Metabolomic fingerprint of heart failure with preserved ejection fraction

Beshay N Zordoky, Miranda M Sung, Justin Ezekowitz, Rupasri Mandal, Beomsoo Han, Trent C Bjorndahl, Souhaila Bouatra, Todd Anderson, Gavin Y Oudit, David S Wishart, Jason R B Dyck, Alberta HEART, Beshay N Zordoky, Miranda M Sung, Justin Ezekowitz, Rupasri Mandal, Beomsoo Han, Trent C Bjorndahl, Souhaila Bouatra, Todd Anderson, Gavin Y Oudit, David S Wishart, Jason R B Dyck, Alberta HEART

Abstract

Background: Heart failure (HF) with preserved ejection fraction (HFpEF) is increasingly recognized as an important clinical entity. Preclinical studies have shown differences in the pathophysiology between HFpEF and HF with reduced ejection fraction (HFrEF). Therefore, we hypothesized that a systematic metabolomic analysis would reveal a novel metabolomic fingerprint of HFpEF that will help understand its pathophysiology and assist in establishing new biomarkers for its diagnosis.

Methods and results: Ambulatory patients with clinical diagnosis of HFpEF (n = 24), HFrEF (n = 20), and age-matched non-HF controls (n = 38) were selected for metabolomic analysis as part of the Alberta HEART (Heart Failure Etiology and Analysis Research Team) project. 181 serum metabolites were quantified by LC-MS/MS and 1H-NMR spectroscopy. Compared to non-HF control, HFpEF patients demonstrated higher serum concentrations of acylcarnitines, carnitine, creatinine, betaine, and amino acids; and lower levels of phosphatidylcholines, lysophosphatidylcholines, and sphingomyelins. Medium and long-chain acylcarnitines and ketone bodies were higher in HFpEF than HFrEF patients. Using logistic regression, two panels of metabolites were identified that can separate HFpEF patients from both non-HF controls and HFrEF patients with area under the receiver operating characteristic (ROC) curves of 0.942 and 0.981, respectively.

Conclusions: The metabolomics approach employed in this study identified a unique metabolomic fingerprint of HFpEF that is distinct from that of HFrEF. This metabolomic fingerprint has been utilized to identify two novel panels of metabolites that can separate HFpEF patients from both non-HF controls and HFrEF patients.

Clinical trial registration: ClinicalTrials.gov NCT02052804.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Flow chart representing patient selection…
Fig 1. Flow chart representing patient selection in the metabolomics sub-study.
649 patients were enrolled in the Alberta HEART study as of March 31st, 2014. Only 82 subjects were included in this metabolomics sub-study. Among these 82 subjects, there were 24 patients with HFpEF, 20 patients with HFrEF, and 38 control subjects without heart failure.
Fig 2. Cardiac peptides and left ventricular…
Fig 2. Cardiac peptides and left ventricular ejection fraction (LVEF) in control and HF patients.
Ambulatory patients with clinical diagnosis of HFpEF (n = 24), HFrEF (n = 20), and age-matched controls (n = 38) were selected for metabolomics analysis as part of the Alberta HEART (Heart Failure Etiology and Analysis Research Team) project. Plasma BNP and NT-proBNP levels were measured using a Biosite Triage reagent pack and Elecsys 2010 proBNP assay, respectively. LVEF was assessed by echocardiography and interpreted by cardiologists blinded to the metabolomics analysis. Data are presented as the median ± IQR. * p < 0.05 compared to the control group, # p < 0.05 compared to the HFpEF group.
Fig 3. Heat map of metabolomic differences…
Fig 3. Heat map of metabolomic differences between HFpEF and controls.
Heat maps were generated with the concentrations of potential candidate metabolites with univariate analysis. Similar metabolites were arranged together for use in pathway analysis through intuitive pattern discovery. The heat map displays an increase in each metabolite in relative concentration as a red color and a decrease in a metabolite as a blue color. The metabolites are listed at the left side of each row, and the subjects are shown at the bottom of each column.
Fig 4
Fig 4
(A) Receiver operator characteristic (ROC) analysis for serum metabolites, cardiac peptides, and combined metabolites and NT-proBNP. Logistic regression (LR) was performed to find the most parsimonious model to discriminate HFpEF patients from control subjects using the minimum number of metabolites and/or cardiac peptides. Octanoylcarnitine, arginine, asparagine, lysophosphatidylcholine acyl C18:2, and sphingomyelin C20:2 were used in the metabolites-only panel. While octanoyl carnitine, arginine, and sphingomyelin C20:2 were used for the combined metabolites and NT-proBNP panel. (B–D) Quantification of the metabolites used to derive the LR equation of the combined metabolites and NT-proBNP model. Data are presented as means ± SD. * p < 0.05 compared to the control group.
Fig 5. Heat map of metabolomic differences…
Fig 5. Heat map of metabolomic differences between HFrEF and controls.
Heat maps were generated with the concentrations of potential candidate metabolites with univariate analysis. Similar metabolites were arranged together for use in pathway analysis through intuitive pattern discovery. The heat map displays an increase in each metabolite in relative concentration as a red color and a decrease in a metabolite as a blue color. The metabolites are listed at the left side of each row, and the subjects are shown at the bottom of each column.
Fig 6
Fig 6
(A) Receiver operator characteristic (ROC) analysis for serum metabolites, cardiac peptides, and combined serum metabolites and NT-proBNP. Logistic regression (LR) was performed to find the most parsimonious model to discriminate HFrEF patients from control subjects using the minimum number of metabolites and/or cardiac peptides. Creatinine, carnitine, acetoacetate, lysophosphatidylcholine acyl C18:2, 2-hydroxybutyrate, and lysophosphatidylcholine acyl C20:4 were used in the metabolites-only panel. While acetoacetate was used for the combined metabolites and NT-proBNP panel. (B) Quantification of acetoacetate which was used to derive the LR equation of the combined metabolites and NT-proBNP model. Data are presented as means ± SD. * p < 0.1 compared to the control group.
Fig 7. Heat map of metabolomic differences…
Fig 7. Heat map of metabolomic differences between HFpEF and HFrEF.
Heat maps were generated with the concentrations of potential candidate metabolites with univariate analysis. Similar metabolites were arranged together for use in pathway analysis through intuitive pattern discovery. The heat map displays an increase in each metabolite in relative concentration as a red color and a decrease in a metabolite as a blue color. The metabolites are listed at the left side of each row, and the subjects are shown at the bottom of each column.
Fig 8
Fig 8
(A) Receiver operator characteristic (ROC) analysis for serum metabolites, cardiac peptides, and combined serum metabolites and BNP. Logistic regression (LR) was performed to find the most parsimonious model to discriminate HFpEF from HFrEF patients using the minimum number of metabolites and/or cardiac peptides. 2-hydroxybutyrate, octadecenoylcarnitine (C18:1), hydroxyprionylcarnitine (C3-OH), and sphingomyelin C24:1 were used in the metabolites-only panel. While acetate, 2-hydroxybutyrate, pimelyl carnitine, and phosphatidyl choline diacyl C40:4 were used for the combined metabolites and NT-proBNP panel. (B–E) Quantification of the metabolites used to derive the LR equation of the combined metabolites and NT-proBNP model. Data are presented as means ± SD. The dashed line represents the metabolite concentration in control subjects. * p < 0.05 compared to the HFpEF group.

References

    1. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ, et al. Heart disease and stroke statistics—2014 update: a report from the american heart association. Circulation. 2014;129(3):e28–e292. 10.1161/01.cir.0000441139.02102.80
    1. Dunlay SM, Roger VL. Understanding the Epidemic of Heart Failure: Past, Present, and Future. Curr Heart Fail Rep. 2014.
    1. Abhayaratna WP, Marwick TH, Smith WT, Becker NG. Characteristics of left ventricular diastolic dysfunction in the community: an echocardiographic survey. Heart. 2006;92(9):1259–64.
    1. Owan TE, Hodge DO, Herges RM, Jacobsen SJ, Roger VL, Redfield MM. Trends in prevalence and outcome of heart failure with preserved ejection fraction. N Engl J Med. 2006;355(3):251–9.
    1. Paulus WJ, Tschope C, Sanderson JE, Rusconi C, Flachskampf FA, Rademakers FE, et al. How to diagnose diastolic heart failure: a consensus statement on the diagnosis of heart failure with normal left ventricular ejection fraction by the Heart Failure and Echocardiography Associations of the European Society of Cardiology. Eur Heart J. 2007;28(20):2539–50.
    1. Meta-analysis Global Group in Chronic Heart Failure (MAGGIC). The survival of patients with heart failure with preserved or reduced left ventricular ejection fraction: an individual patient data meta-analysis. Eur Heart J. 2012;33(14):1750–7. 10.1093/eurheartj/ehr254
    1. McMurray JJ, Adamopoulos S, Anker SD, Auricchio A, Bohm M, Dickstein K, et al. ESC guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC. Eur J Heart Fail. 2012;14(8):803–69. 10.1093/eurjhf/hfs105
    1. Borlaug BA, Paulus WJ. Heart failure with preserved ejection fraction: pathophysiology, diagnosis, and treatment. Eur Heart J. 2011;32(6):670–9. 10.1093/eurheartj/ehq426
    1. Du ZY, Shen A, Su L, Liang JQ, Xu DL. [Application of (1)H-NMR-based pattern recognition in serum metabolomics of patients with chronic heart failure]. Nan Fang Yi Ke Da Xue Xue Bao. 2012;32(3):415–9.
    1. Dunn WB, Broadhurst DI, Deepak SM, Buch MH, McDowell G, Spasic I, et al. Serum metabolomics reveals many novel metabolic markers of heart failure, including pseudouridine and 2-oxoglutarate. Metabolomics. 2007;3(4):413–26.
    1. Kang SM, Park JC, Shin MJ, Lee H, Oh J, Ryu do H, et al. (1)H nuclear magnetic resonance based metabolic urinary profiling of patients with ischemic heart failure. Clin Biochem. 2011;44(4):293–9. 10.1016/j.clinbiochem.2010.11.010
    1. Wang J, Li Z, Chen J, Zhao H, Luo L, Chen C, et al. Metabolomic identification of diagnostic plasma biomarkers in humans with chronic heart failure. Mol Biosyst. 2013;9(11):2618–26. 10.1039/c3mb70227h
    1. Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE Jr., Drazner MH, et al. 2013 ACCF/AHA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2013;62(16):e147–e239. 10.1016/j.jacc.2013.05.019
    1. Zile MR, Baicu CF. Biomarkers of diastolic dysfunction and myocardial fibrosis: application to heart failure with a preserved ejection fraction. J Cardiovasc Transl Res. 2013;6(4):501–15. 10.1007/s12265-013-9472-1
    1. van Veldhuisen DJ, Linssen GC, Jaarsma T, van Gilst WH, Hoes AW, Tijssen JG, et al. B-type natriuretic peptide and prognosis in heart failure patients with preserved and reduced ejection fraction. J Am Coll Cardiol. 2013;61(14):1498–506. 10.1016/j.jacc.2012.12.044
    1. Mamas M, Dunn WB, Neyses L, Goodacre R. The role of metabolites and metabolomics in clinically applicable biomarkers of disease. Arch Toxicol. 2011;85(1):5–17. 10.1007/s00204-010-0609-6
    1. Ezekowitz JA, Becher H, Belenkie I, Clark AM, Duff HJ, Friedrich MG, et al. The Alberta Heart Failure Etiology and Analysis Research Team (HEART) study. BMC Cardiovasc Disord. 2014;14(1):91.
    1. Bionda C, Bergerot C, Ardail D, Rodriguez-Lafrasse C, Rousson R. Plasma BNP and NT-proBNP assays by automated immunoanalyzers: analytical and clinical study. Ann Clin Lab Sci. 2006;36(3):299–306.
    1. Psychogios N, Hau DD, Peng J, Guo AC, Mandal R, Bouatra S, et al. The human serum metabolome. PLoS One. 2011;6(2):e16957 10.1371/journal.pone.0016957
    1. Ojala M, Garriga GC. Permutation Tests for Studying Classifier Performance. J Mach Learn Res. 2010;11:1833–63.
    1. Bijlsma S, Bobeldijk I, Verheij ER, Ramaker R, Kochhar S, Macdonald IA, et al. Large-scale human metabolomics studies: a strategy for data (pre-) processing and validation. Anal Chem. 2006;78(2):567–74.
    1. Pencina MJ, D'Agostino RB Sr., D'Agostino RB Jr., Vasan RS. Evaluating the added predictive ability of a new marker: from area under the ROC curve to reclassification and beyond. Stat Med. 2008;27(2):157–72; discussion 207–12.
    1. Marcovina SM, Sirtori C, Peracino A, Gheorghiade M, Borum P, Remuzzi G, et al. Translating the basic knowledge of mitochondrial functions to metabolic therapy: role of L-carnitine. Transl Res. 2013;161(2):73–84. 10.1016/j.trsl.2012.10.006
    1. Adams SH, Hoppel CL, Lok KH, Zhao L, Wong SW, Minkler PE, et al. Plasma acylcarnitine profiles suggest incomplete long-chain fatty acid beta-oxidation and altered tricarboxylic acid cycle activity in type 2 diabetic African-American women. J Nutr. 2009;139(6):1073–81. 10.3945/jn.108.103754
    1. Rutkowsky JM, Knotts TA, Ono-Moore KD, McCoin CS, Huang S, Schneider D, et al. Acylcarnitines activate proinflammatory signaling pathways. Am J Physiol Endocrinol Metab. 2014;306(12):E1378–87. 10.1152/ajpendo.00656.2013
    1. Ferro F, Ouille A, Tran TA, Fontanaud P, Bois P, Babuty D, et al. Long-chain acylcarnitines regulate the hERG channel. PLoS One. 2012;7(7):e41686 10.1371/journal.pone.0041686
    1. Kalim S, Clish CB, Wenger J, Elmariah S, Yeh RW, Deferio JJ, et al. A plasma long-chain acylcarnitine predicts cardiovascular mortality in incident dialysis patients. J Am Heart Assoc. 2013;2(6):e000542 10.1161/JAHA.113.000542
    1. Putko BN, Wang Z, Lo J, Anderson T, Becher H, Dyck JR, et al. Circulating levels of tumor necrosis factor-alpha receptor 2 are increased in heart failure with preserved ejection fraction relative to heart failure with reduced ejection fraction: evidence for a divergence in pathophysiology. PLoS One. 2014;9(6):e99495 10.1371/journal.pone.0099495
    1. Zakeri R, Chamberlain AM, Roger VL, Redfield MM. Temporal relationship and prognostic significance of atrial fibrillation in heart failure patients with preserved ejection fraction: a community-based study. Circulation. 2013;128(10):1085–93. 10.1161/CIRCULATIONAHA.113.001475
    1. Martin MA, Gomez MA, Guillen F, Bornstein B, Campos Y, Rubio JC, et al. Myocardial carnitine and carnitine palmitoyltransferase deficiencies in patients with severe heart failure. Biochim Biophys Acta. 2000;1502(3):330–6.
    1. El-Aroussy W, Rizk A, Mayhoub G, Aleem SA, El-Tobgy S, Mokhtar MS. Plasma carnitine levels as a marker of impaired left ventricular functions. Mol Cell Biochem. 2000;213(1–2):37–41.
    1. Zamora E, Lupon J, de Antonio M, Vila J, Penafiel J, Galan A, et al. Long-Term Prognostic Value for Patients with Chronic Heart Failure of Estimated Glomerular Filtration Rate Calculated with the New CKD-EPI Equations Containing Cystatin C. Clin Chem. 2013.
    1. Pasini E, Aquilani R, Dioguardi FS, D'Antona G, Gheorghiade M, Taegtmeyer H. Hypercatabolic syndrome: molecular basis and effects of nutritional supplements with amino acids. Am J Cardiol. 2008;101(11A):11E–5E. 10.1016/j.amjcard.2008.02.074
    1. Strilakou AA, Lazaris AC, Perelas AI, Mourouzis IS, Douzis I, Karkalousos PL, et al. Heart dysfunction induced by choline-deficiency in adult rats: the protective role of L-carnitine. Eur J Pharmacol. 2013;709(1–3):20–7. 10.1016/j.ejphar.2013.03.040
    1. Okumura K, Yamada Y, Kondo J, Hashimoto H, Ito T, Kitoh J. Decreased 1,2-diacylglycerol levels in myopathic hamster hearts during the development of heart failure. J Mol Cell Cardiol. 1991;23(4):409–16.
    1. Haris M, Singh A, Cai K, Kogan F, McGarvey J, Debrosse C, et al. A technique for in vivo mapping of myocardial creatine kinase metabolism. Nat Med. 2014.
    1. Tenori L, Hu X, Pantaleo P, Alterini B, Castelli G, Olivotto I, et al. Metabolomic fingerprint of heart failure in humans: a nuclear magnetic resonance spectroscopy analysis. Int J Cardiol. 2013;168(4):e113–5. 10.1016/j.ijcard.2013.08.042
    1. Lommi J, Kupari M, Koskinen P, Naveri H, Leinonen H, Pulkki K, et al. Blood ketone bodies in congestive heart failure. J Am Coll Cardiol. 1996;28(3):665–72.
    1. Lommi J, Koskinen P, Naveri H, Harkonen M, Kupari M. Heart failure ketosis. J Intern Med. 1997;242(3):231–8.
    1. Melenovsky V, Kotrc M, Polak J, Pelikanova T, Bendlova B, Cahova M, et al. Availability of energetic substrates and exercise performance in heart failure with or without diabetes. Eur J Heart Fail. 2012;14(7):754–63. 10.1093/eurjhf/hfs080
    1. Rhee EP, Gerszten RE. Metabolomics and cardiovascular biomarker discovery. Clin Chem. 2012;58(1):139–47. 10.1373/clinchem.2011.169573
    1. Thomas MD, Fox KF, Wood DA, Gibbs JS, Coats AJ, Henein MY, et al. Echocardiographic features and brain natriuretic peptides in patients presenting with heart failure and preserved systolic function. Heart. 2006;92(5):603–8.
    1. Yamaguchi H, Yoshida J, Yamamoto K, Sakata Y, Mano T, Akehi N, et al. Elevation of plasma brain natriuretic peptide is a hallmark of diastolic heart failure independent of ventricular hypertrophy. J Am Coll Cardiol. 2004;43(1):55–60.
    1. Martos R, Baugh J, Ledwidge M, O'Loughlin C, Murphy NF, Conlon C, et al. Diagnosis of heart failure with preserved ejection fraction: improved accuracy with the use of markers of collagen turnover. Eur J Heart Fail. 2009;11(2):191–7. 10.1093/eurjhf/hfn036
    1. Wang YC, Yu CC, Chiu FC, Tsai CT, Lai LP, Hwang JJ, et al. Soluble ST2 as a biomarker for detecting stable heart failure with a normal ejection fraction in hypertensive patients. J Card Fail. 2013;19(3):163–8. 10.1016/j.cardfail.2013.01.010
    1. Tschope C, Kasner M, Westermann D, Gaub R, Poller WC, Schultheiss HP. The role of NT-proBNP in the diagnostics of isolated diastolic dysfunction: correlation with echocardiographic and invasive measurements. Eur Heart J. 2005;26(21):2277–84.
    1. Jorge AJ, Freire MD, Ribeiro ML, Fernandes LC, Lanzieri PG, Jorge BA, et al. [Utility of B-type natriuretic peptide measurement in outpatients with heart failure with preserved ejection fraction]. Rev Port Cardiol. 2013;32(9):647–52. 10.1016/j.repc.2012.10.019
    1. Anjan VY, Loftus TM, Burke MA, Akhter N, Fonarow GC, Gheorghiade M, et al. Prevalence, clinical phenotype, and outcomes associated with normal B-type natriuretic peptide levels in heart failure with preserved ejection fraction. Am J Cardiol. 2012;110(6):870–6. 10.1016/j.amjcard.2012.05.014
    1. Lukacs I, Fey E, Karpati P. [Experiences with serum BNP (B type natriuretic peptide) in patients with systolic and diastolic heart failure]. Orv Hetil. 2005;146(37):1919–23.
    1. Mottram PM, Leano R, Marwick TH. Usefulness of B-type natriuretic peptide in hypertensive patients with exertional dyspnea and normal left ventricular ejection fraction and correlation with new echocardiographic indexes of systolic and diastolic function. Am J Cardiol. 2003;92(12):1434–8.
    1. Fonseca C, Sarmento PM, Minez A, Goncalves E, Covas R, Dias AR, et al. Comparative value of BNP and NT-proBNP in diagnosis of heart failure. Rev Port Cardiol. 2004;23(7–8):979–91.
    1. Oktay AA, Rich JD, Shah SJ. The emerging epidemic of heart failure with preserved ejection fraction. Curr Heart Fail Rep. 2013;10(4):401–10.

Source: PubMed

Подписаться