Aptamer-Based Proteomic Platform Identifies Novel Protein Predictors of Incident Heart Failure and Echocardiographic Traits

Supplemental Digital Content is available in the text. Background: We used a large-scale, high-throughput DNA aptamer-based discovery proteomic platform to identify circulating biomarkers of cardiac remodeling and incident heart failure (HF) in community-dwelling individuals. Methods: We evaluated 1895 FHS (Framingham Heart Study) participants (age 55±10 years, 54% women) who underwent proteomic profiling and echocardiography. Plasma levels of 1305 proteins were related to echocardiographic traits and to incident HF using multivariable regression. Statistically significant protein-HF associations were replicated in the HUNT (Nord-Trøndelag Health) study (n=2497, age 63±10 years, 43% women), and results were meta-analyzed. Genetic variants associated with circulating protein levels (pQTLs) were related to echocardiographic traits in the EchoGen (n=30 201) and to incident HF in the CHARGE (n=20 926) consortia. Results: Seventeen proteins associated with echocardiographic traits in cross-sectional analyses (false discovery rate <0.10), and 8 of these proteins had pQTLs associated with echocardiographic traits in EchoGen (P<0.0007). In Cox models adjusted for clinical risk factors, 29 proteins demonstrated associations with incident HF in FHS (174 HF events, mean follow-up 19 [limits, 0.2–23.7] years). In meta-analyses of FHS and HUNT, 6 of these proteins were associated with incident HF (P<3.8×10−5; 3 with higher risk: NT-proBNP [N-terminal proB-type natriuretic peptide], TSP2 [thrombospondin-2], MBL [mannose-binding lectin]; and 3 with lower risk: ErbB1 [epidermal growth factor receptor], GDF-11/8 [growth differentiation factor-11/8], and RGMC [hemojuvelin]). For 5 of the 6 proteins, pQTLs were associated with echocardiographic traits (P<0.0006) in EchoGen, and for RGMC, a protein quantitative trait loci was associated with incident HF (P=0.001). Conclusions: A large-scale proteomics approach identified new predictors of cardiac remodeling and incident HF. Future studies are warranted to elucidate how biological pathways represented by these proteins may mediate cardiac remodeling and HF risk and to assess if these proteins can improve HF risk prediction.

[1]  M. Soley,et al.  Activated epidermal growth factor receptor (ErbB1) protects the heart against stress-induced injury in mice. , 2003, American journal of physiology. Regulatory, integrative and comparative physiology.

[2]  S. Franklin,et al.  Metabolic Remodeling in Moderate Synchronous versus Dyssynchronous Pacing-Induced Heart Failure: Integrated Metabolomics and Proteomics Study , 2015, PloS one.

[3]  W. Kannel,et al.  An investigation of coronary heart disease in families. The Framingham offspring study. , 1979, American journal of epidemiology.

[4]  Herbert Y. Lin,et al.  The RGM/DRAGON family of BMP co-receptors. , 2009, Cytokine & growth factor reviews.

[5]  Stephen Burgess,et al.  Genomic atlas of the human plasma proteome , 2018, Nature.

[6]  R. Vasan,et al.  Genetic Architecture of the Cardiovascular Risk Proteome , 2017, Circulation.

[7]  Eric Boerwinkle,et al.  Association of Genome-Wide Variation With the Risk of Incident Heart Failure in Adults of European and African Ancestry: A Prospective Meta-Analysis From the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium , 2010, Circulation. Cardiovascular genetics.

[8]  D. Levy,et al.  Cardiac Dysfunction and Noncardiac Dysfunction as Precursors of Heart Failure With Reduced and Preserved Ejection Fraction in the Community , 2011, Circulation.

[9]  D. Eisen,et al.  Impact of mannose-binding lectin on susceptibility to infectious diseases. , 2003, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[10]  Thomas Meitinger,et al.  Genetic variants associated with cardiac structure and function: a meta-analysis and replication of genome-wide association data. , 2009, JAMA.

[11]  G. Felker,et al.  Biomarkers for the Prevention of Heart Failure: Are We There Yet? , 2018, Journal of the American College of Cardiology.

[12]  B. Howard,et al.  Prospective Analysis of Mannose-Binding Lectin Genotypes and Coronary Artery Disease in American Indians: The Strong Heart Study , 2004, Circulation.

[13]  Plasma natriuretic peptides for community screening for left ventricular hypertrophy and systolic dysfunction: the Framingham heart study. , 2002 .

[14]  M. Pfisterer,et al.  Influence of functional deficiency of complement mannose-binding lectin on outcome of patients with acute ST-elevation myocardial infarction undergoing primary percutaneous coronary intervention. , 2010, European heart journal.

[15]  Daniel Levy,et al.  Plasma natriuretic peptide levels and the risk of cardiovascular events and death. , 2004, The New England journal of medicine.

[16]  Thomas J. Wang,et al.  Accelerating Biomarker Discovery Through Electronic Health Records, Automated Biobanking, and Proteomics. , 2019, Journal of the American College of Cardiology.

[17]  P. Amouyel,et al.  Multimarker Proteomic Profiling for the Prediction of Cardiovascular Mortality in Patients with Chronic Heart Failure , 2015, PloS one.

[18]  F. David,et al.  Statistical Estimates and Transformed Beta-Variables. , 1960 .

[19]  M. Schellings,et al.  Thrombospondins in the heart: potential functions in cardiac remodeling , 2009, Journal of Cell Communication and Signaling.

[20]  V. Roger Epidemiology of Heart Failure , 2013, Circulation research.

[21]  Stephen A. Williams,et al.  Improving Assessment of Drug Safety Through Proteomics , 2017, Circulation.

[22]  M. Seidel,et al.  Mannan-binding lectin deficiency - Good news, bad news, doesn't matter? , 2012, Clinical immunology.

[23]  R. Gerszten,et al.  Application of Large-Scale Aptamer-Based Proteomic Profiling to Planned Myocardial Infarctions , 2017, Circulation.

[24]  C. Shovlin,et al.  Antiplatelet and anticoagulant agents in hereditary hemorrhagic telangiectasia. , 2013, The New England journal of medicine.

[25]  P. Ponikowski,et al.  Novel endotypes in heart failure: effects on guideline-directed medical therapy , 2018, European heart journal.

[26]  C. Yancy,et al.  Role of Biomarkers for the Prevention, Assessment, and Management of Heart Failure: A Scientific Statement From the American Heart Association , 2017, Circulation.

[27]  R. Duisters,et al.  Thrombospondin-2 Is Essential for Myocardial Matrix Integrity: Increased Expression Identifies Failure-Prone Cardiac Hypertrophy , 2004, Circulation research.

[28]  B. Kühn,et al.  ERBB2 inhibition and heart failure. , 2013, The New England journal of medicine.

[29]  Sanjiv J. Shah,et al.  Association of Cardiovascular Biomarkers With Incident Heart Failure With Preserved and Reduced Ejection Fraction , 2018, JAMA cardiology.

[30]  Manuel Mayr,et al.  In Aptamers They Trust: Caveats of the SOMAscan Biomarker Discovery Platform From SomaLogic , 2018, Circulation.

[31]  Terrie E. Kitchner,et al.  Probing the Virtual Proteome to Identify Novel Disease Biomarkers , 2018, Circulation.

[32]  J. D. de Lemos,et al.  Impact of Smoking on Circulating Cardiac Troponin I Concentrations and Cardiovascular Events in the General Population: The HUNT Study (Nord-Trøndelag Health Study) , 2016, Circulation.

[33]  Mark R Segal,et al.  Development and Validation of a Protein-Based Risk Score for Cardiovascular Outcomes Among Patients With Stable Coronary Heart Disease. , 2016, JAMA.

[34]  Jie Huang,et al.  Large-scale genome-wide analysis identifies genetic variants associated with cardiac structure and function , 2017, The Journal of clinical investigation.

[35]  D. Levy,et al.  Relations of Biomarkers Representing Distinct Biological Pathways to Left Ventricular Geometry , 2008, Circulation.

[36]  A. Orekhov,et al.  Thrombospondins: A Role in Cardiovascular Disease , 2017, International Journal of Molecular Sciences.

[37]  P. Hildebrandt,et al.  Detection of left ventricular enlargement and impaired systolic function with plasma N-terminal pro brain natriuretic peptide concentrations. , 2002, American heart journal.

[38]  J. Januzzi,et al.  Established and Emerging Roles of Biomarkers in Heart Failure , 2018, Circulation research.

[39]  Robert E. Gerszten,et al.  Emerging Affinity-Based Proteomic Technologies for Large-Scale Plasma Profiling in Cardiovascular Disease , 2017, Circulation.

[40]  Brian R. Kotajarvi,et al.  Quantification of GDF11 and Myostatin in Human Aging and Cardiovascular Disease. , 2016, Cell metabolism.

[41]  Xu Shi,et al.  Aptamer-Based Proteomic Profiling Reveals Novel Candidate Biomarkers and Pathways in Cardiovascular Disease , 2016, Circulation.

[42]  Xia Yang,et al.  Co-regulatory networks of human serum proteins link genetics to disease , 2018, Science.

[43]  Richard T. Lee,et al.  Growth Differentiation Factor 11 Is a Circulating Factor that Reverses Age-Related Cardiac Hypertrophy , 2013, Cell.

[44]  Shane T. Jensen,et al.  Bayesian integration of genetics and epigenetics detects causal regulatory SNPs underlying expression variability , 2015, Nature Communications.

[45]  Y. Benjamini,et al.  Controlling the false discovery rate: a practical and powerful approach to multiple testing , 1995 .