Metabolomics reveals amino acids contribute to variation in response to simvastatin treatment

Miles Trupp, Hongjie Zhu, William R Wikoff, Rebecca A Baillie, Zhao-Bang Zeng, Peter D Karp, Oliver Fiehn, Ronald M Krauss, Rima Kaddurah-Daouk, Miles Trupp, Hongjie Zhu, William R Wikoff, Rebecca A Baillie, Zhao-Bang Zeng, Peter D Karp, Oliver Fiehn, Ronald M Krauss, Rima Kaddurah-Daouk

Abstract

Statins are widely prescribed for reducing LDL-cholesterol (C) and risk for cardiovascular disease (CVD), but there is considerable variation in therapeutic response. We used a gas chromatography-time-of-flight mass-spectrometry-based metabolomics platform to evaluate global effects of simvastatin on intermediary metabolism. Analyses were conducted in 148 participants in the Cholesterol and Pharmacogenetics study who were profiled pre and six weeks post treatment with 40 mg/day simvastatin: 100 randomly selected from the full range of the LDL-C response distribution and 24 each from the top and bottom 10% of this distribution ("good" and "poor" responders, respectively). The metabolic signature of drug exposure in the full range of responders included essential amino acids, lauric acid (p<0.0055, q<0.055), and alpha-tocopherol (p<0.0003, q<0.017). Using the HumanCyc database and pathway enrichment analysis, we observed that the metabolites of drug exposure were enriched for the pathway class amino acid degradation (p<0.0032). Metabolites whose change correlated with LDL-C lowering response to simvastatin in the full range responders included cystine, urea cycle intermediates, and the dibasic amino acids ornithine, citrulline and lysine. These dibasic amino acids share plasma membrane transporters with arginine, the rate-limiting substrate for nitric oxide synthase (NOS), a critical mediator of cardiovascular health. Baseline metabolic profiles of the good and poor responders were analyzed by orthogonal partial least square discriminant analysis so as to determine the metabolites that best separated the two response groups and could be predictive of LDL-C response. Among these were xanthine, 2-hydroxyvaleric acid, succinic acid, stearic acid, and fructose. Together, the findings from this study indicate that clusters of metabolites involved in multiple pathways not directly connected with cholesterol metabolism may play a role in modulating the response to simvastatin treatment.

Trial registration: ClinicalTrials.gov NCT00451828.

Conflict of interest statement

Competing Interests: The authors have read the journal's policy and have the following conflicts. RKD is equity holder in Metabolon in the metabolomics domain, and also an inventor on patents in the metabolomics field. There are no specific patents filed related to the finding reported in this manuscript. RKD, RMK and RAB are inventors on a patent application on statin effects on metabolism. RAB is an employee of Rosa and Co LLC. MT is an employee of SRI International. In none of these cases do these affiliations alter the authors' adherence to all the PLoS ONE policies on sharing data and materials. HZ, OF, PK, WW and ZBZ have no competing interests.

Figures

Figure 1. Correlation matrix of metabolites altered…
Figure 1. Correlation matrix of metabolites altered by simvastatin in full range participants.
Correlations among metabolites in Table 1 were obtained by deriving a Spearman’s correlation coefficient between each pair of metabolites. The color scheme corresponds to correlation strength as shown by the color bar. Red: Better response, more reduction of the metabolite. Blue: Better response, less reduction or increase of the metabolite. The metabolites have been rescaled (divided by the largest absolute value of them) to be clearer on the map. Abbreviations: NIST, National Institute of Standards and Technology.
Figure 2. Correlation matrix illustrating two clusters…
Figure 2. Correlation matrix illustrating two clusters of compounds correlated with simvastatin response in full range participants.
The two clusters were identified in a clustering analysis for the change of all metabolites (results not shown) according to their pairwise correlations using the MMC algorithm [14]). Correlations of metabolites to drug response in LDLC were given in the first row and column, and are rescaled (divided by the largest absolute value of them) to be clearer in the map. The color scheme corresponds to correlation strength as shown by the color bar. Red: Better response, more reduction of the metabolite. Blue: Better response, less reduction or increase of the metabolite. Abbreviations: LDLC, Low-Density Lipoprotein Cholesterol; NIST, National Institute of Standards and Technology.
Figure 3. OPLSDA of baseline metabolites classifies…
Figure 3. OPLSDA of baseline metabolites classifies good and poor responders.
(A) Orthogonal partial least square discriminant analysis was used to classify good and poor responders based on log-transformed baseline concentration of metabolites (R2 = 0.87, Q2 = 0.31). Good responders are shown in black and poor responders in red. Baseline metabolites were log-transformed and normalized (described in methods). Performance evaluation by 7-fold cross validation yielded the following statistics: prediction accuracy: 74%; sensitivity: 70%; specificity: 79% (not shown). (B) ROC curve of true positive rate (x-axis) versus false positive rate (y-axis) yields an area under the curve (AUC) of 0.84. (C) Baseline metabolites ranked by importance in classifying good and poor responders in the OPLS model. *indicates a partially identified compound: pentonic acid is an aldonic acid with five carbons and hexaric acid is an aldonic acid with six carbons. Abbreviations: VIP, variable importance score; cvSE, standard error derived from cross validation.

References

    1. Grundy SM, Cleeman JI, Merz CN, Brewer HB, Jr, Clark LT, et al. Implications of recent clinical trials for the National Cholesterol Education Program Adult Treatment Panel III guidelines. Circulation. 2004;110:227–239.
    1. Jasinska M, Owczarek J, Orszulak-Michalak D. Statins: a new insight into their mechanisms of action and consequent pleiotropic effects. Pharmacol Rep. 2007;59:483–499.
    1. Ridker PM. The JUPITER trial: results, controversies, and implications for prevention. Circulation Cardiovascular quality and outcomes. 2009;2:279–285.
    1. Mangravite LM, Wilke RA, Zhang J, Krauss RM. Pharmacogenomics of statin response. Curr Opin Mol Ther. 2008;10:555–561.
    1. Vaquero MP, Sanchez Muniz FJ, Jimenez Redondo S, Prats Olivan P, Higueras FJ, et al. Major diet-drug interactions affecting the kinetic characteristics and hypolipidaemic properties of statins. Nutr Hosp. 2010;25:193–206.
    1. Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Jr, et al. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008;359:2195–2207.
    1. Bai JP. Ongoing challenges in drug interaction safety: from exposure to pharmacogenomics. Drug Metab Pharmacokinet. 2010;25:62–71.
    1. Clayton TA, Baker D, Lindon JC, Everett JR, Nicholson JK. Pharmacometabonomic identification of a significant host-microbiome metabolic interaction affecting human drug metabolism. Proc Natl Acad Sci U S A. 2009;106:14728–14733.
    1. Kaddurah-Daouk R, Baillie RA, Zhu H, Zeng ZB, Wiest MM, et al. Enteric microbiome metabolites correlate with response to simvastatin treatment. PLoS ONE. 2011;6:e25482.
    1. Kaddurah-Daouk R, Kristal BS, Weinshilboum RM. Metabolomics: a global biochemical approach to drug response and disease. Annu Rev Pharmacol Toxicol. 2008;48:653–683.
    1. Kaddurah-Daouk R, Krishnan KR. Metabolomics: a global biochemical approach to the study of central nervous system diseases. Neuropsychopharmacology. 2009;34:173–186.
    1. Han X, Rozen S, Boyle SH, Hellegers C, Cheng H, et al. Metabolomics in early Alzheimer's disease: identification of altered plasma sphingolipidome using shotgun lipidomics. PLoS ONE. 2011;6:e21643.
    1. Yao JK, Dougherty GG, Jr, Reddy RD, Keshavan MS, Montrose DM, et al. Altered interactions of tryptophan metabolites in first-episode neuroleptic-naive patients with schizophrenia. Mol Psychiatry. 2010;15:938–953.
    1. Yao JK, Dougherty GG, Jr, Reddy RD, Keshavan MS, Montrose DM, et al. Homeostatic imbalance of purine catabolism in first-episode neuroleptic-naive patients with schizophrenia. PLoS ONE. 2010;5:e9508.
    1. Nicholson JK, Wilson ID, Lindon JC. Pharmacometabonomics as an effector for personalized medicine. Pharmacogenomics. 2011;12:103–111.
    1. Wang TJ, Larson MG, Vasan RS, Cheng S, Rhee EP, et al. Metabolite profiles and the risk of developing diabetes. Nat Med. 2011;17:448–453.
    1. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472:57–63.
    1. Ji Y, Hebbring S, Zhu H, Jenkins GD, Biernacka J, et al. Glycine and a glycine dehydrogenase (GLDC) SNP as citalopram/escitalopram response biomarkers in depression: pharmacometabolomics-informed pharmacogenomics. Clin Pharmacol Ther. 2011;89:97–104.
    1. Kaddurah-Daouk R, Baillie RA, Zhu H, Zeng ZB, Wiest MM, et al. Lipidomic analysis of variation in response to simvastatin in the Cholesterol and Pharmacogenetics Study. Metabolomics. 2010;6:191–201.
    1. Kaddurah-Daouk R, Boyle SH, Matson W, Sharma S, Matson S, et al. Pretreatment metabotype as a predictor of response to sertraline or placebo in depressed outpatients: a proof of concept. Transl Psychiatry. 2011;1:e26.
    1. Simon JA, Lin F, Hulley SB, Blanche PJ, Waters D, et al. Phenotypic predictors of response to simvastatin therapy among African-Americans and Caucasians: the Cholesterol and Pharmacogenetics (CAP) Study. Am J Cardiol. 2006;97:843–850.
    1. Pal S, Thomson AM, Bottema CD, Roach PD. Alpha-tocopherol modulates the low density lipoprotein receptor of human HepG2 cells. Nutr J. 2003;2:3.
    1. Rode S, Rubic T, Lorenz RL. alpha-Tocopherol disturbs macrophage LXRalpha regulation of ABCA1/G1 and cholesterol handling. Biochem Biophys Res Commun. 2008;369:868–872.
    1. Devaraj S, Hugou I, Jialal I. Alpha-tocopherol decreases CD36 expression in human monocyte-derived macrophages. J Lipid Res. 2001;42:521–527.
    1. Ricciarelli R, Zingg JM, Azzi A. Vitamin E reduces the uptake of oxidized LDL by inhibiting CD36 scavenger receptor expression in cultured aortic smooth muscle cells. Circulation. 2000;102:82–87.
    1. Human JA, Ubbink JB, Jerling JJ, Delport R, Vermaak WJ, et al. The effect of Simvastatin on the plasma antioxidant concentrations in patients with hypercholesterolaemia. Clin Chim Acta. 1997;263:67–77.
    1. Vasankari T, Ahotupa M, Viikari J, Nuotio I, Strandberg T, et al. Effect of 12-month statin therapy on antioxidant potential of LDL and serum antioxidant vitamin concentrations. Ann Med. 2004;36:618–622.
    1. Neunteufl T, Kostner K, Katzenschlager R, Zehetgruber M, Maurer G, et al. Additional benefit of vitamin E supplementation to simvastatin therapy on vasoreactivity of the brachial artery of hypercholesterolemic men. J Am Coll Cardiol. 1998;32:711–716.
    1. Ryden M, Leanderson P, Kastbom KO, Jonasson L. Effects of simvastatin on carotenoid status in plasma. Nutr Metab Cardiovasc Dis. 2010.
    1. Piorunska-Stolzmann M, Piorunska-Mikolajczak A, Mikolajczyk Z. Effect of simvastatin on trioleylglycerol hydrolysis and transacylation with cholesterol in serum of outpatients with coronary heart disease. Drugs Exp Clin Res. 2003;29:37–43.
    1. Baigent C, Landray M, Leaper C, Altmann P, Armitage J, et al. First United Kingdom Heart and Renal Protection (UK-HARP-I) study: biochemical efficacy and safety of simvastatin and safety of low-dose aspirin in chronic kidney disease. Am J Kidney Dis. 2005;45:473–484.
    1. Huskey J, Lindenfeld J, Cook T, Targher G, Kendrick J, et al. Effect of simvastatin on kidney function loss in patients with coronary heart disease: findings from the Scandinavian Simvastatin Survival Study (4S). Atherosclerosis. 2009;205:202–206.
    1. Namli S, Oflaz H, Turgut F, Alisir S, Tufan F, et al. Improvement of endothelial dysfunction with simvastatin in patients with autosomal dominant polycystic kidney disease. Ren Fail. 2007;29:55–59.
    1. Keshishian H, Addona T, Burgess M, Mani DR, Shi X, et al. Quantification of cardiovascular biomarkers in patient plasma by targeted mass spectrometry and stable isotope dilution. Mol Cell Proteomics. 2009;8:2339–2349.
    1. Laferrere B, Reilly D, Arias S, Swerdlow N, Gorroochurn P, et al. Differential metabolic impact of gastric bypass surgery versus dietary intervention in obese diabetic subjects despite identical weight loss. Sci Transl Med. 2011;3:80re82.
    1. Lee BE, Toledo AH, Anaya-Prado R, Roach RR, Toledo-Pereyra LH. Allopurinol, xanthine oxidase, and cardiac ischemia. J Investig Med. 2009;57:902–909.
    1. Pacher P, Nivorozhkin A, Szabo C. Therapeutic effects of xanthine oxidase inhibitors: renaissance half a century after the discovery of allopurinol. Pharmacol Rev. 2006;58:87–114.
    1. Dawson J, Walters M. Uric acid and xanthine oxidase: future therapeutic targets in the prevention of cardiovascular disease? Br J Clin Pharmacol. 2006.
    1. Schwartz IF, Grupper A, Chernichovski T, Hillel O, Engel A, et al. Hyperuricemia attenuates aortic nitric oxide generation, through inhibition of arginine transport, in rats. J Vasc Res. 2011;48:252–260.
    1. Aura AM, Mattila I, Hyotylainen T, Gopalacharyulu P, Bounsaythip C, et al. Drug metabolome of the simvastatin formed by human intestinal microbiota in vitro. Mol Biosyst. 2011;7:437–446.
    1. Fiehn O, Wohlgemuth G, Scholz M, Kind T, Lee do Y, et al. Quality control for plant metabolomics: reporting MSI-compliant studies. Plant J. 2008;53:691–704.
    1. Fiehn O, Wohlgemuth G, Scholz M. Setup and annotation of metabolomic experiments by integrating biological and mass spectrometric metadata. Proc Lect Notes Bioinformatics. 2005;3615:224–239.
    1. Scholz M, Fiehn O. SetupX–a public study design database for metabolomic projects. Pac Symp Biocomput. 2007. pp. 169–180.
    1. Storey JD, Tibshirani R. Statistical significance for genomewide studies. Proceedings of the National Academy of Sciences of the United States of America. 2003;100:9440–9445.
    1. Stone EA, Ayroles JF. Modulated modularity clustering as an exploratory tool for functional genomic inference. PLoS Genet. 2009;5:e1000479.
    1. Trygg J, Wold S. Orthogonal projections to latent structures (O-PLS). Journal of Chemometrics. 2002;16:119–128.

Source: PubMed

3
Abonnieren