Med. Genet. A. 161A, 3115–3120 (2013).

    Article  PubMed  CAS  Google Scholar 

  87. Mazzotta, C. et al. Plexin-D1/Semaphorin 3E pathway may contribute to dysregulation of vascular tone control and defective angiogenesis in systemic sclerosis. Arthritis Res. Ther. 17, 221 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Yang, W. J. et al. Semaphorin-3C signals through Neuropilin-1 and PlexinD1 receptors to inhibit pathological angiogenesis. EMBO Mol. Med. 7, 1267–1284 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Zygmunt, T. et al. Semaphorin-PlexinD1 signaling limits angiogenic potential via the VEGF decoy receptor sFlt1. Dev. Cell. 21, 301–314 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Kim, J., Oh, W. J., Gaiano, N., Yoshida, Y. & Gu, C. Semaphorin 3E-Plexin-D1 signaling regulates VEGF function in developmental angiogenesis via a feedback mechanism. Genes Dev. 25, 1399–1411 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Bertolino, P. et al. Activin B receptor ALK7 is a negative regulator of pancreatic beta-cell function. Proc. Natl Acad. Sci. USA 105, 7246–7251 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Haworth, K. E. et al. Methylation of the FGFR2 gene is associated with high birth weight centile in humans. Epigenomics 6, 477–491 (2014).

    Article  CAS  PubMed  Google Scholar 

  93. Chi, X. et al. Angiopoietin-like 4 modifies the interactions between lipoprotein lipase and its endothelial cell transporter GPIHBP1. J. Biol. Chem. 290, 11865–11877 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Catoire, M. et al. Fatty acid-inducible ANGPTL4 governs lipid metabolic response to exercise. Proc. Natl Acad. Sci. USA 111, E1043–E1052 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. van Raalte, D. H. et al. Angiopoietin-like protein 4 is differentially regulated by glucocorticoids and insulin in vitro and in vivo in healthy humans. Exp. Clin. Endocrinol. Diabetes. 120, 598–603 (2012).

    Article  PubMed  CAS  Google Scholar 

  96. Koster, A. et al. Transgenic angiopoietin-like (angptl)4 overexpression and targeted disruption of angptl4 and angptl3: regulation of triglyceride metabolism. Endocrinology 146, 4943–4950 (2005).

    Article  CAS  PubMed  Google Scholar 

  97. Thiagalingam, A. et al. RREB-1, a novel zinc finger protein, is involved in the differentiation response to Ras in human medullary thyroid carcinomas. Mol. Cell. Biol. 16, 5335–5345 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Bonomo, J. A. et al. The ras responsive transcription factor RREB1 is a novel candidate gene for type 2 diabetes associated end-stage kidney disease. Hum. Mol. Genet. 23, 6441–6447 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Thiagalingam, A., Lengauer, C., Baylin, S. B. & Nelkin, B. D. RREB1, a ras responsive element binding protein, maps to human chromosome 6p25. Genomics 45, 630–632 (1997).

    Article  CAS  PubMed  Google Scholar 

  100. Bisogno, T. et al. Cloning of the first sn1-DAG lipases points to the spatial and temporal regulation of endocannabinoid signaling in the brain. J. Cell Biol. 163, 463–468 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Global Lipids Genetics, C. et al. Discovery and refinement of loci associated with lipid levels. Nat. Genet. 45, 1274–1283 (2013).

    Article  CAS  Google Scholar 

  102. Kooner, J. S. et al. Genome-wide scan identifies variation in MLXIPL associated with plasma triglycerides. Nat. Genet. 40, 149–151 (2008).

    Article  CAS  PubMed  Google Scholar 

  103. Pan, L. A. et al. G771C polymorphism in the mlxipl gene is associated with a risk of coronary artery disease in the chinese: a case-control study. Cardiology 114, 174–178 (2009).

    Article  CAS  PubMed  Google Scholar 

  104. Kang, G., Leech, C. A., Chepurny, O. G., Coetzee, W. A. & Holz, G. G. Role of the cAMP sensor Epac as a determinant of KATP channel ATP sensitivity in human pancreatic beta-cells and rat INS-1 cells. J. Physiol. 586, 1307–1319 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Ji, Z., Mei, F. C. & Cheng, X. Epac, not PKA catalytic subunit, is required for 3T3-L1 preadipocyte differentiation. Front Biosci. 2, 392–398 (2010).

    Article  Google Scholar 

  106. Martini, C. N., Plaza, M. V. & Vila Mdel, C. PKA-dependent and independent cAMP signaling in 3T3-L1 fibroblasts differentiation. Mol. Cell. Endocrinol. 298, 42–47 (2009).

    Article  CAS  PubMed  Google Scholar 

  107. Petersen, R. K. et al. Cyclic AMP (cAMP)-mediated stimulation of adipocyte differentiation requires the synergistic action of Epac- and cAMP-dependent protein kinase-dependent processes. Mol. Cell. Biol. 28, 3804–3816 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Yan, J. et al. Enhanced leptin sensitivity, reduced adiposity, and improved glucose homeostasis in mice lacking exchange protein directly activated by cyclic AMP isoform 1. Mol. Cell. Biol. 33, 918–926 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Gesta, S. et al. Evidence for a role of developmental genes in the origin of obesity and body fat distribution. Proc. Natl Acad. Sci. USA 103, 6676–6681 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Gesta, S. et al. Mesodermal developmental gene Tbx15 impairs adipocyte differentiation and mitochondrial respiration. Proc. Natl Acad. Sci. USA 108, 2771–2776 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Lee, K. Y. et al. Tbx15 controls skeletal muscle fibre-type determination and muscle metabolism. Nat. Commun. 6, 8054 (2015).

    Article  CAS  PubMed  Google Scholar 

  112. Sudlow, C. et al. UK Biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 12, e1001779 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  113. Liu, D. J. et al. Meta-analysis of gene-level tests for rare variant association. Nat. Genet. 46, 200–204 (2014).

    Article  CAS  PubMed  Google Scholar 

  114. Goldstein, J. I. et al. zCall: a rare variant caller for array-based genotyping: genetics and population analysis. Bioinformatics 28, 2543–2545 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Winkler, T. W. et al. Quality control and conduct of genome-wide association meta-analyses. Nat. Protoc. 9, 1192–1212 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  116. Winkler, T. W. et al. EasyStrata: evaluation and visualization of stratified genome-wide association meta-analysis data. Bioinformatics 31, 259–261 (2015).

    Article  CAS  PubMed  Google Scholar 

  117. Purcell, S. M. et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature 506, 185–190 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Yang, J. et al. Genomic inflation factors under polygenic inheritance. Eur. J. Hum. Genet. 19, 807–812 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  119. Yang, J. et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat. Genet. 44, S1–S3 (2012).

    Article  CAS  Google Scholar 

  120. Marchini, J., Howie, B., Myers, S., McVean, G. & Donnelly, P. A new multipoint method for genome-wide association studies by imputation of genotypes. Nat. Genet. 39, 906–913 (2007).

    Article  CAS  PubMed  Google Scholar 

  121. Wellcome Trust Case Control, C. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661–678 (2007).

    Article  CAS  Google Scholar 

  122. Marchini, J. & Howie, B. Genotype imputation for genome-wide association studies. Nat. Rev. Genet. 11, 499–511 (2010).

    Article  CAS  PubMed  Google Scholar 

  123. Frey, B. J. & Dueck, D. Clustering by passing messages between data points. Science 315, 972–976 (2007).

    Article  CAS  PubMed  Google Scholar 

  124. Moayyeri, A., Hammond, C. J., Valdes, A. M. & Spector, T. D. Cohort profile: TwinsUK and healthy ageing twin study. Int. J. Epidemiol. 42, 76–85 (2013).

    Article  PubMed  Google Scholar 

  125. Boyd, A. et al. Cohort profile: the ‘children of the 90s’–the index offspring of the Avon Longitudinal Study of parents and children. Int. J. Epidemiol. 42, 111–127 (2013).

    Article  PubMed  Google Scholar 

  126. Kutalik, Z., Whittaker, J., Waterworth, D., Beckmann, J. S. & Bergmann, S. Novel method to estimate the phenotypic variation explained by genome-wide association studies reveals large fraction of the missing heritability. Genet. Epidemiol. 35, 341–349 (2011).

    Article  PubMed  Google Scholar 

  127. Billingsley, P. Probability and Measure (Wiley, New York, 1986).

    Google Scholar 

  128. Surendran, P. et al. Trans-ancestry meta-analyses identify rare and common variants associated with blood pressure and hypertension. Nat. Genet. 48, 1151–1161 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Nikpay, M. et al. A comprehensive 1000 Genomes-based genome-wide association meta-analysis of coronary artery disease. Nat. Genet. 47, 1121–1130 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Storey, J. D. & Tibshirani, R. Statistical significance for genomewide studies. Proc. Natl Acad. Sci. USA 100, 9440–9445 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Civelek, M. et al. Genetic regulation of adipose gene expression and cardio-metabolic traits. Am. J. Hum. Genet. 100, 428–443 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was primarily supported through funding from the National Institute of Health (NIH): 1K99HL130580, R01-DK089256, 2R01HD057194, U01HG007416, R01DK101855, T32 HL007055, KL2TR001109; and the American Heart Association (AHA): 13POST16500011 and 13GRNT16490017. Co-author Y. Jia recently passed away while this work was in process. This study was completed as part of the Genetic Investigation of ANtropometric Traits (GIANT) Consortium. This research has been conducted using the UK Biobank resource. A full list of acknowledgements is provided in the Supplementary Data 18.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

Writing Group: L.A.C., R.S.F., T.M.F., M. Graff, H.M.H., J.N.H., A.E.J., T.K., Z.K., C.M.L., R.J.F.L., Y.L., K.E.N., V.T., K.L.Y.

Data preparation group: T.A., I.B.B., T.E., S.F., M. Graff, H.M.H., A.E.J., T.K., D.J.L., K.S.L., A.E.L., R.J.F.L., Y.L., E. Marouli, N.G.D.M., C.M.-G, P.M., M.C.Y.N., M.A.R., S.S., C.S., K. Stirrups, V.T., S.V., S.M.W., T.W.W., K.L.Y., X.Z.

WHR meta-analyses: P.L.A., H.M.H., A.E.J., T.K., M. Graff, C.M.L., R.J.F.L., K.E.N., V.T., K.L.Y.

Pleiotropy working group: G.A., M. Boehnke, J.P.C., P.D., F.D., J.C.F., H.M.H., H.K., H.M.H., A.E.J., C.M.L., D.J.L., R.J.F.L., A. Mahajan, E. Marouli, G.M., M.I.M., P.B.M., G.M.P., J.R.B.P., K.S.R., X.S., S.W., J.W., C.J.W.

Phenome-wide association studies: L. Bastarache, J.C.D., A.G., A. Mahajan, M.I.M.

Gene-set enrichment analyses: S.B., R.S.F., J.N.H., Z.K., D.L., T.H.P., T.F.V.

eQTL analyses: C.K.R., Y.L., K.L.M.

Monogenic and syndromic gene enrichment analyses: H.M.H., A.K.M.

Fly Obesity Screen: A. Lempradl, J.A. Pospisilik.

Overseeing of contributing studies and consortia: (1958 Birth Cohort) P.D.; (Airwave) P.E.; (AMC PAS) G.K.H.; (Amish) J.R.O.; (ARIC) E.B.; (ARIC, Add Health) K.E.N.; (BRAVE) E.D.A., R.C.; (BRIGHT) P.B.M.; (CARDIA) M.F., P.J.S.; (Cebu Longitudinal Health and Nutrition Survey) K.L.M.; (CHD Exome + Consortium) A.S.B., J.M.M.H., D.F.R., J.D.; (CHES) R.V.; (Clear/eMERGE (Seattle)) G.P.J.; (CROATIA_Korcula) V.V., O. Polasek, I.R.; (deCODE) K. Stefansson, U.T.; (DHS) D.W.B.; (DIACORE) C.A.B.; (DPS) J.T., J. Lindström, M.U.; (DRSEXTRA) T.A.L., R.R.; (EFSOCH) A.T.H., T.M.F.; (EGCUT) T.E.; (eMERGE (Seattle)) E.B.L.; (EPIC-Potsdam) M.B.S., H.B.; (EpiHealth) E.I., P.W.F.; (EXTEND) A.T.H., T.M.F.; (Family Heart Study) I.B.B.; (Fenland, EPIC) R.A.S.; (Fenland, EPIC, InterAct) N.J.W., C.L.; (FINRISK) S. Männistö; (FINRISK 2007 (T2D)) P.J., V. Salomaa; (Framingham Heart Study) L.A.C.; (FUSION) M. Boehnke, F.S.C.; (FVG) P.G.; (Generation Scotland) C.H., B.H.S.; (Genetic Epidemiology Network of Arteriopathy (GENOA)) S.L.R.K.; (GRAPHIC) N.J.S.; (GSK-STABILITY) D.M.W., L.W., H.D.W.; (Health) A. Linneberg; (HELIC MANOLIS) E.Z., G.D.; (HELIC Pomak) E.Z., G.D.; (HUNT-MI) K.H., C.J.W.; (Inter99) T.H., T.J.; (IRASFS) L.E.W., E.K.S.; (Jackson Heart Study (JHS)) J.G.W.; (KORA S4) K. Strauch, I.M.H.; (Leipzig-Adults) M. Blüher, P. Kovacs.; (LOLIPOP-Exome) J.C.C., J.S.K.; (LOLIPOP-OmniEE) J.C.C., J.S.K.; (MESA) J.I.R., X.G.; (METSIM) J.K., M.L.; (MONICA-Brianza) G.C.; (Montreal Heart Institute Biobank (MHIBB)) M.P.D., G.L., S.d.D., J.C.T.; (MORGAM Central Laboratory) M.P.; (MORGAM Data Centre) K.K.; (OBB) F. Karpe; (PCOS) A.P.M., C.M.L.; (PIVUS) C.M.L., L.L.; (PRIME—Belfast) F. Kee; (PRIME—Lille) P.A.; (PRIME—Strasbourg) M.M.; (PRIME—Toulouse) J.F.; (PROMIS) D.S.; (quality control) M.A.R.; (RISC) B.B., E.F., M.W.; (Rotterdam Study I) A.G.U., M.A.I.; (SEARCH) A.M.D.; (SHIP/SHIP-Trend) M.D.; (SIBS) D.F.E.; (SOLID TIMI-52) D.M.W.; (SORBS) A.P.M., M.S., A. Tönjes; (The Mount Sinai BioMe Biobank) E.P.B., R.J.F.L.; (The NEO Study) D.O.M.K.; (The NHAPC Study, The GBTDS Study) X.L.; (The Western Australian Pregnancy Cohort (Raine) Study) C.E.P., S. Macgregor; (TwinsUK) T.D.S.; (ULSAM) A.P.M.; (Vejle Biobank) I.B., C.C., O. Pedersen; (WGHS) D.I.C., P.M.R.; (Women’s Health Initiative) P.L.A.; (WTCCC-UKT2D) M.I.M., K.R.O.; (YFS) T.L., O.T.R.

Genotyping of contributing studies and consortia: (1958 Birth Cohort) K.E.S.; (Airwave) E.E., M.S.L.; (AMC PAS) S.S.; (Amish) L.M.Y.A., J.A. Perry; (ARIC) E.W.D., M.L.G.; (BBMRI-NL) S.H.V., L. Broer, C.M.v.D., P.I.W.d.B.; (BRAVE) E.D.A.; (Cambridge Cancer Studies) J.G.D.; (CARDIA) M.F.; (CHD Exome + Consortium) A.S.B., J.M.M.H., D.F.R., J.D., R.Y.; (Clear/eMERGE (Seattle)) G.P.J.; (CROATIA_Korcula) V.V.; (DIACORE) C.A.B., M. Gorski; (DPS) A.U.J., J. Lindström; (DRSEXTRA) P. Komulainen; (EGCUT) T.E.; (EPIC-Potsdam) M.B.S., K.M.; (EpiHealth) E.I., P.W.F.; (Family Heart Study) K.D.T.; (Fenland, EPIC) R.A.S.; (Fenland, EPIC, InterAct) N.J.W., C.L.; (FUSION) N.N.; (FVG) I.G., A. Morgan; (Generation Scotland) C.H.; (Genetic Epidemiology Network of Arteriopathy (GENOA)) S.L.R.K., J.A.S.; (GRAPHIC) N.J.S.; (GSK-STABILITY) D.M.W.; (Health) J.B.J.; (HELIC MANOLIS) L. Southam; (HELIC Pomak) L. Southam; (Inter99) T.H., N.G.; (KORA) M.M.N.; (KORA S4) K. Strauch, H.G.; (Leipzig-Adults) A. Mahajan; (LOLIPOP-Exome) J.C.C., J.S.K.; (LOLIPOP-OmniEE) J.C.C., J.S.K.; (MESA) J.I.R., Y.D.I.C., K.D.T.; (METSIM) J.K., M.L.; (Montreal Heart Institute Biobank (MHIBB)) M.P.D.; (OBB) F. Karpe; (PCOS) A.P.M.; (PIVUS) C.M.L.; (Rotterdam Study I) A.G.U., C.M.G., F.R.; (SDC) J.M.J., H.V.; (SEARCH) A.M.D.; (SOLID TIMI-52) D.M.W.; (SORBS) A.P.M.; (The Mount Sinai BioMe Biobank) E.P.B., R.J.F.L., Y.L., C.S.; (The NEO Study) R.L.G.; (The NHAPC study, The GBTDS study) X.L., H.L., Y.H.; (The Western Australian Pregnancy Cohort (Raine) Study) C.E.P., S. Macgregor; (TUDR) Z.A.; (TwinsUK) A.P.M.; (ULSAM) A.P.M.; (WGHS) D.I.C., A.Y.C.; (Women’s Health Initiative) A.P.R.; (WTCCC-UKT2D) M.I.M.; (YFS) T.L., L.P.L.

Phenotyping of contributing studies and consortia: (Airwave) E.E.; (AMC PAS) S.S.; (Amish) L.M.Y.A.; (ARIC) E.W.D.; (ARIC, Add Health) K.E.N.; (BBMRI-NL) S.H.V.; (BRAVE) E.D.A.; (BRIGHT) M.J.C.; (CARL) A. Robino, G.G.; (Cebu Longitudinal Health and Nutrition Survey) N.R.L.; (CHES) R.V., M.T.; (Clear/eMERGE (Seattle)) G.P.J., A.A.B.; (CROATIA_Korcula) O. Polasek, I.R.; (DIACORE) C.A.B., B.K.K.; (DPS) A.U.J., J. Lindström; (EFSOCH) A.T.H.; (EGCUT) E. Mihailov; (EPIC-Potsdam) H.B.; (EpiHealth) E.I.; (EXTEND) A.T.H.; (Family Heart Study) M.F.F.; (Fenland, EPIC, InterAct) N.J.W.; (FIN-D2D 2007) L.M., M.V.; (FINRISK) S. Männistö; (FINRISK 2007 (T2D)) P.J., H.M.S.; (Framingham Heart Study) C.S.F.; (Generation Scotland) C.H., B.H.S.; (Genetic Epidemiology Network of Arteriopathy (GENOA)) S.L.R.K., J.A.S.; (GRAPHIC) N.J.S.; (GSK-STABILITY) L.W., H.D.W.; (Health) A. Linneberg, B.H.T.; (HELIC MANOLIS) L. Southam, A.E.F., E.T.; (HELIC Pomak) L. Southam, A.E.F., M.K.; (HUNT-MI) K.H., O.L.H.; (Inter99) T.J., N.G.; (IRASFS) L.E.W., B.K.; (KORA) M.M.N.; (BBMRI-NL) K.M.A.S.; (Leipzig-Adults) M. Blüher, P. Kovacs; (LOLIPOP-Exome) J.C.C., J.S.K.; (LOLIPOP-OmniEE) J.C.C., J.S.K.; (MESA) M.A.; (Montreal Heart Institute Biobank (MHIBB)) G.L., K.S.L., V.T.; (MORGAM Data Centre) K.K.; (OBB) F. Karpe, M.N.; (PCOS) C.M.L.; (PIVUS) L.L.; (PRIME—Belfast) F. Kee; (PRIME—Lille) P.A.; (PRIME—Strasbourg) M.M.; (PRIME—Toulouse) J.F.; (RISC) B.B., E.F.; (Rotterdam Study I) M.A.I., C.M.G., F.R., M.C.Z.; (SHIP/SHIP-Trend) N.F.; (SORBS) M.S., A. Tönjes; (The Mount Sinai BioMe Biobank) E.P.B., Y.L., C.S.; (The NEO Study) R.d.M.; (The NHAPC study, The GBTDS study) X.L., H.L., L. Sun, F.W.; (The Western Australian Pregnancy Cohort (Raine) Study) C.E.P.; (TUDR) Y.J.H., W.J.L.; (TwinsUK) T.D.S., K.S.S.; (ULSAM) V.G.; (WGHS) D.I.C., P.M.R.; (Women’s Health Initiative) A.P.R.; (WTCCC-UKT2D) M.I.M., K.R.O.; (YFS) T.L., O.T.R.

Data analysis of contributing studies and consortia: (1958 Birth Cohort) K.E.S., I.N.; (Airwave) E.E., M.S.L.; (AMC PAS) S.S.; (Amish) J.R.O., L.M.Y.A., J.A. Perry; (ARIC, Add Health) K.E.N., K.L.Y., M. Graff; (BBMRI-NL) L. Broer; (BRAVE) R.C., D.S.A.; (BRIGHT) H.R.W.; (Cambridge Cancer Studies) J.G.D., A.E., D.J.T.; (CARDIA) C.E.L, M.F., L.-A.L.; (CARL) A. Robino, D.V.; (Cebu Longitudinal Health and Nutrition Survey) Y.W.; (CHD Exome + Consortium) A.S.B., J.M.M.H., D.F.R., R.Y., P.S.; (CHES) Y.J.; (CROATIA_Korcula) V.V.; (deCODE) V. Steinthorsdottir, G.T.; (DHS) A.J.C., P.M., M.C.Y.N.; (DIACORE) C.A.B., M. Gorski; (EFSOCH) H.Y.; (EGCUT) T.E., R.M.; (eMERGE (Seattle)) D.S.C.; (ENDO) T.K.; (EPIC) J.H.Z.; (EPIC-Potsdam) K.M.; (EpiHealth) S.G.; (EXTEND) H.Y.; (Family Heart Study) M.F.F.; (Fenland) J.Luan.; (Fenland, EPIC) R.A.S.; (Fenland, InterAct) S.M.W.; (FIN-D2D 2007) M.V., L.M.; (Finrisk Extremes and quality control) S.V.; (Framingham Heart Study) C.T.L., N.L.H.C.; (FVG) I.G.; (Generation Scotland) C.H., J.M.; (Genetic Epidemiology Network of Arteriopathy (GENOA)) L.F.B.; (GIANT-Analyst) A.E.J.; (GRAPHIC) N.J.S., N.G.D.M., C.P.N.; (GSK-STABILITY) D.M.W., A.J.S.; (Health) J.B.J.; (HELIC MANOLIS) L. Southam; (HELIC Pomak) L. Southam; (HUNT-MI) W. Zhou; (Inter99) N.G.; (IRASFS) B.K.; (Jackson Heart Study (JHS)) L.A.L., J. Li; (KORA S4) T.W.W.; (BBMRI-NL) K.M.A.S.; (Leipzig-Adults) A. Mahajan; (LOLIPOP-Exome) J.C.C., J.S.K., W. Zhang; (LOLIPOP-OmniEE) J.C.C., J.S.K., Weihua Zhang; (MESA) J.I.R., X.G., J.Y.; (METSIM) X.S.; (Montreal Heart Institute Biobank (MHIBB)) J.C.T., G.L., K.S.L., V.T.; (OBB) A. Mahajan; (PCOS) A.P.M., T.K.; (PIVUS) N.R.R.; (PROMIS) A. Rasheed, W. Zhao; (quality control GoT2D/T2D-GENES (FUSION, METSIM and so on)) A.E.L.; (RISC) H.Y.; (Rotterdam Study I) C.M.G., F.R.; (SHIP/SHIP-Trend) A. Teumer; (SOLID TIMI-52) D.M.W., A.J.S.; (SORBS) A.P.M; (The Mount Sinai BioMe Biobank) Y.L., C.S.; (The NEO Study) R.L.G.; (The NHAPC study, The GBTDS study) X.L., H.L., Y.H., H.Z.; (The Western Australian Pregnancy Cohort (Raine) Study) C.A.W.; (UK Biobank) A.R.W.; (ULSAM) A.P.M., A. Mahajan; (WGHS) D.I.C., A.Y.C.; (Women’s Health Initiative) P.L.A., J.H.; (WTCCC-UKT2D) W.G.; (YFS) L.P.L.

Corresponding authors

Correspondence to Kari E. North or Cecilia M. Lindgren.

Ethics declarations

Competing interests

The authors declare the following competing interests: A.S.B. holds interest in AstraZeneca, Biogen, Bioverativ, Merck, Novartis and Pfizer. A.S.C. and C.S.F. are current employees of Merck. Authors affiliated with deCODE (V.St., G.T., U.T. and K.S.) are employed by deCODE Genetics/Amgen, Inc. H.D.W. has the following financial and non-financial competing interests to declare: research grants: Sanofi Aventis; Eli Lilly; NIH; Omthera Pharmaceuticals, Pfizer, Elsai Inc. AstraZeneca; DalCor and Services; lecture fees: Sanofi Aventis; advisory boards: Acetelion, Sirtex, CSL Boehring. J.D. has received grants from AstraZeneca, Biogen, Merck, Novartis and Pfizer. L.M.Y.A. and R.A.S. are employee stock holders of GlaxoSmithKline. M.P.D. received honoraria and holds minor equity in Dalcor. V.S. has participated in a conference trip sponsored by Novo Nordisk.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–24, Supplementary Tables 1–10 and Supplementary Note

Reporting Summary

Supplementary Data 1

Study design, number of individuals and sample quality control for ExomeChip study cohorts.

Supplementary Data 2

Information on genotyping methods, quality control of SNPs, imputation, and statistical analysis for ExomeChip study cohorts.

Supplementary Data 3

Study-specific descriptive statistics for anthropometric measurements in men and women of the ExomeChip cohorts.

Supplementary Data 4

Sex combined stage 1 and stage 2 results for all variants with initial stage 1 results that met P value

Supplementary Data 5

Male-Specific stage 1 and stage 2 results for all variants with initial stage 1 results that met P value

Supplementary Data 6

Female-Specific stage 1 and stage 2 results for all variants with initial stage 1 results that met P value

Supplementary Data 7

Conditional analysis and reciprocal

Supplementary Data 8

ExomeChip Data-driven ExpressionPrioritized Integration for Complex Traits (EC-DEPICT) results when including all variants associated WHRadjBMI with P value

Supplementary Data 9

ExomeChip Data-driven ExpressionPrioritized Integration for Complex Traits (EC-DEPICT) results when including all variants associated with WHRadjBMI with P value

Supplementary Data 10

Results from PASCAL (Pathway scoring algorithm) for Exomechip coding variants and genome-wide coding and regulatory variants associated with WHRadjBMI. Pascal (Pathway scoring algorithm) details and software are available at https://www2.unil.ch/cbg/index.php?title=Pascal

Supplementary Data 11

Associations between the WHRadjBMI that

Supplementary Data 12

Previously-reported associations with other traits in the GWAS Catalog for significant WHRadjBMI variants from the current study. This table lists all previously reported associations within 1 MB (+/- 500 kb) and in high LD (r2 >0.2) with our lead novel SNPs along with relevant annotation (for example miRNA target binding site, variant location relevant to nearest gene) reported in the GWAS Catalog.

Supplementary Data 13

Genes that have been linked to monogenic and syndromic obesity.

Supplementary Data 14

Exome variant - expression quantitative trait loci (eQTL) gene associations at false discovery rate

Supplementary Data 15

Detailed description for each of the variants that met array-wide significance in the stage 1 plus stage 2 sex-combined analyses, men only analyses, or women only analyses (see also Tables 1 and 2).

Supplementary Data 16

Exome variant - expression quantitative trait loci (eQTL) gene associations at false discovery rate <5% identified in the Genotype-Tissue Expression (GTEx) database. The results were extracted from the GTEx project (version 6):

Supplementary Data 17

Variant Effect Predictor (VEP) annotation

Supplementary Data 18

Detailed acknowledgements by study and/or contributing author(s).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Justice, A.E., Karaderi, T., Highland, H.M. et al. Protein-coding variants implicate novel genes related to lipid homeostasis contributing to body-fat distribution. Nat Genet 51, 452–469 (2019). https://doi.org/10.1038/s41588-018-0334-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-018-0334-2

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing