Large-scale metabolomic profiling identifies novel biomarkers for incident coronary heart disease

Andrea Ganna, Samira Salihovic, Johan Sundström, Corey D Broeckling, Asa K Hedman, Patrik K E Magnusson, Nancy L Pedersen, Anders Larsson, Agneta Siegbahn, Mihkel Zilmer, Jessica Prenni, Johan Arnlöv, Lars Lind, Tove Fall, Erik Ingelsson, Andrea Ganna, Samira Salihovic, Johan Sundström, Corey D Broeckling, Asa K Hedman, Patrik K E Magnusson, Nancy L Pedersen, Anders Larsson, Agneta Siegbahn, Mihkel Zilmer, Jessica Prenni, Johan Arnlöv, Lars Lind, Tove Fall, Erik Ingelsson

Abstract

Analyses of circulating metabolites in large prospective epidemiological studies could lead to improved prediction and better biological understanding of coronary heart disease (CHD). We performed a mass spectrometry-based non-targeted metabolomics study for association with incident CHD events in 1,028 individuals (131 events; 10 y. median follow-up) with validation in 1,670 individuals (282 events; 3.9 y. median follow-up). Four metabolites were replicated and independent of main cardiovascular risk factors [lysophosphatidylcholine 18∶1 (hazard ratio [HR] per standard deviation [SD] increment = 0.77, P-value<0.001), lysophosphatidylcholine 18∶2 (HR = 0.81, P-value<0.001), monoglyceride 18∶2 (MG 18∶2; HR = 1.18, P-value = 0.011) and sphingomyelin 28∶1 (HR = 0.85, P-value = 0.015)]. Together they contributed to moderate improvements in discrimination and re-classification in addition to traditional risk factors (C-statistic: 0.76 vs. 0.75; NRI: 9.2%). MG 18∶2 was associated with CHD independently of triglycerides. Lysophosphatidylcholines were negatively associated with body mass index, C-reactive protein and with less evidence of subclinical cardiovascular disease in additional 970 participants; a reverse pattern was observed for MG 18∶2. MG 18∶2 showed an enrichment (P-value = 0.002) of significant associations with CHD-associated SNPs (P-value = 1.2×10-7 for association with rs964184 in the ZNF259/APOA5 region) and a weak, but positive causal effect (odds ratio = 1.05 per SD increment in MG 18∶2, P-value = 0.05) on CHD, as suggested by Mendelian randomization analysis. In conclusion, we identified four lipid-related metabolites with evidence for clinical utility, as well as a causal role in CHD development.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Figure 1. Study flow chart.
Figure 1. Study flow chart.
Overview of the study design and analyses performed.
Figure 2. Association between four metabolites and…
Figure 2. Association between four metabolites and cardiovascular traits and genotypes.
Panel A: Association with main cardiovascular risk factors in three population-based studies. Panel B: Minus log10(P-value) for association with markers of inflammation, oxidative stress and subclinical CVD in PIVUS. Sex-adjusted analysis (upper panel) and adjusted by sex, systolic blood pressure, body mass index, current smoker, antihypertensive treatment, LDL-C, HDL-C, log-triglycerides and diabetes at baseline (lower panel). * indicates the alpha threshold after multiple-testing correction. Panel C: Minus log10(P-value) for association with 51 SNPs previously reported for association with CHD (44 SNPs) or selected from candidate pathways (7 SNPs).
Figure 3. Mendelian randomization analysis.
Figure 3. Mendelian randomization analysis.
A significant deviation from zero of the estimate of causal effect using all SNPs (solid red line) suggests a causal relationship between the metabolite and CHD.

References

    1. Suhre K, Shin SY, Petersen AK, Mohney RP, Meredith D, et al. (2011) Human metabolic individuality in biomedical and pharmaceutical research. Nature 477: 54–60.
    1. Magnusson M, Lewis GD, Ericson U, Orho-Melander M, Hedblad B, et al. (2013) A diabetes-predictive amino acid score and future cardiovascular disease. Eur Heart J 34: 1982–1989.
    1. Shah SH, Bain JR, Muehlbauer MJ, Stevens RD, Crosslin DR, et al. (2010) Association of a peripheral blood metabolic profile with coronary artery disease and risk of subsequent cardiovascular events. Circ Cardiovasc Genet 3: 207–214.
    1. Boyanovsky BB, Webb NR (2009) Biology of secretory phospholipase A2. Cardiovasc Drugs Ther 23: 61–72.
    1. Wilson PW, D'Agostino RB, Levy D, Belanger AM, Silbershatz H, et al. (1998) Prediction of coronary heart disease using risk factor categories. Circulation 97: 1837–1847.
    1. Lemaitre RN, Tanaka T, Tang W, Manichaikul A, Foy M, et al. (2011) Genetic loci associated with plasma phospholipid n-3 fatty acids: a meta-analysis of genome-wide association studies from the CHARGE Consortium. PLoS Genet 7: e1002193.
    1. Schunkert H, Konig IR, Kathiresan S, Reilly MP, Assimes TL, et al. (2011) Large-scale association analysis identifies 13 new susceptibility loci for coronary artery disease. Nat Genet 43: 333–338.
    1. Demirkan A, van Duijn CM, Ugocsai P, Isaacs A, Pramstaller PP, et al. (2012) Genome-wide association study identifies novel loci associated with circulating phospho- and sphingolipid concentrations. PLoS Genet 8: e1002490.
    1. CARDIoGRAMplusC4D Consortium (2013) Deloukas P, Kanoni S, Willenborg C, Farrall M, et al. (2013) Large-scale association analysis identifies new risk loci for coronary artery disease. Nat Genet 45: 25–33.
    1. Miller M, Stone NJ, Ballantyne C, Bittner V, Criqui MH, et al. (2011) Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation 123: 2292–2333.
    1. Do R, Willer CJ, Schmidt EM, Sengupta S, Gao C, et al. (2013) Common variants associated with plasma triglycerides and risk for coronary artery disease. Nat Genet 45: 1345–1352.
    1. Rozenberg O, Shih DM, Aviram M (2003) Human serum paraoxonase 1 decreases macrophage cholesterol biosynthesis: possible role for its phospholipase-A2-like activity and lysophosphatidylcholine formation. Arterioscler Thromb Vasc Biol 23: 461–467.
    1. Gauster M, Rechberger G, Sovic A, Horl G, Steyrer E, et al. (2005) Endothelial lipase releases saturated and unsaturated fatty acids of high density lipoprotein phosphatidylcholine. J Lipid Res 46: 1517–1525.
    1. Sekas G, Patton GM, Lincoln EC, Robins SJ (1985) Origin of plasma lysophosphatidylcholine: evidence for direct hepatic secretion in the rat. J Lab Clin Med 105: 190–194.
    1. Shamburek RD, Zech LA, Cooper PS, Vandenbroek JM, Schwartz CC (1996) Disappearance of two major phosphatidylcholines from plasma is predominantly via LCAT and hepatic lipase. Am J Physiol 271: E1073–1082.
    1. Schmitz G, Ruebsaamen K (2010) Metabolism and atherogenic disease association of lysophosphatidylcholine. Atherosclerosis 208: 10–18.
    1. Wang-Sattler R, Yu Z, Herder C, Messias AC, Floegel A, et al. (2012) Novel biomarkers for pre-diabetes identified by metabolomics. Mol Syst Biol 8: 615.
    1. Fernandez C, Sandin M, Sampaio JL, Almgren P, Narkiewicz K, et al. (2013) Plasma lipid composition and risk of developing cardiovascular disease. PLoS One 8: e71846.
    1. Stegemann C, Pechlaner R, Willeit P, Langley S, Mangino M, et al. (2014) Lipidomics Profiling and Risk of Cardiovascular Disease in the Prospective Population-Based Bruneck Study. Circulation
    1. Tzoulaki I, Ebbels TM, Valdes A, Elliott P, Ioannidis JP (2014) Design and analysis of metabolomics studies in epidemiologic research: a primer on -omic technologies. Am J Epidemiol 180: 129–139.
    1. Ganna A, Fall T, Lee W, Broeckling CD, Kumar J, et al. (2014) A workflow for UPLC-MS non-targeted metabolomic profiling in large human population-based studies. bioRxiv
    1. Ganna A, Lee D, Ingelsson E, Pawitan Y (2014) Rediscovery rate estimation for assessing the validation of significant findings in high-throughput studies. Brief Bioinform
    1. Magnusson PK, Almqvist C, Rahman I, Ganna A, Viktorin A, et al. (2013) The Swedish Twin Registry: establishment of a biobank and other recent developments. Twin Res Hum Genet 16: 317–329.
    1. Ganna A, Reilly M, de Faire U, Pedersen N, Magnusson P, et al. (2012) Risk prediction measures for case-cohort and nested case-control designs: an application to cardiovascular disease. Am J Epidemiol 175: 715–724.
    1. Byberg L, Siegbahn A, Berglund L, McKeigue P, Reneland R, et al. (1998) Plasminogen activator inhibitor-1 activity is independently related to both insulin sensitivity and serum triglycerides in 70-year-old men. Arterioscler Thromb Vasc Biol 18: 258–264.
    1. Lind L, Fors N, Hall J, Marttala K, Stenborg A (2005) A comparison of three different methods to evaluate endothelium-dependent vasodilation in the elderly: the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study. Arterioscler Thromb Vasc Biol 25: 2368–2375.
    1. Broeckling CD, Heuberger AL, Prenni JE (2013) Large scale non-targeted metabolomic profiling of serum by ultra performance liquid chromatography-mass spectrometry (UPLC-MS). J Vis Exp e50242.
    1. Lind L, Fors N, Hall J, Marttala K, Stenborg A (2006) A comparison of three different methods to determine arterial compliance in the elderly: the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study. J Hypertens 24: 1075–1082.
    1. Lind L, Siegbahn A, Hulthe J, Elmgren A (2008) C-reactive protein and e-selectin levels are related to vasodilation in resistance, but not conductance arteries in the elderly: the prospective investigation of the Vasculature in Uppsala Seniors (PIVUS) study. Atherosclerosis 199: 129–137.
    1. Lind L, Siegbahn A, Ingelsson E, Sundstrom J, Arnlov J (2011) A detailed cardiovascular characterization of obesity without the metabolic syndrome. Arterioscler Thromb Vasc Biol 31: e27–34.
    1. Smith CA, Want EJ, O'Maille G, Abagyan R, Siuzdak G (2006) XCMS: processing mass spectrometry data for metabolite profiling using nonlinear peak alignment, matching, and identification. Anal Chem 78: 779–787.
    1. Sumner LW, Amberg A, Barrett D, Beale MH, Beger R, et al. (2007) Proposed minimum reporting standards for chemical analysis Chemical Analysis Working Group (CAWG) Metabolomics Standards Initiative (MSI). Metabolomics 3: 211–221.
    1. Leening MJ, Vedder MM, Witteman JC, Pencina MJ, Steyerberg EW (2014) Net reclassification improvement: computation, interpretation, and controversies: a literature review and clinician's guide. Ann Intern Med 160: 122–131.

Source: PubMed

3
Sottoscrivi