Integrative genomic analysis implicates limited peripheral adipose storage capacity in the pathogenesis of human insulin resistance

Luca A Lotta, Pawan Gulati, Felix R Day, Felicity Payne, Halit Ongen, Martijn van de Bunt, Kyle J Gaulton, John D Eicher, Stephen J Sharp, Jian'an Luan, Emanuella De Lucia Rolfe, Isobel D Stewart, Eleanor Wheeler, Sara M Willems, Claire Adams, Hanieh Yaghootkar, EPIC-InterAct Consortium, Cambridge FPLD1 Consortium, Nita G Forouhi, Kay-Tee Khaw, Andrew D Johnson, Robert K Semple, Timothy Frayling, John R B Perry, Emmanouil Dermitzakis, Mark I McCarthy, Inês Barroso, Nicholas J Wareham, David B Savage, Claudia Langenberg, Stephen O'Rahilly, Robert A Scott, Claudia Langenberg, Robert A Scott, Stephen J Sharp, Nita G Forouhi, Nicola D Kerrison, Matt Sims, Debora M E Lucarelli, Inês Barroso, Panos Deloukas, Mark I McCarthy, Larraitz Arriola, Beverley Balkau, Aurelio Barricarte, Heiner Boeing, Paul W Franks, Carlos Gonzalez, Sara Grioni, Rudolf Kaaks, Timothy J Key, Carmen Navarro, Peter M Nilsson, Kim Overvad, Domenico Palli, Salvatore Panico, J Ramón Quirós, Olov Rolandsson, Carlotta Sacerdote, Elena Salamanca-Fernández, Nadia Slimani, Anne Tjonneland, Rosario Tumino, Annemieke M W Spijkerman, Daphne L van der A, Yvonne T van der Schouw, Elio Riboli, Nicholas J Wareham, Robert K Semple, Claire Adams, Anna Stears, Stella George, Mark Walker, Ellie Gurnell, Deirdre Maguire, Rasha Mukhtar, Sath Nag, Amanda Adler, Maarten R Soeters, Ken Laji, Alistair Watt, Simon Aylwin, Andrew Johnson, Gerry Rayman, Fahmy Hanna, Sian Ellard, Richard Ross, Kristina Blaslov, Lea Smirčić Duvnjak, Stephen O'Rahilly, David B Savage, Luca A Lotta, Pawan Gulati, Felix R Day, Felicity Payne, Halit Ongen, Martijn van de Bunt, Kyle J Gaulton, John D Eicher, Stephen J Sharp, Jian'an Luan, Emanuella De Lucia Rolfe, Isobel D Stewart, Eleanor Wheeler, Sara M Willems, Claire Adams, Hanieh Yaghootkar, EPIC-InterAct Consortium, Cambridge FPLD1 Consortium, Nita G Forouhi, Kay-Tee Khaw, Andrew D Johnson, Robert K Semple, Timothy Frayling, John R B Perry, Emmanouil Dermitzakis, Mark I McCarthy, Inês Barroso, Nicholas J Wareham, David B Savage, Claudia Langenberg, Stephen O'Rahilly, Robert A Scott, Claudia Langenberg, Robert A Scott, Stephen J Sharp, Nita G Forouhi, Nicola D Kerrison, Matt Sims, Debora M E Lucarelli, Inês Barroso, Panos Deloukas, Mark I McCarthy, Larraitz Arriola, Beverley Balkau, Aurelio Barricarte, Heiner Boeing, Paul W Franks, Carlos Gonzalez, Sara Grioni, Rudolf Kaaks, Timothy J Key, Carmen Navarro, Peter M Nilsson, Kim Overvad, Domenico Palli, Salvatore Panico, J Ramón Quirós, Olov Rolandsson, Carlotta Sacerdote, Elena Salamanca-Fernández, Nadia Slimani, Anne Tjonneland, Rosario Tumino, Annemieke M W Spijkerman, Daphne L van der A, Yvonne T van der Schouw, Elio Riboli, Nicholas J Wareham, Robert K Semple, Claire Adams, Anna Stears, Stella George, Mark Walker, Ellie Gurnell, Deirdre Maguire, Rasha Mukhtar, Sath Nag, Amanda Adler, Maarten R Soeters, Ken Laji, Alistair Watt, Simon Aylwin, Andrew Johnson, Gerry Rayman, Fahmy Hanna, Sian Ellard, Richard Ross, Kristina Blaslov, Lea Smirčić Duvnjak, Stephen O'Rahilly, David B Savage

Abstract

Insulin resistance is a key mediator of obesity-related cardiometabolic disease, yet the mechanisms underlying this link remain obscure. Using an integrative genomic approach, we identify 53 genomic regions associated with insulin resistance phenotypes (higher fasting insulin levels adjusted for BMI, lower HDL cholesterol levels and higher triglyceride levels) and provide evidence that their link with higher cardiometabolic risk is underpinned by an association with lower adipose mass in peripheral compartments. Using these 53 loci, we show a polygenic contribution to familial partial lipodystrophy type 1, a severe form of insulin resistance, and highlight shared molecular mechanisms in common/mild and rare/severe insulin resistance. Population-level genetic analyses combined with experiments in cellular models implicate CCDC92, DNAH10 and L3MBTL3 as previously unrecognized molecules influencing adipocyte differentiation. Our findings support the notion that limited storage capacity of peripheral adipose tissue is an important etiological component in insulin-resistant cardiometabolic disease and highlight genes and mechanisms underpinning this link.

Conflict of interest statement

Competing Financial Interests Statement

The authors report no conflict of interest relative to this study.

Figures

Figure 1. Combined associations with detailed anthropometry…
Figure 1. Combined associations with detailed anthropometry and metabolic disease risk at the 53 genomic loci.
Panel A: association of the 53-SNP genetic score with anthropometric and glycaemic traits in meta-analyses of genetic association studies. Body mass index (BMI), waist-to-hip ratio (WHR), waist and hip circumference data are from the GIANT consortium and the UK Biobank study. Body fat percentage data are from the UK Biobank, EPIC-Norfolk and Fenland studies. Fasting plasma glucose (FPG), 2 hour glucose and HbA1c data are from the MAGIC consortium. Squares with error bars represent the per-allele beta coefficients in standard deviation units and their 95% confidence intervals. Panel B: association of genetic scores with compartmental body masses. Data are from 12,848 participants of the Fenland and EPIC-Norfolk studies who underwent a DEXA scan. Squares with error bars represent the per-allele beta coefficients in standard deviation units and their 95% confidence intervals.Panel C: association with lower levels of leg fat mass and higher hazard of incident type 2 diabetes by quintiles of the 53-SNP genetic risk scores. Associations are reported for individuals in the exposed category compared with the bottom quintile (reference category). Associations with leg fat mass are from 9,747 participants of the Fenland study and are reported on the left. Associations with incident type 2 diabetes are from 7,420 incident cases and 9,267 controls of the InterAct study and are reported on the right. Squares represent the beta coefficients in grams of leg fat mass (left plot) or the hazard ratio (HR) for incident type 2 diabetes (right plot) in each category compared with the lowest quintile. Error bars represent the 95% confidence intervals of these estimates. Panel D: associations of individual lead SNPs at eight loci with waist, hip circumference (left) and type 2 diabetes (right). Loci were selected on the basis of their genome-wide significant association with hip circumference or body fat percentage (i.e.PIK3R1). Waist and hip circumference analyses are from a meta-analysis of the GIANT and UK Biobank studies. Type 2 diabetes analyses are from a meta-analysis of the DIAGRAM, InterAct and UK Biobank studies. Squares with error bars represent the per-allele beta coefficients in standard deviation units of waist and hip circumference (left plot) or the per-allele odds ratio (OR) of type 2 diabetes (right plot). Error bars represent the 95% confidence intervals of these estimates. *Detailed associations at thePIK3R1 locus, which was primarily associated with lower body fat percentage, are presented in Supplementary Figure 9.
Figure 2. Associations at the 53 genomic…
Figure 2. Associations at the 53 genomic loci with familial partial lipodystrophy type 1 (FPLD1).
Panel A: distribution of leg fat mass as a function of the fat mass of the rest of the body (from DEXA) in women of the Fenland study at the extreme quintiles (Q) of the 53-SNP genetic score and in 14 FPLD1 subjects. Q1 represents a low genetic burden, whereas Q5 a high genetic burden. Lines of fit are plotted for each group. Panel B: histograms of the distribution of risk alleles in the FPLD1 subjects and in control women from the UKHLS study. Panel C: bi-dimensional box plots of the distribution of leg fat mass as a function of the distribution of the number of risk alleles in women of the Fenland study at the extreme quintiles (Q) of the 53-SNP genetic score and in FPLD1 subjects. Q1 represents a low genetic burden, whereas Q5 a high genetic burden. Each rectangle represents a group of individuals. For each dimension, the two sides of the rectangle represent the interquartile range and the central line the median. Data for obese women from Fenland were plotted to show the relationship between genetic risk and levels of leg fat in a group of women with a similar body mass index to that of FPLD1 patients.
Figure 3. Putative effector genes, tissues and…
Figure 3. Putative effector genes, tissues and cell types.
Panel A: schematic representation of some established components of the insulin signalling pathway with stars reporting the location in the pathway of putative effector genes, with their respective lead single nucleotide polymorphism listed. Panel B: associations of gain- and loss-of-function genetic variants in the LPL gene with type 2 diabetes. The reference number in parenthesis refers to the study reporting the association with triglycerides and coronary heart disease (see reference number 38 of this manuscript).Panel C: summary of evidence about links between genetic variants, lipodystrophy, insulin resistance, and type 2 diabetes at different levels of the population phenotypic distribution. *Rare syndromes caused by autosomal dominant INSR mutations are not usually associated with lipodystrophy and the INSR rs8101064 polymorphism is not associated with body fat percentage. Panel D: overlap of the 53 loci (lead SNPs plus proxy variants in r2>0.8) with chromatin state annotations from the NIH Roadmap. Panel E: DEPICT’s annotation of cell types and tissues on the basis of expression patterns in 37,427 human microarray samples. The y-axis represents the –log10(p-value) for enrichment of signal in a cell or tissue type attributed by DEPICT. The horizontal broken line represents the multiple-test corrected threshold of statistical significance (Bonferroni p=0.00072).
Figure 4. Experimental knockdown of putative effector…
Figure 4. Experimental knockdown of putative effector genes in cellular adipogenesis models and comparisons with phenotypic associations.
Panel A: results of experimental knockdown in OP9-K cells. Full circles represent the difference of the means from knockout experiments of a given gene compared with control experiments (n=4-7). Error bars represent the 95% confidence intervals of the difference of the means. Top graph: effect on mRNA levels of knockdown experiments of target genes using short interfering RNA (siRNA) in OP9-K cells. The two-tailed t-test p-values for differences in means were: IRS1, p=4.6 x 10-06; CCDC92, p=2.7 x 10-09, DNAH10, p=2.4 x 10-06;L3MBTL3, p=4.6 x 10-06; FAM13A, p=2.4 x 10-05. Bottom graph: effect on lipid accumulation in siRNA knockdown experiments. The two-tailed t-test p-values for differences in means were: IRS1, p=0.0047;CCDC92, p=1.2 x 10-05, DNAH10,p=0.0027; L3MBTL3, p=0.00013; FAM13A,p=0.92.Panel B: illustrative images showing florescence microscopy from lipid accumulation experiments. Red indicates adipored staining of neutral lipid, blue is hoechst staining of nuclei. Panel C: Association of the risk (insulin-raising) allele of the lead single nucleotide polymorphism in or near each of the putative effector genes with (a) the expression of the corresponding gene in subcutaneous adipocytes in the EUROBATS project (top graph in the panel); (b) hip circumference in a meta-analysis of GIANT and UK Biobank (mid graph); and (c) type 2 diabetes in a meta-analysis of InterAct, DIAGRAM and UK Biobank (bottom graph). Full circles represent the –log10(p-value) for the association of the insulin-raising allele multiplied by the direction of the beta coefficient (i.e. a “directional” –log10(p)). For graphic display purposes, the –log10(p-value) for the association with type 2 diabetes of the rs2943645-T allele near IRS1 is represented as 10 instead of 16.9.

References

    1. Samuel VT, Shulman GI. The pathogenesis of insulin resistance: integrating signaling pathways and substrate flux. The Journal of clinical investigation. 2016;126:12–22. doi: 10.1172/JCI77812.
    1. Ginsberg HN. Insulin resistance and cardiovascular disease. The Journal of clinical investigation. 2000;106:453–458. doi: 10.1172/JCI10762.
    1. Shulman GI. Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic disease. The New England journal of medicine. 2014;371:1131–1141. doi: 10.1056/NEJMra1011035.
    1. Lillioja S, Bogardus C. Obesity and insulin resistance: lessons learned from the Pima Indians. Diabetes/metabolism reviews. 1988;4:517–540.
    1. Perry RJ, Samuel VT, Petersen KF, Shulman GI. The role of hepatic lipids in hepatic insulin resistance and type 2 diabetes. Nature. 2014;510:84–91. doi: 10.1038/nature13478.
    1. Arner P. The adipocyte in insulin resistance: key molecules and the impact of the thiazolidinediones. Trends in endocrinology and metabolism: TEM. 2003;14:137–145.
    1. Friedman JM. Obesity in the new millennium. Nature. 2000;404:632–634. doi: 10.1038/35007504.
    1. Guilherme A, Virbasius JV, Puri V, Czech MP. Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Nature reviews. Molecular cell biology. 2008;9:367–377. doi: 10.1038/nrm2391.
    1. Friedman JM. A war on obesity, not the obese. Science. 2003;299:856–858. doi: 10.1126/science.1079856.
    1. Hardy OT, Czech MP, Corvera S. What causes the insulin resistance underlying obesity? Current opinion in endocrinology, diabetes, and obesity. 2012;19:81–87. doi: 10.1097/MED.0b013e3283514e13.
    1. Stefan N, Haring HU, Hu FB, Schulze MB. Metabolically healthy obesity: epidemiology, mechanisms, and clinical implications. Lancet Diabetes Endocrinol. 2013;1:152–162. doi: 10.1016/S2213-8587(13)70062-7.
    1. Robbins AL, Savage DB. The genetics of lipid storage and human lipodystrophies. Trends in molecular medicine. 2015;21:433–438. doi: 10.1016/j.molmed.2015.04.004.
    1. Semple RK, Savage DB, Cochran EK, Gorden P, O'Rahilly S. Genetic syndromes of severe insulin resistance. Endocrine reviews. 2011;32:498–514. doi: 10.1210/er.2010-0020.
    1. Samuel VT, Petersen KF, Shulman GI. Lipid-induced insulin resistance: unravelling the mechanism. Lancet. 2010;375:2267–2277. doi: 10.1016/S0140-6736(10)60408-4.
    1. Danforth E., Jr Failure of adipocyte differentiation causes type II diabetes mellitus? Nature genetics. 2000;26:13. doi: 10.1038/79111.
    1. Unger RH. Lipid overload and overflow: metabolic trauma and the metabolic syndrome. Trends in endocrinology and metabolism: TEM. 2003;14:398–403.
    1. Virtue S, Vidal-Puig A. Adipose tissue expandability, lipotoxicity and the Metabolic Syndrome--an allostatic perspective. Biochimica et biophysica acta. 2010;1801:338–349. doi: 10.1016/j.bbalip.2009.12.006.
    1. Shulman GI. Cellular mechanisms of insulin resistance. The Journal of clinical investigation. 2000;106:171–176. doi: 10.1172/JCI10583.
    1. Karpe F, Pinnick KE. Biology of upper-body and lower-body adipose tissue--link to whole-body phenotypes. Nature reviews. Endocrinology. 2015;11:90–100. doi: 10.1038/nrendo.2014.185.
    1. Robbins DC, et al. The effect of diet on thermogenesis in acquired lipodystrophy. Metabolism: clinical and experimental. 1979;28:908–916.
    1. Shimomura I, Hammer RE, Ikemoto S, Brown MS, Goldstein JL. Leptin reverses insulin resistance and diabetes mellitus in mice with congenital lipodystrophy. Nature. 1999;401:73–76. doi: 10.1038/43448.
    1. Oral EA, et al. Leptin-replacement therapy for lipodystrophy. The New England journal of medicine. 2002;346:570–578. doi: 10.1056/NEJMoa012437.
    1. Gavrilova O, et al. Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice. The Journal of clinical investigation. 2000;105:271–278. doi: 10.1172/JCI7901.
    1. Kim JY, et al. Obesity-associated improvements in metabolic profile through expansion of adipose tissue. The Journal of clinical investigation. 2007;117:2621–2637. doi: 10.1172/JCI31021.
    1. Gray SL, et al. Leptin deficiency unmasks the deleterious effects of impaired peroxisome proliferator-activated receptor gamma function (P465L PPARgamma) in mice. Diabetes. 2006;55:2669–2677. doi: 10.2337/db06-0389.
    1. Medina-Gomez G, et al. PPAR gamma 2 prevents lipotoxicity by controlling adipose tissue expandability and peripheral lipid metabolism. PLoS genetics. 2007;3:e64. doi: 10.1371/journal.pgen.0030064.
    1. Knowles JW, et al. Identification and validation of N-acetyltransferase 2 as an insulin sensitivity gene. The Journal of clinical investigation. 2016;126:403. doi: 10.1172/JCI85921.
    1. Scott RA, et al. Large-scale association analyses identify new loci influencing glycemic traits and provide insight into the underlying biological pathways. Nature genetics. 2012;44:991–1005. doi: 10.1038/ng.2385.
    1. Manning AK, et al. A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance. Nature genetics. 2012;44:659–669. doi: 10.1038/ng.2274.
    1. Salazar MR, et al. Comparison of the abilities of the plasma triglyceride/high-density lipoprotein cholesterol ratio and the metabolic syndrome to identify insulin resistance. Diabetes & vascular disease research. 2013;10:346–352. doi: 10.1177/1479164113479809.
    1. Scott RA, et al. Common genetic variants highlight the role of insulin resistance and body fat distribution in type 2 diabetes, independent of obesity. Diabetes. 2014;63:4378–4387. doi: 10.2337/db14-0319.
    1. Global Lipids Genetics Consortium. Discovery and refinement of loci associated with lipid levels. Nature genetics. 2013;45:1274–1283. doi: 10.1038/ng.2797.
    1. Prokopenko I, et al. A central role for GRB10 in regulation of islet function in man. PLoS genetics. 2014;10:e1004235. doi: 10.1371/journal.pgen.1004235.
    1. Morris AP, et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nature genetics. 2012;44:981–990. doi: 10.1038/ng.2383.
    1. Nikpay M, et al. A comprehensive 1,000 Genomes-based genome-wide association meta-analysis of coronary artery disease. Nature genetics. 2015;47:1121–1130. doi: 10.1038/ng.3396.
    1. Shungin D, et al. New genetic loci link adipose and insulin biology to body fat distribution. Nature. 2015;518:187–196. doi: 10.1038/nature14132.
    1. Kozaki K, et al. Mutational analysis of human lipoprotein lipase by carboxy-terminal truncation. Journal of lipid research. 1993;34:1765–1772.
    1. Myocardial Infarction Genetics and CARDIoGRAM Exome Consortia Investigators. Coding Variation in ANGPTL4, LPL, and SVEP1 and the Risk of Coronary Disease. The New England journal of medicine. 2016 doi: 10.1056/NEJMoa1507652.
    1. Mailly F, et al. A common variant in the gene for lipoprotein lipase (Asp9-->Asn). Functional implications and prevalence in normal and hyperlipidemic subjects. Arteriosclerosis, thrombosis, and vascular biology. 1995;15:468–478.
    1. Avila M, et al. Clinical reappraisal of SHORT syndrome with PIK3R1 mutations: towards recommendation for molecular testing and management. Clinical genetics. 2015 doi: 10.1111/cge.12688.
    1. Thauvin-Robinet C, et al. PIK3R1 mutations cause syndromic insulin resistance with lipoatrophy. American journal of human genetics. 2013;93:141–149. doi: 10.1016/j.ajhg.2013.05.019.
    1. Chudasama KK, et al. SHORT syndrome with partial lipodystrophy due to impaired phosphatidylinositol 3 kinase signaling. American journal of human genetics. 2013;93:150–157. doi: 10.1016/j.ajhg.2013.05.023.
    1. Dyment DA, et al. Mutations in PIK3R1 cause SHORT syndrome. American journal of human genetics. 2013;93:158–166. doi: 10.1016/j.ajhg.2013.06.005.
    1. Pers TH, et al. Biological interpretation of genome-wide association studies using predicted gene functions. Nature communications. 2015;6:5890. doi: 10.1038/ncomms6890.
    1. Lane JM, Doyle JR, Fortin JP, Kopin AS, Ordovas JM. Development of an OP9 derived cell line as a robust model to rapidly study adipocyte differentiation. PloS one. 2014;9:e112123. doi: 10.1371/journal.pone.0112123.
    1. Yaghootkar H, et al. Genetic evidence for a normal-weight "metabolically obese" phenotype linking insulin resistance, hypertension, coronary artery disease, and type 2 diabetes. Diabetes. 2014;63:4369–4377. doi: 10.2337/db14-0318.
    1. Lu Y, et al. New loci for body fat percentage reveal link between adiposity and cardiometabolic disease risk. Nature communications. 2016;7:10495. doi: 10.1038/ncomms10495.
    1. Kilpelainen TO, et al. Genetic variation near IRS1 associates with reduced adiposity and an impaired metabolic profile. Nature genetics. 2011;43:753–760. doi: 10.1038/ng.866.
    1. Yaghootkar H, et al. Genetic evidence for a link between favorable adiposity and lower risk of type 2 diabetes, hypertension and heart disease. Diabetes. 2016 doi: 10.2337/db15-1671.
    1. Biggs ML, et al. Association between adiposity in midlife and older age and risk of diabetes in older adults. JAMA : the journal of the American Medical Association. 2010;303:2504–2512. doi: 10.1001/jama.2010.843.
    1. Pischon T, et al. General and abdominal adiposity and risk of death in Europe. The New England journal of medicine. 2008;359:2105–2120. doi: 10.1056/NEJMoa0801891.
    1. Vague J. The degree of masculine differentiation of obesities: a factor determining predisposition to diabetes, atherosclerosis, gout, and uric calculous disease. The American journal of clinical nutrition. 1956;4:20–34.
    1. Smith U. Abdominal obesity: a marker of ectopic fat accumulation. The Journal of clinical investigation. 2015;125:1790–1792. doi: 10.1172/JCI81507.
    1. Despres JP, Lemieux I. Abdominal obesity and metabolic syndrome. Nature. 2006;444:881–887. doi: 10.1038/nature05488.
    1. Dahlman I, et al. Numerous Genes in Loci Associated With Body Fat Distribution Are Linked to Adipose Function. Diabetes. 2016;65:433–437. doi: 10.2337/db15-0828.
    1. Ryden M, Andersson DP, Bergstrom IB, Arner P. Adipose tissue and metabolic alterations: regional differences in fat cell size and number matter, but differently: a cross-sectional study. The Journal of clinical endocrinology and metabolism. 2014;99:E1870–1876. doi: 10.1210/jc.2014-1526.
    1. Pinnick KE, et al. Distinct developmental profile of lower-body adipose tissue defines resistance against obesity-associated metabolic complications. Diabetes. 2014;63:3785–3797. doi: 10.2337/db14-0385.
    1. Baughman BM, Pattenden SG, Norris JL, James LI, Frye SV. The L3MBTL3 Methyl-Lysine Reader Domain Functions As a Dimer. ACS chemical biology. 2016;11:722–728. doi: 10.1021/acschembio.5b00632.
    1. Randall JC, et al. Sex-stratified genome-wide association studies including 270,000 individuals show sexual dimorphism in genetic loci for anthropometric traits. PLoS genetics. 2013;9:e1003500. doi: 10.1371/journal.pgen.1003500.
    1. Paternoster L, et al. Adult height variants affect birth length and growth rate in children. Human molecular genetics. 2011;20:4069–4075. doi: 10.1093/hmg/ddr309.
    1. Wood AR, et al. Defining the role of common variation in the genomic and biological architecture of adult human height. Nature genetics. 2014;46:1173–1186. doi: 10.1038/ng.3097.
    1. Gupta GD, et al. A Dynamic Protein Interaction Landscape of the Human Centrosome-Cilium Interface. Cell. 2015;163:1484–1499. doi: 10.1016/j.cell.2015.10.065.
    1. Singaraja RR, et al. Identification of four novel genes contributing to familial elevated plasma HDL cholesterol in humans. Journal of lipid research. 2014;55:1693–1701. doi: 10.1194/jlr.M048710.
    1. Chasman DI, et al. Forty-three loci associated with plasma lipoprotein size, concentration, and cholesterol content in genome-wide analysis. PLoS genetics. 2009;5:e1000730. doi: 10.1371/journal.pgen.1000730.
    1. DeFronzo RA, et al. Pioglitazone for diabetes prevention in impaired glucose tolerance. The New England journal of medicine. 2011;364:1104–1115. doi: 10.1056/NEJMoa1010949.
    1. DREAM (Diabetes REduction Assessment with ramipril and rosiglitazone Medication) Trial Investigators. Effect of rosiglitazone on the frequency of diabetes in patients with impaired glucose tolerance or impaired fasting glucose: a randomised controlled trial. Lancet. 2006;368:1096–1105. doi: 10.1016/S0140-6736(06)69420-8.
    1. Lincoff AM, Wolski K, Nicholls SJ, Nissen SE. Pioglitazone and risk of cardiovascular events in patients with type 2 diabetes mellitus: a meta-analysis of randomized trials. JAMA : the journal of the American Medical Association. 2007;298:1180–1188. doi: 10.1001/jama.298.10.1180.
    1. Kernan WN, et al. Pioglitazone after Ischemic Stroke or Transient Ischemic Attack. The New England journal of medicine. 2016 doi: 10.1056/NEJMoa1506930.
    1. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. The New England journal of medicine. 2007;356:2457–2471. doi: 10.1056/NEJMoa072761.
    1. Food and Drug Administration. Guidance for Industry. Diabetes Mellitus — Evaluating Cardiovascular Risk in New Antidiabetic Therapies to Treat Type 2 Diabetes. 2008 URL: .
    1. Swerdlow DI, et al. HMG-coenzyme A reductase inhibition, type 2 diabetes, and bodyweight: evidence from genetic analysis and randomised trials. Lancet. 2015;385:351–361. doi: 10.1016/S0140-6736(14)61183-1.
    1. Dewey FE, et al. Inactivating Variants in ANGPTL4 and Risk of Coronary Artery Disease. The New England journal of medicine. 2016 doi: 10.1056/NEJMoa1510926.
    1. Do R, et al. Exome sequencing identifies rare LDLR and APOA5 alleles conferring risk for myocardial infarction. Nature. 2015;518:102–106. doi: 10.1038/nature13917.
    1. Jorgensen AB, Frikke-Schmidt R, Nordestgaard BG, Tybjaerg-Hansen A. Loss:of-function mutations in APOC3 and risk of ischemic vascular disease. The New England journal of medicine. 2014;371:32–41. doi: 10.1056/NEJMoa1308027.
    1. TG and HDL Working Group of the Exome Sequencing Project, National Heart, Lung, and Blood Institute. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. The New England journal of medicine. 2014;371:22–31. doi: 10.1056/NEJMoa1307095.
    1. Myocardial Infarction Genetics Consortium Investigators. Inactivating mutations in NPC1L1 and protection from coronary heart disease. The New England journal of medicine. 2014;371:2072–2082. doi: 10.1056/NEJMoa1405386.
    1. Moltke I, et al. A common Greenlandic TBC1D4 variant confers muscle insulin resistance and type 2 diabetes. Nature. 2014;512:190–193. doi: 10.1038/nature13425.
    1. Day N, et al. EPIC-Norfolk: study design and characteristics of the cohort. European Prospective Investigation of Cancer. British journal of cancer. 1999;80(Suppl 1):95–103.
    1. Riboli E, et al. European Prospective Investigation into Cancer and Nutrition (EPIC): study populations and data collection. Public health nutrition. 2002;5:1113–1124. doi: 10.1079/PHN2002394.
    1. InterAct Consortium et al. Design and cohort description of the InterAct Project: an examination of the interaction of genetic and lifestyle factors on the incidence of type 2 diabetes in the EPIC Study. Diabetologia. 2011;54:2272–2282. doi: 10.1007/s00125-011-2182-9.
    1. Collins R. What makes UK Biobank special? Lancet. 2012;379:1173–1174. doi: 10.1016/S0140-6736(12)60404-8.
    1. Lyssenko V, et al. Mechanisms by which common variants in the TCF7L2 gene increase risk of type 2 diabetes. The Journal of clinical investigation. 2007;117:2155–2163. doi: 10.1172/JCI30706.
    1. Aschard H, Vilhjalmsson BJ, Joshi AD, Price AL, Kraft P. Adjusting for heritable covariates can bias effect estimates in genome-wide association studies. American journal of human genetics. 2015;96:329–339. doi: 10.1016/j.ajhg.2014.12.021.
    1. Day FR, Loh PR, Scott RA, Ong KK, Perry JR. A Robust Example of Collider Bias in a Genetic Association Study. American journal of human genetics. 2016;98:392–393. doi: 10.1016/j.ajhg.2015.12.019.
    1. Burgess S, Butterworth A, Thompson SG. Mendelian randomization analysis with multiple genetic variants using summarized data. Genetic epidemiology. 2013;37:658–665. doi: 10.1002/gepi.21758.
    1. Lawrence RD. Types of human diabetes. British medical journal. 1951;1:373–375.
    1. Herbst KL, et al. Kobberling type of familial partial lipodystrophy: an underrecognized syndrome. Diabetes care. 2003;26:1819–1824.
    1. Payne F, et al. Hypomorphism in human NSMCE2 linked to primordial dwarfism and insulin resistance. The Journal of clinical investigation. 2014;124:4028–4038. doi: 10.1172/JCI73264.
    1. UK10K Consortium. The UK10K project identifies rare variants in health and disease. Nature. 2015;526:82–90. doi: 10.1038/nature14962.
    1. Purcell S, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. American journal of human genetics. 2007;81:559–575. doi: 10.1086/519795.
    1. Delaneau O, Marchini J, Zagury JF. A linear complexity phasing method for thousands of genomes. Nature methods. 2012;9:179–181. doi: 10.1038/nmeth.1785.
    1. Marchini J, Howie B, Myers S, McVean G, Donnelly P. A new multipoint method for genome-wide association studies by imputation of genotypes. Nature genetics. 2007;39:906–913. doi: 10.1038/ng2088.
    1. Pruim RJ, et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics. 2010;26:2336–2337. doi: 10.1093/bioinformatics/btq419.
    1. Ward LD, Kellis M. HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic acids research. 2012;40:D930–934. doi: 10.1093/nar/gkr917.
    1. Johnson AD, et al. SNAP: a web-based tool for identification and annotation of proxy SNPs using HapMap. Bioinformatics. 2008;24:2938–2939. doi: 10.1093/bioinformatics/btn564.
    1. Welter D, et al. The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic acids research. 2014;42:D1001–1006. doi: 10.1093/nar/gkt1229.
    1. Zhang X, et al. Synthesis of 53 tissue and cell line expression QTL datasets reveals master eQTLs. BMC genomics. 2014;15:532. doi: 10.1186/1471-2164-15-532.
    1. Buil A, et al. Gene-gene and gene-environment interactions detected by transcriptome sequence analysis in twins. Nature genetics. 2015;47:88–91. doi: 10.1038/ng.3162.
    1. GTEx Consortium. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science. 2015;348:648–660. doi: 10.1126/science.1262110.
    1. Nica AC, et al. Candidate causal regulatory effects by integration of expression QTLs with complex trait genetic associations. PLoS genetics. 2010;6:e1000895. doi: 10.1371/journal.pgen.1000895.

Source: PubMed

3
Iratkozz fel